WO1997035990A2 - Histone deacetylases, and uses related thereto - Google Patents

Histone deacetylases, and uses related thereto Download PDF

Info

Publication number
WO1997035990A2
WO1997035990A2 PCT/US1997/005275 US9705275W WO9735990A2 WO 1997035990 A2 WO1997035990 A2 WO 1997035990A2 US 9705275 W US9705275 W US 9705275W WO 9735990 A2 WO9735990 A2 WO 9735990A2
Authority
WO
WIPO (PCT)
Prior art keywords
hdx
polypeptide
protein
cell
nucleic acid
Prior art date
Application number
PCT/US1997/005275
Other languages
French (fr)
Other versions
WO1997035990A9 (en
WO1997035990A3 (en
Inventor
Stuart L. Schreiber
Jack Taunton
Christian A. Hassig
Timothy F. Jamison
Original Assignee
President And Fellows Of Harvard College
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by President And Fellows Of Harvard College filed Critical President And Fellows Of Harvard College
Priority to AU29905/97A priority Critical patent/AU2990597A/en
Publication of WO1997035990A2 publication Critical patent/WO1997035990A2/en
Publication of WO1997035990A3 publication Critical patent/WO1997035990A3/en
Publication of WO1997035990A9 publication Critical patent/WO1997035990A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/78Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5)
    • C12N9/80Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5) acting on amide bonds in linear amides (3.5.1)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/12Cyclic peptides with only normal peptide bonds in the ring
    • C07K5/126Tetrapeptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the nucleosome consists of 146 bp of DNA wrapped around a protein core of the histones H2A, H2B, H3, and H4, known as the core histones. These histones are arranged as an (H3-H4) 2 tetramer and two H2A-H2B dimers positioned on each face of the tetramer.
  • the DNA joining the nucleosomes is called linker DNA; it is to the linker DNA to which the HI or linker histones bind.
  • the 10 nm fibre is compacted further into the 30 nm fibre.
  • Linker histones and amino-terminal regions ("tails") of the core histones maintain the higher order folding of chromatin (Garcia Ramirez et al.. (1992) J. Biol Chem 267:19587). This chromatin structure must be relaxed when DNA is transcribed or translated.
  • Histones of the nucleosome core particle are subject to reversible acetylation at the ⁇ -amino group of lysines present in their amino terminus (Csordas et al. (1990) Biochem J 265:23-38). Transcriptionally silent regions of the genome are enriched in underacetyiated histone H4 (Turner (1993) Cell 75:5-8), and histone hyperacetylation facilitates the ability of transcription factor TFHIA to bind to chromatin templates (Lee et al. (1993) Cell 72:73-84). Recent genetic, biochemical and immunological approaches have provided substantial evidence indicating that histones associated with actively transcribed genes are more highly acetylated than those from nontranscribed regions.
  • histone acetylation may influence transcription at several stages, for example, by causing transcription factors to bind or by inducing structural transitions in chromatin, or by facilitating histone displacement and repositioning during polymerase elongation.
  • acetylation and deacetylation are catalyzed by specific enzymes, histone acetyltransferase and deacetylase, respectively, and the net level of the acetylation is controlled by the equilibrium between these enzymes.
  • the steady state level of acetylation and the rates at which acetate groups are turned over vary both between and within different cell types, with half-lives that vary from a few minutes to several hours.
  • HAT1 histone acetyltransferase gene
  • the present invention relates to the discovery of a novel family of genes, and gene products, expressed in mammals, which genes are referred to hereinafter as the "histone deacetylase” genes or “HDx” gene family, the products of which are referred to as histone deacetylases or HDx proteins.
  • the invention features isolated HDx polypeptides, preferably substantially pure preparations of one or more of the subject HDx polypeptides.
  • the invention also provides recombinantly produced HDx polypeptides.
  • the polypeptide has a biological activity including an ability to deacetylate an acetylated histone substrate, preferably a substrate analog of histone H3 and/or H4.
  • the HDx polypeptides of the present invention bind to trapoxin or to trichostatin, such binding resulting in the inhibition a deacetylase activity of the HDx polypeptide.
  • HDx polypeptides which specifically antagonize such activities such as may be provided by dominant negative mutants, are also specifically contemplated.
  • the HDx polypeptides disclosed herein are capable of modulating proliferation, survival and/or differentiation of cells, because of their ability to alter chromatin structure by deacetylating histones such as H3 or H4. Moreover, in preferred embodiments, the subject HDx proteins have the ability to modulate cell growth by influencing cell cycle progression or to modulate gene transcription.
  • the polypeptide is identical with or homologous to an HDx protein.
  • Exemplary HDx polypeptide include amino acid sequences represented in any one of SEQ ID Nos 5-8.
  • an HDx polypeptide preferably has an amino acid sequence at least 85% homologous to a polypeptide represented by one or more of the polypeptides designated SEQ ID Nos: 5-8, though polypeptides with higher sequence homologies of, for example, 88, 90% and 95% or are also contemplated.
  • the HDx polypeptide is encoded by a nucleic acid which hybridizes under stringent conditions with a nucleic acid sequence represented in one or more of SEQ ID Nos. 1-4.
  • Homologs of the subject HDx proteins also include versions of the protein which are resistant to post-translation modification, as for example, due to mutations which alter modification sites (such as tyrosine, threonine, serine or aspargine residues), or which inactivate an enzymatic activity associated with the protein.
  • the HDx polypeptide can comprise a full length protein, such as represented in SEQ ID No. 5, or it can comprise a fragment corresponding to particular motifs/domains, or to arbitrary sizes, e.g., at least 5, 10, 25, 50, 100, 150 or 200 amino acids in length.
  • the polypeptide, or fragment thereof specifically deacetylates histone H4.
  • the HDx polypeptide includes both a v motif (SEQ ID No. 12) and a ⁇ motif (SEQ ID No. 14), preferably a v motif represented in the general formula SEQ ID No. 13, and a ⁇ motif represented in the general formula SEQ ID No. 15.
  • the invention features a purified or recombinant HDx polypeptide having a molecular weight in the range of 40kd to 60kd.
  • preferred HDx polypeptides have molecular weights in the range of 50kd to about 60kd, even more preferably in the range of 53-58kd.
  • certain post ⁇ translational modifications e.g., phosphorylation, prenylation and the like, can increase the apparent molecular weight of the HDx protein relative to the unmodified polypeptide chain.
  • die HDx protein can be provided as a recombinant fusion protein which includes a second polypeptide portion, e.g., a second polypeptide having an amino acid sequence unrelated (heterologous) to the HDx polypeptide, e.g. the second polypeptide portion is glutathione-S-transferase, e.g. the second polypeptide portion is an enzymatic activity such as alkaline phosphatase, e.g. the second polypeptide portion is an epitope tag.
  • a second polypeptide portion e.g., a second polypeptide having an amino acid sequence unrelated (heterologous) to the HDx polypeptide, e.g. the second polypeptide portion is glutathione-S-transferase, e.g. the second polypeptide portion is an enzymatic activity such as alkaline phosphatase, e.g. the second polypeptide portion is an epitope tag.
  • the invention features a nucleic acid encoding a an HDx polypeptide, or polypeptide homologous thereto, which polypeptide has the ability to modulate, e.g., either mimic or antagonize, at least a portion of the activity of a wild ⁇ type HDx polypeptide.
  • Exemplary HDx-encoding nucleic acid sequences are represented by SEQ ID Nos: 1-4.
  • the nucleic acid of the present invention includes a coding sequence which hybridizes under stringent conditions with one or more of the nucleic acid sequences in SEQ ID Nos: 1-4.
  • the coding sequence of the nucleic acid can comprise a sequence which is identical to a coding sequence represented in one of SEQ ID Nos: 1-4, or it can merely be homologous to one or more of those sequences.
  • the nucleic acid encodes a polypeptide which specifically modulates, by acting as either an agonist or antagonist, the enzymatic activity of an HDx polypeptide.
  • the subject HDx nucleic acid will include a transcriptional regulatory sequence, e.g. at least one of a transcriptional promoter or transcriptional enhancer sequence, which regulatory sequence is operably linked to the HDx gene sequence.
  • a transcriptional regulatory sequence e.g. at least one of a transcriptional promoter or transcriptional enhancer sequence, which regulatory sequence is operably linked to the HDx gene sequence.
  • Such regulatory sequences can be used in to render the HDx gene sequence suitable for use as an expression vector.
  • This invention also contemplates the cells transfected with said expression vector whether prokaryotic or eukaryotic and a method for producing HDx proteins by employing said expression vectors.
  • the nucleic acid hybridizes under stringent conditions to a nucleic acid probe corresponding to at least 12 consecutive nucleotides of either sense or antisense sequence of one or more of SEQ ID Nos: 1-4; though preferably to at least 25 consecutive nucleotides; and more preferably to at least 40, 50 or 75 consecutive nucleotides of either sense or antisense sequence of one or more of SEQ ID Nos: 1-4.
  • an immunogen comprising an HDx polypeptide in an immunogenic preparation, the immunogen being capable of eliciting an immune response specific for an HDx polypeptide; e.g. a humoral response, e.g. an antibody response; e.g. a cellular response.
  • the immunogen comprising an antigenic determinant, e.g. a unique determinant, from a protein represented by one of SEQ ID Nos. 5-8.
  • a still further aspect of the present invention features antibodies and antibody preparations specifically reactive with an epitope of the HDx immunogen.
  • the invention also features transgenic non-human animals, e.g. mice, rats, rabbits, chickens, frogs or pigs, having a transgene, e.g., animals which include (and preferably express) a heterologous form of an HDx gene described herein, or which misexpress an endogenous HDx gene, e.g., an animal in which expression of one or more of the subject HDx proteins is disrupted.
  • a transgenic animal can serve as an animal model for studying cellular and tissue disorders comprising mutated or mis-expressed HDx alleles or for use in drug screening.
  • the invention also provides a probe/primer comprising a substantially purified oligonucleotide, wherein the oligonucleotide comprises a region of nucleotide sequence which hybridizes under stringent conditions to at least 12 consecutive nucleotides of sense or antisense sequence of SEQ DD Nos: 1-4, or naturally occurring mutants thereof.
  • Nucleic acid probes which are specific for each of the HDx proteins are contemplated by the present invention, e.g. probes which can discern between nucleic acid encoding a human or bovine HD.
  • the probe/primer further includes a label group attached thereto and able to be detected.
  • the label group can be selected, e.g., from a group consisting of radioisotopes, fluorescent compounds, enzymes, and enzyme co-factors.
  • Probes of the invention can be used as a part of a diagnostic test kit for identifying dysfunctions associated with mis-expression of an HDx protein, such as for detecting in a sample of cells isolated from a patient, a level of a nucleic acid encoding a subject HDx protein; e.g. measuring an HDx mRNA level in a cell, or determining whether a genomic HDx gene has been mutated or deleted.
  • probes/primers of the invention can also be used as a part of "antisense” therapy which refers to administration or in situ generation of oligonucleotide probes or their derivatives which specifically hybridize (e.g. bind) under cellular conditions, with the cellular mRNA and or genomic DNA encoding one or more of the subject HDx proteins so as to inhibit expression of that protein, e.g. by inhibiting transcription and/or translation.
  • die oligonucleotide is at least 12 nucleotides in length, though primers of 25, 40, 50, or 75 nucleotides in length are also contemplated.
  • the invention provides an assay for screening test compounds for inhibitors, or alternatively, potentiators, of an interaction between an HDx protein and an HDx binding protein or nucleic acid sequence.
  • An exemplary method includes the steps of (i) combining an HDx polypeptide or fragment thereof, one or more HDx target polypeptide (such as a histone, SIN3, RpAp48 or other protein which participates in HDx complexes, e.g., one or more proteins having molecular weights of 250 kDa, 180 kDa, 55 kDa, 50 kDa, 42 kDa, 33-36 kDa and 30 kDa, see also Example 3), and a test compound, e.g., under conditions wherein, but for the test compound, the HDx protein and target polypeptide(s) are able to interact; and (ii) detecting the formation of a complex which includes the HDx protein and target polypeptide(s) either by directly quantitating the complex, the de
  • a statistically significant change, such as a decrease, in the formation of the complex in the presence of a test compound (relative to what is seen in the absence of the test compound) is indicative of a modulation, e.g., inhibition, of the interaction between the HDx protein and its target polypeptide.
  • a modulation e.g., inhibition
  • the drug screening assay can be derived with a fungal homolog of an HDx protein, such as RPD3, in order to identify agents which inhibit histone deacetylation in a yeast cell.
  • a fungal homolog of an HDx protein such as RPD3
  • Yet another aspect of the present invention concerns a method for modulating one or more of growth, differentiation, or survival of a mammalian cell by modulating HDx bioactivity, e.g., by inhibiting the deacetylase activity of HDx proteins, or disrupting certain protein-protein interactions.
  • the method comprises treating the cell with an effective amount of an HDx therapeutic so as to alter, relative to the cell in the absence of treatment, at least one of (i) rate of growth, (ii) differentiation, or (iii) survival of the cell.
  • the method can be carried out with HDx therapeutics such as peptide and peptidomimetics or other molecules identified in the above-referenced drug screens which antagonize the effects of a naturally-occurring HDx protein on said cell.
  • Other HDx therapeutics include antisense constructs for inhibiting expression of HDx proteins, and dominant negative mutants of HDx proteins which competitively inhibit protein-substrate and/or protein- protein interactions upstream and downstream of the wild-type HDx protein.
  • the subject method is used to treat tumor cells by antagonizing HDx activity and blocking cell cycle progression.
  • the subject method includes the treatment of testicular cells, so as modulate spermatogenesis.
  • the subject method is used to modulate osteogenesis, comprising the treatment of osteogenic cells with an HDx polypeptide.
  • the treated cell is a chondrogenic cell
  • the present method is used to modulate chondrogenesis.
  • HDx polypeptides can be used to modulate the differentiation of progenitor cells, e.g., the method can be used to cause differentiation of a hematopoietic cells, neuronal cells, or other stem/progenitor cell populations, to maintain a that cell in a differentiated state, and/or to enhance the survival of a differentiated cell, e.g., to prevent apoptosis or other forms of cell death.
  • anti-fungal agents developed with such screening assays as described herein can be used, for example, as preservatives in foodstuff, feed supplement for promoting weight gain in livestock, or in disinfectant formulations for treatment of non-living matter, e.g., for decontaminating hospital equipment and rooms.
  • assays provided herein will permit selection of deacetylase inhibitors which discriminate between the human and insect deacetylase enzymes.
  • d e present invention expressly contemplates the use and formulations of the deacetylase inhibitors in insecticides, such as for use in management of insects like the fruit fly.
  • certain of the inhibitors can be selected on the basis of inhibitory specificity for plant H ⁇ x-related activities relative to the mammalian enzymes.
  • the present invention specifically contemplates formulations of deacetylase inhibitors for agricultural applications, such as in the form of a defoliant or the like.
  • the present method is applicable, for example, to cell culture technique, such as in the culturing of hematopoietic cells and other cells whose survival or differentiative state is dependent on HDx function.
  • HDx agonists and antagonists can be used for therapeutic intervention, such as to enhance survival and maintenance of cells, as well as to influence organogenic pathways, such as tissue patterning and other differentiation processes.
  • the method is practiced for modulating, in an animal, cell growth, cell differentiation or cell survival, and comprises administering a therapeutically effective amount of an HDx polypeptide to alter, relative the absence of HDx treatment, at least one of (i) rate of growth, (ii) differentiation, or (iii) survival of one or more cell-types in the animal.
  • Another aspect of the present invention provides a method of determining if a subject, e.g. a human patient, is at risk for a disorder characterized by unwanted cell proliferation or aberrant control of differentiation.
  • the method includes detecting, in a tissue of the subject, the presence or absence of a genetic lesion characterized by at least one of (i) a mutation of a gene encoding an HDx protein, e.g. represented in one of SEQ ID Nos: 1-4, or a homolog thereof; or (ii) the mis-expression of an HDx gene.
  • detecting the genetic lesion includes ascertaining the existence of at least one of: a deletion of one or more nucleotides from an HDx gene; an addition of one or more nucleotides to the gene, a substitution of one or more nucleotides of the gene, a gross chromosomal rearrangement of the gene; an alteration in the level of a messenger RNA transcript of the gene; the presence of a non- wild type splicing pattern of a messenger RNA transcript of the gene; or a non-wild type level of the protein.
  • detecting the genetic lesion can include (i) providing a probe/primer including an oligonucleotide containing a region of nucleotide sequence which hybridizes to a sense or antisense sequence of an HDx gene, e.g. a nucleic acid represented in one of SEQ ID Nos: 1-4, or naturally occurring mutants thereof, or 5' or 3' flanking sequences naturally associated with the HDx gene; (ii) exposing the probe primer to nucleic acid of the tissue; and (iii) detecting, by hybridization of the probe/primer to the nucleic acid, the presence or absence of the genetic lesion; e.g.
  • detecting the lesion comprises utilizing the probe/primer to determine the nucleotide sequence of the HDx gene and, optionally, of the flanking nucleic acid sequences.
  • the probe/primer can be employed in a polymerase chain reaction (PCR) or in a ligation chain reaction (LCR).
  • PCR polymerase chain reaction
  • LCR ligation chain reaction
  • d e level of an HDx protein is detected in an immunoassay using an antibody which is specifically immunoreactive with the HDx protein.
  • an HDx inhibitor compound of the invention can be represented by the formula A-B-C, in which A is a specificity element for selective binding to an HDx, B is a linker element, and C is an electrophilic moiety capable of reacting with a nucleophilic moiety of an HDx; with the proviso that the compound is not butyrate, trapoxin, or trichostatin.
  • composition for inhibiting a histone deacetylase comprising a compound represented by the general formula A-B-C, wherein A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclopeptides;
  • B is selected from d e group consisting of substituted and unsubstituted C 4 -C 8 alkylidenes, C -Cg alkenylidenes, C -C 8 alkynylidenes, and -(D-E-F)-, in which D and F are, independently, absent or represent a C2-C 7 alkylidene, a C 2 -C 7 alkenylidene or a C2- C7 alkynylidene, and E represents O, S, or NR', in which R' represents H, a lower alkyl, a lower alkenyl, a lower alkynyl, an aralkyl, aryl, or a heterocyclyl; and
  • C is selected from the group consisting of , and a boronic acid; in which Z represents O, S, or NR 5 , and Y; R5 represents a hydrogen, an alkyl, an alkoxycarbonyl, an aryloxycarbonyl, an alkylsulfonyl, an arylsulfonyl or an aryl; R' 6 represents hydrogen, an alkyl, an alkenyl, an alkynyl or an aryl; and R represents a hydrogen, an alkyl, an aryl, an alkoxy, an aryloxy, an amino, a hydroxylamino, an alkoxylamino or a halogen; with the proviso that the compound is not trapoxin.
  • A is selected from the group consisting of cycloalk ls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclopeptides;
  • B is selected from the group consisting of substituted and unsubstituted C -C 8 alkylidenes, C -C 8 alkenylidenes, C 4 -C 8 alkynylidenes, and -(D-E-F)-, in which D and F are, independently, absent or represent C 2 -C alkylidenes, C 2 -C 7 alkenylidenes or C 2 -C 7 alkynylidenes, and E represents O, S, or NR', in which R' represents H, a lower alkyl, a lower alkenyl, a lower alkynyl, an aralkyl, an aryl, or a heterocyclyl; and
  • C is selected from the group consisting of
  • R 9 represents a hydrogen, an alkyl, an aryl, a hydroxyl, an alkoxy, an aryloxy or an amino, with the proviso that the inhibitor compound is not trichostatin.
  • the compound is represented by the general formula A-B-C, wherein
  • A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclopeptides;
  • B is selected from the group consisting of substituted and unsubstituted C -C 8 alkylidenes, C -C 8 alkenylidenes, C -C alkynylidenes, and -(D-E-F)-, in which D and F are, independently, absent or a C 2 -C 7 alkylidene, a C 2 -C alkenylidene, or a C 2 -C 7 alkynylidene, and E represents O, S, or NR', in which R' is H, lower alkyl, lower alkenyl, lower alkynyl, aralkyl, aryl, or heterocyclyl; and
  • C represents ; in which Y is O or S, and R7 represents a hydrogen, an alkyl, an aryl, an alkoxy, an aryloxy, an amino, a hydroxylamino, an alkoxylamino or a halogen.
  • the present invention also contemplates pharmaceutical preparations of such compounds, e.g., in an amount effective for inhibiting proliferation of a cell, formulated in a pharmaceutically acceptable diluent.
  • such compounds can be used for modulating one or more of growth, differentiation, or survival of a mammalian cell responsive to HDx-mediated histone deacetylation, by treating the cell with an effective amount of the deacetylase inhibitor so as to modulate the deacetylase activity and alter, relative to the cell in the absence of the agent, at least one of (i) the rate of growth, (ii) the differentiation state, or (iii) the rate of survival of the cell.
  • Figure IA illustrates the chemical structures of trapoxin and trichostatin, natural products diat inhibit the enzymatic deacetylation of lysine residues near the NH 2 -terminus of histones.
  • the epoxyketone side chain of trapoxin is approximately isosteric with N- acetyl lysine and likely alkylates an active site nucleophile.
  • Figure IB illustrates the copurification of trapoxin binding and histone deacetylase activities.
  • Nuclear proteins from bovine thymus were precipitated with ammonium sulfate and fractionated on a Mono Q column. Trapoxin binding was assayed by charcoal precipitation with [ 3 H]trapoxin.
  • a histone deacetylase assay a peptide corresponding to bovine histone H4 (1-24) was synthesized. The peptide was chemically acetylated with sodium [ 3 H]acetate (5.3 Ci/mmol, New England Nuclear) BOP reagent (Aldrich) and purified by reverse phase HPLC. Two microliters of
  • [ 3 H]peptide( ⁇ 40,000dpm) were used per 200 ⁇ l assay. After incubation at 37°C for one hour, the reaction was quenched with 1 M HC1/0.16 M acetic acid (50 ⁇ l). Released
  • FIG. 2A shows the synthesis of K-trap and the K-trap affinity matrix.
  • K-trap contains a protected lysine residue in place of the phenylalanine at position two in trapoxin.
  • Alloc allyloxycarbonyl.
  • Figure 2B is a silver stained gel showing bovine and human trapoxin binding proteins. Proteins bound to the K-trap affinity matrix in the presence or absence of trapoxin or trichostatin were eluted by boiling in SDS loading buffer and analyzed by SDS-PAGE (9% gel). Nuclear proteins from human Jurkat T cells were prepared identically to those from bovine thymus ( Figure IB). Molecular size standards (in kilodaltons) are indicated to the right.
  • Figure 3 A is the predicted amino acid sequence of human HD An in-frame stop codon was found upstream of the starting methionine. Regions equivalent to microsequenced tryptic peptides from the purified bovine protein are boxed.
  • Underlined amino acids 319-334 and 467-482 denote the sequences of synthetic peptides that were conjugated to KLH and used to generate polyclonal antisera.
  • Abbreviations for the amino acid residues are: A, Ala; C, Cys; D, Asp; E, Glu; F, Phe; G, Gly; H, His; I, He; K, Lys; L, Leu; M, Met; N, Asn; P, Pro; Q, Gin; R, Arg; S, Ser; T, Thr; V, Val; W, Trp; and Y, Tyr.
  • Figure 3B is a protein immunoblot analogous to d e silver stained gel in Figure 2B, showing the relationship between bovine p46-p49 and human p55 (top panels) and confirming the identity of p50 (bovine and human) as RbAp48 (bottom panels).
  • Proteins eluted from the K-trap affinity matrix ( Figure 2) were separated by SDS-PAGE and transferred to Immobilon-P (Millipore). Blots were probed with polyclonal anti-HDl (319-336) or monoclonal anti-RbAp48 and bound antibodies were detected with enhanced chemilurninescence (Amersham).
  • Figure 4A is an immunoprecipitation of endogenous histone deacetylase activity with affinity purified anti-HDl (467-482) antibodies.
  • Anti-HDl (467-482) immunoprecipitates from equivalent amounts of Jurkat nuclear extract (1 mg nuclear protein supplemented with 0.5 M NaCl, 1% BSA, and 0.1% NP-40) were isolated in the presence or absence of HD 1(467-482) peptide competitor. After resuspending the immunoprecipitates in HDx buffer [20 mM tris (p ⁇ 8), 150 mM NaCl, 10% glycerol], inhibitors were added as indicated, and histone deacetylase activity was measured as described in Figure 1 A.
  • Figure 4B shows the coprecipitation of HD1 and RbAp48, as detected by protein immunoblot analysis.
  • Figure 4C demonstrates the histone deacetylase activity of recombinant HD1-F.
  • Tag Jurkat cells (Clipstone et al. (1992) Nature 357, 695-7) were transfected with pFJ5 (vector alone) or pBJ5/HDl-F (encoding COO ⁇ -terminal FLAG epitope tagged HD1) by electroporation and detergent lysates were prepared [0.5% Triton X-100, 50 mM tris (p ⁇ 8), 100 mM NaCl, 10% glycerol].
  • Anti-FLAG antibodies conjugated to agarose beads IBI were used to immunoprecipitate recombinant HD1 in the presence or absence of FLAG peptide competitor, and histone deacetylase activity was measured as described above.
  • Figure 4D shows die interaction between recombinant ⁇ D1-F and the K-trap affinity matrix.
  • Lysates from Jurkat cells transfected with pBJ5/ ⁇ Dl-F were incubated with the K-trap affinity matrix in the presence or absence of inhibitors.
  • Immunoblots of the eluted proteins were probed with the anti-FLAG M2 monoclonal antibody (IBI).
  • Figures 5 A and 5B are sequence alignments for various HDx and HDx-related cDNAs and proteins, respectively.
  • Figure 6 depicts exemplary specificity elements (A), linker elements (B), and electrophilic moieties (C) for generating compounds which are capable of reacting with a nucleophilic moiety of an HDx protein.
  • Figure 7 illustrates an exemplary synthesis of trichostatin analogs.
  • Figures 8A-8C illustrate a synthesis of tritiated Trapoxin B.
  • Figures 9A-9C depict a synthesis of the K-trap and K-trap affinity matrix .
  • FIGS 10A-10B mSin3A is present in cells as a large stable multiprotein complex. Nuclear lysates were prepared from U937 cells metabolically labeled with [ 35 ]S-methionine and low stringency immunoprecipitations performed with antiserum specific for mSin3A. "+ block” shows proteins immunoprecipitated when the anti- mSin3 A was preincubated with purified GST-PA ⁇ 2 (A). In (B), low stringency mSin3A immunoprecipitates were washed for an additional 60 minutes using the salt and detergent conditions indicated at the top of die Figure. In (A) and (B), the immunoprecipitates were analyzed by SDS-PAGE and autoradiography. Apparent molecular weight of the coprecipitating proteins and the sizes of die molecular weight markers are given in kilodaltons.
  • FIGS 11 A-D mSin3A and EMACI associate in vivo. Immunoprecipitations were performed using nuclear extracts from [ 35 ]S-methionine labeled U937 cells.
  • A The left lane shows proteins from an anti-mSin3A immunoprecipitate.
  • the right lane shows proteins eluted from an anti-mSin3A immunoprecipitate and reprecipitated with anti ⁇ serum specific for HD1.
  • B and (C) low stringency immunoprecipitations were performed using antiserum specific for the carboxy-terminus of HD1. "+ block" indicates diat the HD1 antiserum was preincubated with the immunizing peptide.
  • proteins immunoprecipitated with anti-mS3A are shown for reference, protei ⁇ s eluted from a low stringency anti-HDl immunoprecipitate and reprecipitated with anti-mSin3A are shown in die right most lane.
  • A), (B), and (C) autoradiographs of SDS-PAGE gels are shown. Apparent molecular weight of the coprecipitating, proteins and die sizes of the molecular weight markers are given in kilodaltons.
  • D in vitro histone deacetylase activity in anti-mSin3A immunoprecipitates is shown.
  • Figures 13A-C Trapoxin reverses transcriptional repression by mSin3A.
  • A The structure of the minimal reporter gene derived from the myelomonocytic growth factor gene and the expression vectors.
  • Mad(Pro)N35GALVPI6 has leucine at position 12 and alanine at position 16 mutated to proline as indicated. These point mutations prevent association between mSin3A and Mad (Ayer et al., 1995).
  • the transcriptional activity of MadN35GALVP16 and Mad(Pro)N35GALVP16 was determined by measuring luciferase activity (Relative Light Units, RLU) of transfected 293 cells following an 8 hour treatment with 0 (solid bars) or 10 nM trapoxin (striped bars) (B). To control for differences in transfection efficiency, the RLU values were normalized to the ⁇ -galactosidase activity produced by a cotransfected CMV- ⁇ GAL construct. Shown is data from representative experiment and the error is reported as die standard error of the mean (s.e.m). This experiment has been done a minimum of five times in triplicate with similar results.
  • RLU Relative Light Units
  • trapoxin inhibits histone deacetylase activity of human 293 cells in vivo.
  • 2 x IO 8 cells were cultured for 8 hours in the absence, "O", or in the presence of 10 nM trapoxin.
  • Cells were harvested and crude extracts from approximately 1 x IO 7 cells (solid bars) or anti- HDl immunoprecipitations of extracts from approximately 4 x IO 7 cells (white bars) were assayed for histone deacetylase activity in vitro.
  • nucleosomes The positioning of nucleosomes relative to particular regulatory elements in genomic DNA has emerged as a mechanism for managing the association of sequence- specific DNA-binding proteins with promoters, enhancers and odier transcriptional regulatory sequences. Two modifications to nucleosomes have been observed to influence the association of DNA-binding proteins with chromatin. Depletion of histones H2A/H2B from the nucleosome facilitates the binding of RNA polymerase II (Baer et al. (1983) Nature 301:482-488) and TFIHA (Hayes et al. (1992) PNAS 89:1229-1233).
  • acetylation of die core histones apparently destabilizes the nucleosome and is thought to modulate the accessibility of transcription factors to their respective enhancer and promoter elements (Oliva et al. (1990) Nuc Acid Res 18:2739-2747; and Walker et al. (1990) J Biol Chem 265:5622-5746). In both cases, overall histone-DNA contacts are altered.
  • die present invention concerns die discovery of a family of genes in mammals, the gene products of which are referred to herein as "histone deacetylases" or "HDxV.
  • HDxV histone deacetylases
  • Experimental evidence indicates a functional role for the HDx gene products as catalysts of the deacetylation of histones in mammalian cells, and accordingly play a role in determining tissue fate and maintenance.
  • the results provided below indicate that proteins encoded by the HDx genes may participate, under various circumstances, in the control of proliferation, differentiation and cell death.
  • the family of HDx gene apparently encode at least three different sub-families, e.g., paralogs, and have been identified from the cells of various mammals.
  • the HDx proteins were first isolated from bovine thymus nuclei by use of a binding assay which exploited the ability of trapoxin, an inhibitor of histone deacetylase activity, to isolate proteins which co-purified widi a histone acetylase activity.
  • the partial identity of the isolated proteins were determined by peptide microsequencing, and primers based on the peptide sequences were used to clone human cDNAs from a T cell library.
  • HDl One of the HDx gene products described below is referred to herein as HDl, and is represented in SEQ ID No.
  • the first apparently conserved structural element of the HDx family occurs in the N- terminal portion of the molecule, and is designated herein as the "v motif'.
  • d e v motif corresponds to amino acid residues Asp 130- Phe 198.
  • DXXX TXXC ⁇ LHHAKKXEASGFCy ⁇ VMTXSF (SEQ ID No. 12) more preferably by the consensus sequence: DDVX 1 iNWAGGLHHAKKX2EASGFCY t roiV?C3X 4 7 TD-RVMTVSF (SEQ ID No. 13) wherein each of X n represents any single amino acid, though more preferably represents an amino acid residue in the corresponding human HDx sequences of the appended sequence listing.
  • a second motif, herein designated the ⁇ motif is represented by die consensus sequence:
  • the family of HDx proteins apparently ranges in size from about 40kd to about
  • certain aspects of the present invention relate to nucleic acids encoding HDx proteins, the HDx proteins themselves, antibodies immunoreactive with HDx proteins, and preparations of such compositions. Moreover, the present invention provides diagnostic and therapeutic assays and reagents for detecting and treating disorders involving, for example, aberrant expression (or loss thereof) of HDx homologs.
  • drug discovery assays are provided for identifying agents which can modulate the biological function of HDx proteins, such as by altering the binding of HDx molecules to either proteins or nucleic acids. Such agents can be useful therapeutically to alter the growth and/or differentiation of a cell.
  • agents which can modulate the biological function of HDx proteins, such as by altering the binding of HDx molecules to either proteins or nucleic acids.
  • Such agents can be useful therapeutically to alter the growth and/or differentiation of a cell.
  • drug discovery assays are provided for identifying agents which can modulate the biological function of "HDx-related" proteins, such as RPD3 homologs, by altering the enzymatic activity of the deacetylase, or its binding to odier cellular components including homologs of RbAp48 (described infra).
  • agents can be useful therapeutically to alter the growth and/or differentiation of non-human cells, such as in the treatment of mycotic infections, or as additives to livestock feed, e.g., to promote weight gain, or as topical antiseptics for sterilizing medical equipment.
  • RbAp48 (Qian et al. (1993) Nature 364:648; SEQ ID No. 11).
  • RbAp48 was originally identified as a protein that binds to the retinoblastoma (Rb) gene product.
  • the retinoblastoma (RB) gene product plays a role in tumor suppression (Weinberg, R.A., (Sept 1988) Scientific Amer.pp 44-51; Hansen et al. (1988) Trends Genet 4:125- 128).
  • RB as a tumor-suppressor protein in cell-cycle control
  • p53 Green (1989) Cell 56:1-3; Mowat et al (1985 Nature 314:633-636).
  • Inactivation or mutation of the second RB allele in one of the somatic cells of these susceptible individuals appears to be the molecular event that leads to tumor formation (Caveneee et al. (1983) Nature 305:799-784; Friend et al. (1987) PNAS 84:9059-9063).
  • RbAp48 is one of the major proteins that binds to a putative functional domain at the carboxy terminus of the Rb protein. Complex formation between RbAp48 and Rb occurs in vitro and in vivo, and apparently involves direct interaction between the proteins. Like Rb, RbAp48 is a ubiquitously expressed nuclear protein. RbAp48 share sequence homology with MS11, a negative regulator of the Ras-cyclic AMP pathway in the yeast Saccharomyces cerevisiae. Furthermore, like MS11, human RbAp48 suppresses the heat-shock sensitivity of the yeast iral strains and RAS2Vall9 strains. Interaction with RbAp48 may be one of the mechanisms for suppression of growth mediated by Rb. Accordingly, d e interaction of RbAp48 with HDx proteins further implicates the HDx proteins in cell-cycle regulation.
  • RpAp48 interaction with HDx and HDx-related proteins represents yet another therapeutic target.
  • drug discovery assays are provided for identifying agents which can modulate the interaction of RbAp48 proteins and the like with HDx-related proteins.
  • Such assays can be derived to detect the ability of a test agent to alter protein-protein contacts, or to alter the enzymatic activity of the deacetylase in complexes including an RbAp48 protein (e.g., were such complexes allosterically modulate the HDx enzymatic activity).
  • such agents can be useful therapeutically to alter the growth and/or differentiation of cells.
  • Sin3 proteins are an in vivo component of large, heterogeneous multiprotein complexes and is tightly and specifically associated with at least seven polypeptides.
  • Two of the Sin3-associated proteins, p50 and p55, are members of the histone deacetylase family described herein.
  • Sin3 immunecomplexes possess histone deacetylase activity that is sensitive to the specific deacetylase inhibitor trapoxin.
  • Sin3 targeted repression of a reporter gene is reduced by trapoxin treatment, suggesting that histone deacetylation mediates transcriptional repression through Mad-Max-Sin3 A multimeric complexes.
  • the Sin3 interaction with HDx and HDx-related proteins represents still another therapeutic target.
  • drug discovery assays for identifying agents which can modulate the interaction of Sin3 proteins and the like with HDx-related proteins.
  • Such assays can be derived to detect the ability of a test agent to alter protein-protein contacts, or to alter the enzymatic activity of the deacetylase in complexes including Sin3 or other transcriptional regulatory proteins.
  • such agents can be useful therapeutically to alter the growth and/or differentiation of cells.
  • nucleic acid refers to polynucleotides such as deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA).
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • the term should also be understood to include, as equivalents, analogs of either RNA or
  • DNA made from nucleotide analogs, and, as applicable to the embodiment being described, single (sense or antisense) and double-stranded polynucleotides.
  • the term “gene” or “recombinant gene” refers to a nucleic acid comprising an open reading frame encoding one of the HDx polypeptides of the present invention, including both exon and (optionally) intron sequences.
  • a “recombinant gene” refers to nucleic acid encoding an HDx polypeptide and comprising HDx-encoding exon sequences, though it may optionally include intron sequences which are either derived from a chromosomal HDx gene or from an unrelated chromosomal gene. Exemplary recombinant genes encoding the subject HDx polypeptide are represented in the appended Sequence Listing.
  • the term “intron” refers to a DNA sequence present in a given HDx gene which is not translated into protein and is generally found between exons.
  • transfection means the introduction of a nucleic acid, e.g., an expression vector, into a recipient cell by nucleic acid-mediated gene transfer.
  • Transformation refers to a process in which a cell's genotype is changed as a result of the cellular uptake of exogenous DNA or RNA, and, for example, die transformed cell expresses a recombinant form of an HDx polypeptide or, where anti ⁇ sense expression occurs from die transferred gene, the expression of a naturally- occurring form of die HDx protein is disrupted.
  • the term “specifically hybridizes” refers to the ability of the probe/primer of die invention to hybridize to at least 15 consecutive nucleotides of an HDx gene, such as an HDx sequence designated in one of SEQ ID Nos: 1-4, or a sequence complementary thereto, or naturally occurring mutants thereof, such that it has less than 15%, preferably less than 10%, and more preferably less than 5% background hybridization to a cellular nucleic acid (e.g., mRNA or genomic DNA) encoding a protein other than an HDx protein, as defined herein.
  • the oligonucleotide probe specifically detects only one of the subject HDx paralogs, e.g., does not substantially hybridize to transcripts for other HDx homologs in the same species.
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • One type of preferred vector is an episome, i.e., a nucleic acid capable of extra-chromosomal replication.
  • Preferred vectors are diose capable of autonomous replication and/expression of nucleic acids to which they are linked.
  • Vectors capable of directing the expression of genes to which they are operatively linked are referred to herein as "expression vectors”.
  • expression vectors of utility in recombinant DNA techniques are often in the form of "plasmids" which refer generally to circular double stranded DNA loops which, in their vector form are not bound to the chromosome.
  • plasmid and “vector” are used interchangeably as d e plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors which serve equivalent functions and which become known in the art subsequently hereto.
  • Transcriptional regulatory sequence is a generic term used throughout the specification to refer to DNA sequences, such as initiation signals, enhancers, and promoters, which induce or control transcription of protein coding sequences widi which they are operably linked.
  • transcription of one of the recombinant HDx genes is under the control of a promoter sequence (or other transcriptional regulatory sequence) which controls the expression of the recombinant gene in a cell-type in which expression is intended.
  • tissue-specific promoter means a DNA sequence that serves as a promoter, i.e., regulates expression of a selected DNA sequence operably linked to the promoter, and which effects expression of the selected DNA sequence in specific cells of a tissue, such as cells of hepatic, pancreatic, neuronal or hematopoietic origin.
  • tissue-specific promoter means a DNA sequence that serves as a promoter, i.e., regulates expression of a selected DNA sequence operably linked to the promoter, and which effects expression of the selected DNA sequence in specific cells of a tissue, such as cells of hepatic, pancreatic, neuronal or hematopoietic origin.
  • leaky promoters, which regulate expression of a selected DNA primarily in one tissue, but can cause at least low level expression in other tissues as well.
  • a "transgenic animal” is any animal, preferably a non-human mammal, bird or an amphibian, in which one or more of the cells of the animal contain heterologous nucleic acid introduced by way of human intervention, such as by transgenic techniques well known in the art.
  • the nucleic acid is introduced into the cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection wid a recombinant virus.
  • the term genetic manipulation does not include classical cross-breeding, or in vitro fertilization, but rather is directed to the introduction of a recombinant DNA molecule. This molecule may be integrated within a chromosome, or it may be extrachromosomally replicating DNA.
  • transgenic animal In the typical transgenic animals described herein, the transgene causes cells to express a recombinant form of one of the HDx proteins, e.g. either agonistic or antagonistic forms.
  • transgenic animals in which the recombinant HDx gene is silent are also contemplated, as for example, the FLP or CRE recombinase dependent constructs described below.
  • transgenic animal also includes those recombinant animals in which gene disruption of one or more HDx genes is caused by human intervention, including both recombination and antisense techniques.
  • non-human animals include vertebrates such as rodents, non-human primates, sheep, dog, cow, chickens, amphibians, reptiles, etc.
  • Preferred non- human animals are selected from the rodent family including rat and mouse, most preferably mouse, though transgenic amphibians, such as members of the Xenopus genus, and transgenic chickens can also provide important tools for understanding and identifying agents which can affect, for example, embryogenesis and tissue formation.
  • transgenic insects including those of the genus Drosophila, such as D. melanogaster.
  • chimeric animal is used herein to refer to animals in which the recombinant gene is found, or in which the recombinant is expressed in some but not all cells of the animal.
  • tissue-specific chimeric animal indicates that one of the recombinant HDx genes is present and/or expressed or disrupted in some tissues but not others.
  • transgene means a nucleic acid sequence (encoding, e.g., one of the HDx polypeptides, or pending an antisense transcript thereto), which is partly or entirely heterologous, i.e., foreign, to the transgenic animal or cell into which it is introduced, or, is homologous to an endogenous gene of the transgenic animal or cell into which it is introduced, but which is designed to be inserted, or is inserted, into the animal's genome in such a way as to alter the genome of the cell into which it is inserted (e.g., it is inserted at a location which differs from that of the natural gene or its insertion results in a knockout).
  • a transgene can include one or more transcriptional regulatory sequences and any other nucleic acid, such as introns, that may be necessary for optimal expression of a selected nucleic acid.
  • genes for a particular polypeptide may exist in single or multiple copies within the genome of an individual. Such duplicate genes may be identical or may have certain modifications, including nucleotide substitutions, additions or deletions, which all still code for polypeptides having substantially the same activity.
  • the term "DNA sequence encoding an HDx polypeptide" may thus refer to one or more genes within a particular individual.
  • certain differences in nucleotide sequences may exist between individuals of the same species, which are called alleles.
  • allelic differences may or may not result in differences in amino acid sequence of the encoded polypeptide yet still encode a protein with the same biological activity.
  • Homology refers to sequence similarity between two peptides or between two nucleic acid molecules.
  • Homology can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base or amino acid, then the molecules are homologous at that position. A degree of homology between sequences is a function of die number of matching or homologous positions shared by the sequences. An "unrelated" or “non-homologous" sequence shares less tiian 40 percent identity, though preferably less than 25 percent identity, with one of the HDx sequences of the present invention.
  • an "HDx-related" protein refers to die HDx proteins described herein, and other human homologs of those HDx sequences, as well as orthologs and paralogs (homologs) of the HDx proteins in other species, ranging from yeast to other mammals, e.g., homologous histone deacetylase,
  • the term "ortholog” refers to genes or proteins which are homologs via speciation, e.g., closely related and assumed to have common descent based on structural and functional considerations. Orthologous proteins function as recognizably the same activity in different species.
  • paralog refers to genes or proteins which are homologs via gene duplication, e.g., duplicated variants of a gene within a genome. See also, Fritch, WM (1970) Syst Zool 19:99-113.
  • Cells “host cells” or “recombinant host cells” are terms used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • a "chimeric protein” or “fusion protein” is a fusion of a first amino acid sequence encoding one of the subject HDx polypeptides with a second amino acid sequence defining a domain (e.g. polypeptide portion) foreign to and not substantially homologous with any domain of one of the HDx proteins.
  • a chimeric protein may present a foreign domain which is found (albeit in a different protein) in an organism which also expresses the first protein, or it may be an "interspecies", "intergenic”, etc. fusion of protein structures expressed by different kinds of organisms.
  • a fusion protein can be represented by the general formula X-HDx-Y, wherein HDx represents a portion of the protein which is derived from one of the HDx proteins, and X and Y are, independently, absent or represent amino acid sequences which are not related to one of the HDx sequences in an organism.
  • an isolated nucleic acid encoding one of the subject HDx polypeptides preferably includes no more than 10 kilobases (kb) of nucleic acid sequence which naturally immediately flanks the HDx gene in genomic DNA, more preferably no more than 5kb of such naturally occurring flanking sequences, and most preferably less than 1.5kb of such naturally occurring flanking sequence.
  • kb kilobases
  • isolated also refers to a nucleic acid or peptide that is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or odier chemicals when chemically synthesized.
  • isolated nucleic acid is meant to include nucleic acid fragments which are not naturally occurring as fragments and would not be found in the natural state.
  • nucleic acid as used herein is intended to include fragments as equivalents.
  • equivalent is understood to include nucleotide sequences encoding functionally equivalent HDx polypeptides or functionally equivalent peptides having an activity of an HDx protein such as described herein.
  • Equivalent nucleotide sequences will include sequences that differ by one or more nucleotide substitutions, additions or deletions, such as allelic variants; and will, therefore, include sequences that differ from the nucleotide sequence of the HDx cDNA sequences shown in any of SEQ ID Nos: 1-4 due to the degeneracy of the genetic code. Equivalents will also include nucleotide sequences diat hybridize under stringent conditions (i.e., equivalent to about 20-27°C below the melting temperature (Tm) of the DNA duplex formed in about IM salt) to the nucleotide sequences represented in one or more of SEQ ID Nos: 1-4. In one embodiment, equivalents will further include nucleic acid sequences derived from and evolutionarily related to, a nucleotide sequences shown in any of SEQ ID Nos: 1-4.
  • an HDx agonist mimetic
  • an HDx antagonist e.g., an HDx antagonist
  • Homologs of each of the subject HDx proteins can be generated by mutagenesis, such as by discrete point mutation(s), or by truncation. For instance, mutation can give rise to homologs which retain substantially the same, or merely a subset, of the biological activity of the HDx polypeptide from which it was derived.
  • antagonistic forms of the protein can be generated which are able to inhibit the function of the naturally occurring form of the protein, such as by competitively binding to an HDx substrate or HDx associated protein, as for example competing with wild-type HDx in the binding of RbAp48 or a histone.
  • agonistic forms of the protein may be generated which are constitutively active, or have an altered K ⁇ or K m for deacetylation reactions.
  • the HDx protein and homologs thereof provided by the subject invention may be either positive or negative regulators of transcription and/or replication.
  • polypeptides referred to herein as having an activity of an HDx protein are defined as polypeptides which include an amino acid sequence corresponding (e.g., identical or homologous) to all or a portion of die arnino acid sequences of an HDx proteins shown in any one or more of SEQ ID Nos:5-8 and which mimic or antagonize all or a portion of the biological/biochemical activities of a naturally occurring HDx protein.
  • biological activity include the ability to modulate proliferation of cells. For example, inhibiting histone deacetylation causes cells to arrest in GI and G2 phases of the cell cycle.
  • the biochemical activity associated with HDx proteins of the present invention can also characterized in terms of binding to and (optionally) catalyzing the deacetylation of an acetylated histone.
  • Another biochemical property of certain of the subject HDx proteins involves binding to other cellular proteins, such as RbAp48 or Sin3A.
  • Other biological activities of die subject HDx proteins are described herein or will be reasonably apparent to those skilled in the art.
  • a polypeptide has biological activity if it is a specific agonist or antagonist of a naturally- occurring form of an HDx protein.
  • nucleic acids encode an HDx polypeptide comprising an amino acid sequence at least 80% homologous, more preferably at least 85% homologous and most preferably at least 88% homologous with an amino acid sequence of a human HDx, e.g., such as selected from the group consisting of SEQ ID Nos: 5-8.
  • Nucleic acids which encode polypeptides at least about 90%, more preferably at least about 95%, and most preferably at least about 98-99% homology with an amino acid sequence represented in one of SEQ ID Nos: 5-8 are of course also within the scope of the invention, as are nucleic acids identical in sequence with any of the enumerated HDx sequences of the sequence listing.
  • the nucleic acid is a cDNA encoding a polypeptide having at least one activity of the subject HDx polypeptide.
  • the invention features a purified or recombinant HDx polypeptide having peptide chain with a molecular weight in the range of 40kd to 60kd, even more preferably in the range of 45-50 kd or 53-58kd. It will be understood diat certain post-translational modifications, e.g., phosphorylation and the like, can increase the apparent molecular weight of the HDx protein relative to the unmodified polypeptide chain, and cleavage of certain sequences, such as pro-sequences, can likewise decrease the apparent molecular weight.
  • the nucleic acid encodes an HDx polypeptide which includes both the v and ⁇ motifs, and preferably possess a histone deacetylase activity.
  • preferred HDx proteins are represented by the general formula A- (v motif)-B-( ⁇ motif)-C, wherein the v motif is an amino acid sequence represented in SEQ ID NO. 12, more preferably SEQ ID No. 13, the ⁇ motif is an amino acid sequence represented in SEQ ID No. 14, more preferably SEQ ID No. 15, and A, B and C represent amino acid sequences which are correspond to HDx or HDx-related proteins.
  • nucleic acids of the present invention encode an HDx polypeptide which includes a polypeptide sequence corresponding to all or a portion of amino acid residues of any one of SEQ ID Nos: 5-8, e.g., at least 5, 10, 25, 50 or 100 amino acid residues of that region.
  • nucleic acid which hybridizes under high or low stringency conditions to the nucleic acid represented by SEQ ID No: 1.
  • Appropriate stringency conditions which promote DNA hybridization for example, 6.0 x sodium chloride/sodium citrate (SSC) at about 45°C, followed by a wash of 2.0 x SSC at 50°C, are known to those skilled in the art or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6.
  • the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at 50°C to a high stringency of about 0.2 x SSC at 50°C.
  • the temperature in the wash step can be increased from low stringency conditions at room temperature, about 22°C, to high stringency conditions at about 65°C.
  • Nucleic acids having a sequence that differs from the nucleotide sequences shown in one of SEQ ID Nos: 1-4 due to degeneracy in the genetic code are also within the scope of the invention.
  • Such nucleic acids encode functionally equivalent peptides (i.e., a peptide having a biological activity of an HDx polypeptide) but differ in sequence from the sequence shown in the sequence listing due to degeneracy in the genetic code. For example, a number of amino acids are designated by more than one triplet. Codons diat specify the same amino acid, or synonyms (for example, CAU and CAC each encode histidine) may result in "silent" mutations which do not affect die amino acid sequence of an HDx polypeptide.
  • DNA sequence polymorphisms that do lead to changes in the amino acid sequences of the subject HDx polypeptides will exist among, for example, humans.
  • these variations in one or more nucleotides (up to about 3-5% of the nucleotides) of the nucleic acids encoding polypeptides having an activity of an HDx polypeptide may exist among individuals of a given species due to natural allelic variation.
  • an HDx gene fragment refers to a nucleic acid having fewer nucleotides than the nucleotide sequence encoding the entire mature form of an HDx protein yet which (preferably) encodes a polypeptide which retains some biological activity of the full length protein. Fragment sizes contemplated by the present invention include, for example, 5, 10, 25, 50, 75, 100, or 200 amino acids in length.
  • HDx protein-encoding nucleic acids can be obtained from mRNA present in any of a number of eukaryotic cells. It should also be possible to obtain nucleic acids encoding HDx polypeptides of the present invention from genomic DNA from both adults and embryos.
  • a gene encoding an HDx protein can be cloned from either a cDNA or a genomic library in accordance with protocols described herein, as well as those generally known to persons skilled in the art.
  • a cDNA encoding an HDx protein can be obtained by isolating total mRNA from a cell, e.g. a mammalian cell, e.g. a human cell, including embryonic cells.
  • Double stranded cDNAs can then be prepared from the total mRNA, and subsequently inserted into a suitable plasmid or bacteriophage vector using any one of a number of known techniques.
  • the gene encoding an HDx protein can also be cloned using established polymerase chain reaction techniques in accordance with the nucleotide sequence information provided by the invention.
  • the nucleic acid of the invention can be DNA or RNA.
  • a preferred nucleic acid is a cDNA including a nucleotide sequence represented by one of SEQ ID Nos: 1-4.
  • antisense therapy refers to administration or in situ generation of oligonucleotide probes or their derivatives which specifically hybridize (e.g. binds) under cellular conditions, with the cellular mRNA and/or genomic DNA encoding one or more of the subject HDx proteins so as to inhibit expression of diat protein, e.g. by inhibiting transcription and/or translation.
  • the binding may be by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interactions in the major groove of the double helix.
  • antisense refers to the range of techniques generally employed in the art, and includes any therapy which relies on specific binding to oligonucleotide sequences.
  • an antisense construct of the present invention can be delivered, for example, as an expression plasmid which, when transcribed in the cell, produces RNA which is complementary to at least a unique portion of the cellular mRNA which encodes an HDx protein.
  • the antisense construct is an oligonucleotide probe which is generated ex vivo and which, when introduced into the cell causes inhibition of expression by hybridizing with the mRNA and/or genomic sequences of an HDx gene.
  • Such oligonucleotide probes are preferably modified oligonucleotides which are resistant to endogenous nucleases, e.g. exonucleases and/or endonucleases, and are therefore stable in vivo.
  • Exemplary nucleic acid molecules for use as antisense oligonucleotides are phosphoramidate, phosphothioate and methylphosphonate analogs of DNA (see also U.S.
  • the modified oligomers of the invention are useful in therapeutic, diagnostic, and research contexts.
  • the oligomers are utilized in a manner appropriate for antisense therapy in general.
  • the oligomers of the invention can be formulated for a variety of routes of administration, including systemic and topical or localized administration. Techniques and formulations generally may be found in Remmington's Pharmaceutical Sciences, Meade Publishing Co., Easton, PA.
  • injection is preferred, including intramuscular, intravenous, intraperitoneal, and subcutaneous.
  • the oligomers of the invention can be formulated in liquid solutions, preferably in physiologically compatible buffers such as Hank's solution or Ringer's solution.
  • oligomers may be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms are also included.
  • Systemic administration can also be by transmucosal or transdermal means, or the compounds can be administered orally.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration bile salts and fusidic acid derivatives.
  • detergents may be used to facilitate permeation.
  • Transmucosal administration may be through nasal sprays or using suppositories.
  • the oligomers are formulated into conventional oral administration forms such as capsules, tablets, and tonics.
  • the oligomers of the invention are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the oligomers of the invention may be used as diagnostic reagents to detect the presence or absence of the target DNA or RNA sequences to which they specifically bind. Such diagnostic tests are described in further detail below.
  • die antisense constructs of the present invention by antagonizing the normal biological activity of one of the HDx proteins, can be used in the manipulation of tissue, e.g. tissue differentiation or growth, both in vivo and ex vivo.
  • die anti-sense techniques e.g. microinjection of antisense molecules, or transfection with plasmids whose transcripts are anti-sense with regard to an HDx mRNA or gene sequence
  • Such techniques can be utilized in cell culture, but can also be used in d e creation of transgenic animals (described infra).
  • This invention also provides expression vectors containing a nucleic acid encoding an HDx polypeptide, operably linked to at least one transcriptional regulatory sequence.
  • Operably linked is intended to mean that the nucleotide sequence is linked to a regulatory sequence in a manner which allows expression of the nucleotide sequence.
  • Regulatory sequences are art-recognized and are selected to direct expression of the subject HDx proteins. Accordingly, the term transcriptional regulatory sequence includes promoters, enhancers and other expression control elements. Such regulatory sequences are described in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990).
  • any of a wide variety of expression control sequences, sequences diat control the expression of a DNA sequence when operatively linked to it, may be used in these vectors to express DNA sequences encoding HDx polypeptides of this invention.
  • useful expression control sequences include, for example, a viral LTR, such as the LTR of the Moloney murine leukemia virus, the early and late promoters of SV40, adenovirus or cytomegalovirus immediate early promoter, the lac system, the trp system, the TAC or TRC system, T7 promoter whose expression is directed by T7 RNA polymerase, the major operator and promoter regions of phage ⁇ , the control regions for fd coat protein, the promoter for 3- phosphogiycerate kinase or other glycolytic enzymes, the promoters of acid phosphatase, e.g., Pho5, the promoters of die yeast ⁇ -mating factors, the polyhedron promoter of the bacul
  • the design of the expression vector may depend on such factors as the choice of the host cell to be transformed and/or the type of protein desired to be expressed. Moreover, the vector's copy number, the ability to control that copy number and the expression of any other proteins encoded by the vector, such as antibiotic markers, should also be considered.
  • the expression vector includes a recombinant gene encoding a peptide having an agonistic activity of a subject HDx polypeptide, or alternatively, encoding a peptide which is an antagonistic form of the HDx protein, such as a catalytically-inactive deacetylase.
  • Such expression vectors can be used to transfect cells and thereby produce polypeptides, including fusion proteins, encoded by nucleic acids as described herein.
  • the gene constructs of the present invention can also be used as a part of a gene therapy protocol to deliver nucleic acids, e.g., encoding either an agonistic or antagonistic form of one of the subject HDx proteins or an antisense molecule described above.
  • another aspect of the invention features expression vectors for in vivo or in vitro transfection and expression of an HDx polypeptide or antisense molecule in particular cell types so as to reconstitute the function of, or alternatively, abrogate the function of HDx-induced transcription in a tissue in which the naturally-occurring form of the protein is misexpressed; or to deliver a form of the protein which alters differentiation of tissue, or which inhibits neoplastic transformation.
  • Expression constructs of the subject HDx polypeptides, as well as antisense constructs may be administered in any biologically effective carrier, e.g. any formulation or composition capable of effectively delivering the recombinant gene to cells in vivo.
  • Approaches include insertion of the subject gene in viral vectors including recombinant retroviruses, adenovirus, adeno-associated virus, and herpes simplex virus- 1, or recombinant bacterial or eukaryotic plasmids.
  • Viral vectors transfect cells directly; plasmid DNA can be delivered with the help of, for example, cationic liposomes (lipofectin) or derivatized (e.g. antibody conjugated), polylysine conjugates, gramacidin S, artificial viral envelopes or other such intracellular carriers, as well as direct injection of the gene construct or CaPO precipitation carried out in vivo.
  • transduction of appropriate target cells represents the critical first step in gene therapy
  • choice of the particular gene delivery system will depend on such factors as the phenotype of the intended target and the route of administration, e.g. locally or systemically.
  • the particular gene construct provided for in vivo transduction of HDx expression are also useful for in vitro transduction of cells, such as for use in the ex vivo tissue culture systems described below.
  • a preferred approach for in vivo introduction of nucleic acid into a cell is by use of a viral vector containing nucleic acid, e.g. a cDNA encoding the particular HDx polypeptide desired.
  • a viral vector containing nucleic acid e.g. a cDNA encoding the particular HDx polypeptide desired.
  • Infection of cells with a viral vector has the advantage that a large proportion of the targeted cells can receive the nucleic acid.
  • molecules encoded within the viral vector e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells which have taken up viral vector nucleic acid.
  • Retrovirus vectors, adenovirus vectors and adeno-associated virus vectors are exemplary recombinant gene delivery system for the transfer of exogenous genes in vivo, particularly into humans. These vectors provide efficient delivery of genes into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host.
  • non-viral methods can also be employed to cause expression of a subject HDx polypeptide in the tissue of an animal.
  • Most nonviral methods of gene transfer rely on normal mechanisms used by mammalian cells for the uptake and intracellular transport of macromolecules.
  • non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject HDx polypeptide gene by the targeted cell.
  • Exemplary gene delivery systems of this type include liposomal derived systems, poly-lysine conjugates, and artificial viral envelopes.
  • the gene delivery systems for the therapeutic HDx gene can be introduced into a patient by any of a number of methods, each of which is familiar in the art.
  • a pharmaceutical preparation of the gene delivery system can be introduced systemically, e.g. by intravenous injection, and specific transduction of the protein in the target cells occurs predominantly from specificity of transfection provided by the gene delivery vehicle, cell-type or tissue-type expression due to the transcriptional regulatory sequences controlling expression of the receptor gene, or a combination thereof.
  • initial delivery of the recombinant gene is more limited with introduction into the animal being quite localized.
  • the gene delivery vehicle can be introduced by catheter (see U.S. Patent 5,328,470) or by stereotactic injection (e.g.
  • AN HDx gene such as any one of the clones represented in the group consisting of SEQ ID NO: 1-4, can be delivered in a gene therapy construct by electroporation using techniques described, for example, by Dev et al. ((1994) Cancer Treat Rev 20: 105-115).
  • the pharmaceutical preparation of the gene therapy construct can consist essentially of the gene delivery system in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the pharmaceutical preparation can comprise one or more cells which produce the gene delivery system.
  • HDx polypeptides preferred by the present invention are at least 80% homologous, more preferably at least 85% homologous and most preferably at least 88% homologous with an amino acid sequence represented by any of SEQ ID Nos: 5-8.
  • Polypeptides which possess an activity of an HDx protein (i.e. either agonistic or antagonistic), and which are at least 90%, more preferably at least 95%, and most preferably at least about 98-99% homologous with a sequence selected from the group consisting of SEQ ID Nos: 5-8 are also within the scope of the invention.
  • the HDx polypeptide includes both the v and ⁇ motifs, and preferably possess a histone deacetylase activity.
  • recombinant HDx protein refers to a polypeptide which is produced by recombinant DNA techniques, wherein generally, DNA encoding an HDx polypeptide is inserted into a suitable expression vector which is in turn used to transform a host cell to produce the heterologous protein.
  • the phrase "derived from”, with respect to a recombinant HDx gene is meant to include within the meaning of "recombinant protein” those proteins having an amino acid sequence of a native HDx protein, or an amino acid sequence similar thereto which is generated by mutations including substitutions and deletions (including truncation) of a naturally occurring form of the protein.
  • the present invention further pertains to recombinant forms of the subject HDx polypeptides which are encoded by genes derived from a mammal (e.g. a human), and which have amino acid sequences evolutionarily related to the HDx proteins represented in SEQ ID Nos: 5-8.
  • Such recombinant HDx polypeptides preferably are capable of functioning in one of either role of an agonist or antagonist of at least one biological activity of a wild-type ("authentic") HDx protein of the appended sequence listing.
  • the present invention also provides methods of producing the subject HDx polypeptides.
  • a host cell transfected with a nucleic acid vector directing expression of a nucleotide sequence encoding the subject polypeptides can be cultured under appropriate conditions to allow expression of the peptide to occur.
  • the cells may be harvested, lysed and the protein isolated.
  • a cell culture includes host cells, media and other byproducts. Suitable media for cell culture are well known in the art.
  • the recombinant HDx polypeptide can be isolated from cell culture medium, host cells, or both using techniques known in the art for purifying proteins including ion-exchange chromatography, gel filtration chromatography, ultrafiltration, electrophoresis, and immunoaffinity purification with antibodies specific for such peptide.
  • the recombinant HDx polypeptide is a fusion protein containing a domain which facilitates its purification, such as GST fusion protein or poly( ⁇ is) fusion protein.
  • This invention also pertains to a host cell transfected to express recombinant forms of the subject HDx polypeptides.
  • the host cell may be any prokaryotic or eukaryotic cell.
  • a nucleotide sequence derived from the cloning of HDx proteins, encoding all or a selected portion of a full-length protein can be used to produce a recombinant form of an HDx polypeptide via microbial or eukaryotic cellular processes.
  • a gene construct such as an expression vector
  • transforming or transfecting into hosts either eukaryotic (yeast, avian, insect or mammalian) or prokaryotic (bacterial cells) are standard procedures used in producing other well-known proteins, e.g. MAP kinases, p53, WTl, PTP phosphatases, SRC, and the like. Similar procedures, or modifications thereof, can be employed to prepare recombinant HDx polypeptides by microbial means or tissue-culture technology in accord with the subject invention.
  • the recombinant HDx genes can be produced by ligating nucleic acid encoding an phosphoses, p53, WTl, PTP phosphatases, SRC, and the like.
  • Similar procedures, or modifications thereof can be employed to prepare recombinant HDx polypeptides by microbial means or tissue-culture technology in accord with the subject invention.
  • the recombinant HDx genes can be produced by ligating nucleic acid en
  • HDx protein or a portion thereof, into a vector suitable for expression in either prokaryotic cells, eukaryotic cells, or both.
  • Expression vectors for production of recombinant forms of the subject HDx polypeptides include plasmids and other vectors.
  • suitable vectors for the expression of an HDx polypeptide include plasmids of die types: pBR322-derived plasmids, pEMBL-derived plasmids, pEX-derived plasmids, pBTac-derived plasmids and pUC-derived plasmids for expression in prokaryotic cells, such as E. coli.
  • YEP24, YIP5, YEP51, YEP52, ⁇ YES2, and YRP17 are cloning and expression vehicles useful in the introduction of genetic constructs into S. cerevisiae (see, for example, Broach et al. (1983) in Experimental Manipulation of Gene Expression, ed. M. Inouye Academic Press, p. 83, inco ⁇ orated by reference herein).
  • These vectors can replicate in E. coli due the presence of the pBR322 ori, and in S. cerevisiae due to the replication determinant of the yeast 2 micron plasmid.
  • an HDx polypeptide is produced recombinantly utilizing an expression vector generated by sub ⁇ cloning the coding sequence of one of the HDx genes represented in SEQ ID Nos: 1-4.
  • the preferred mammalian expression vectors contain both prokaryotic sequences, to facilitate the propagation of the vector in bacteria, and one or more eukaryotic transcription units that are expressed in eukaryotic cells.
  • the pcDNAI/amp, pcDNAI/neo, pRc/CMV, pSV2gpt, pSV2neo, pSV2-dhfr, pTk2, pRSVneo, pMSG, pSVT7, pko-neo and pHyg derived vectors are examples of mammalian expression vectors suitable for transfection of eukaryotic cells.
  • vectors are modified with sequences from bacterial plasmids, such as pBR322, to facilitate replication and drug resistance selection in both prokaryotic and eukaryotic cells.
  • derivatives of viruses such as the bovine papillomavirus (BPV-1), or Epstein-Barr virus (pHEBo, pREP-derived and p205) can be used for transient expression of proteins in eukaryotic cells.
  • BBV-1 bovine papillomavirus
  • pHEBo Epstein-Barr virus
  • the various methods employed in the preparation of the plasmids and transformation of host organisms are well known in the art.
  • suitable expression systems for both prokaryotic and eukaryotic cells, as well as general recombinant procedures see Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989) Chapters 16 and 17.
  • baculovirus expression systems include pVL-derived vectors (such as pVL1392, pVL1393 and pVL941), pAcUW-derived vectors (such as pAcUWl), and pBlueBac-derived vectors
  • MAP e.g., E. coli or CM89 or S. cerevisiae
  • in vitro by use of purified MAP e.g., procedure of Miller et al., supra.
  • the coding sequences for the polypeptide can be inco ⁇ orated as a part of a fusion gene including a nucleotide sequence encoding a different polypeptide.
  • This type of expression system can be useful under conditions where it is desirable to produce an immunogenic fragment of an HDx protein.
  • the VP6 capsid protein of rotavirus can be used as an immunologic carrier protein for portions of the HDx polypeptide, either in the monomeric form or in the form of a viral particle.
  • the nucleic acid sequences corresponding to the portion of a subject HDx protein to which antibodies are to be raised can be inco ⁇ orated into a fusion gene construct which includes coding sequences for a late vaccinia virus structural protein to produce a set of recombinant viruses expressing fusion proteins comprising HDx epitopes as part of the virion.
  • the Multiple Antigen Peptide system for peptide-based immunization can also be utilized to generate an immunogen, wherein a desired portion of an HDx polypeptide is obtained directly from organo-chemical synthesis of the peptide onto an oligomeric branching lysine core (see, for example, Posnett et al. (1988) JBC 263:1719 and Nardelli et al. (1992) J. Immunol. 148:914).
  • Antigenic determinants of HDx proteins can also be expressed and presented by bacterial cells.
  • HDx polypeptides can be generated as glutathione-S-transferase (GST-fusion) proteins.
  • GST-fusion proteins can enable easy purification of the HDx polypeptide, as for example by the use of glutathione-derivatized matrices (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. (N.Y.: John Wiley & Sons, 1991)).
  • a fusion gene coding for a purification leader sequence such as a poly-( ⁇ is)/enterokinase cleavage site sequence at the N-terminus of the desired portion of the recombinant protein, can allow purification of the expressed fusion protein by affinity chromatography using a Ni2+ metal resin.
  • the purification leader sequence can then be subsequently removed by treatment with enterokinase to provide the purified protein (e.g., see Hochuli et al. (1987) J. Chromatography 411:177; and Janknecht et al. PNAS 88:8972). Techniques for making fusion genes are known to those skilled in the art.
  • the joining of various DNA fragments coding for different polypeptide sequences is performed in accordance with conventional techniques, employing blunt- ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation.
  • the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers.
  • PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. John Wiley & Sons: 1992).
  • HDx polypeptides may also be chemically modified to create HDx derivatives by forming covalent or aggregate conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like.
  • Covalent derivatives of HDx proteins can be prepared by linking the chemical moieties to functional groups on amino acid sidechains of the protein or at the N-te ⁇ ninus or at the C-terminus of the polypeptide.
  • the present invention also makes available isolated HDx polypeptides which are isolated from, or otherwise substantially free of other cellular proteins, especially other signal transduction factors and/or transcription factors which may normally be associated with the HDx polypeptide.
  • the term "substantially free of other cellular proteins" (also referred to herein as "contaminating proteins") or “substantially pure or purified preparations” are defined as encompassing preparations of HDx polypeptides having less than 20% (by dry weight) contaminating protein, and preferably having less tiian 5% contaminating protein.
  • Functional forms of the subject polypeptides can be prepared, for the first time, as purified preparations by using a cloned gene as described herein.
  • purified it is meant, when referring to a peptide or DNA or RNA sequence, that the indicated molecule is present in the substantial absence of other biological macromolecules, such as other proteins.
  • the term “purified” as used herein preferably means at least 80% by dry weight, more preferably in the range of 95-99% by weight, and most preferably at least 99.8% by weight, of biological macromolecules of the same type present (but water, buffers, and other small molecules, especially molecules having a molecular weight of less than 5000, can be present).
  • pure as used herein preferably has the same numerical limits as “purified” immediately above.
  • isolated and purified do not encompass either natural materials in their native state or natural materials that have been separated into components (e.g., in an acrylamide gel) but not obtained either as pure (e.g. lacking contaminating proteins, or chromatography reagents such as denaturing agents and polymers, e.g. acrylamide or agarose) substances or solutions.
  • purified HDx preparations will lack any contaminating proteins from the same animal from that HDx is normally produced, as can be accomplished by recombinant expression of, for example, a human HDx protein in a non-human cell.
  • isolated HDx polypeptides can include all or a portion of an amino acid sequences corresponding to an HDx polypeptide represented in any one of SEQ ID Nos: 5-8 or homologous sequences thereto.
  • the HDx polypeptide includes both die v and ⁇ motifs, and preferably possess a histone deacetylase activity.
  • Isolated peptidyl portions of HDx proteins can be obtained by screening peptides recombinantly produced from the corresponding fragment of the nucleic acid encoding such peptides.
  • fragments can be chemically synthesized using techniques known in the art such as conventional Merrifield solid phase f-Moc or t-Boc chemistry.
  • an HDx polypeptide of the present invention may be arbitrarily divided into fragments of desired length with no overlap of the fragments, or preferably divided into overlapping fragments of a desired length. The fragments can be produced (recombinantly or by chemical synthesis) and tested to identify those peptidyl fragments which can function as either agonists or antagonists of a wild-type (e.g., "authentic") HDx protein.
  • the recombinant HDx polypeptides of the present invention also include homologs of the authentic HDx proteins, such as versions of those protein which are resistant to proteolytic cleavage, as for example, due to mutations which alter ubiquitination or other enzymatic targeting associated with the protein.
  • Modification of the structure of the subject HDx polypeptides can be for such pu ⁇ oses as enhancing therapeutic or prophylactic efficacy, stability (e.g., ex vivo shelf life and resistance to proteolytic degradation in vivo), or post-translational modifications (e.g., to alter phosphorylation pattern of protein).
  • Such modified peptides when designed to retain at least one activity of die naturally-occurring form of the protein, or to produce specific antagonists thereof, are considered functional equivalents of the HDx polypeptides described in more detail herein.
  • Such modified peptides can be produced, for instance, by amino acid substitution, deletion, or addition.
  • Whether a change in the amino acid sequence of a peptide results in a functional HDx homolog can be readily determined by assessing the ability of the variant peptide to produce a response in cells in a fashion similar to the wild-type protein, or competitively inhibit such a response.
  • Polypeptides in which more than one replacement has taken place can readily be tested in the same manner.
  • This invention further contemplates a method for generating sets of combinatorial mutants of the subject HDx proteins as well as truncation mutants, and is especially useful for identifying potential variant sequences (e.g. homologs) that are functional in modulating histone deacetylation.
  • the pu ⁇ ose of screening such combinatorial libraries is to generate, for example, novel HDx homologs which can act as either agonists or antagonist, or alternatively, possess novel activities all together.
  • HDx homologs can be engineered by the present method to provide selective, constitutive activation of enzymatic activity.
  • combinatorially-derived homologs can be generated to have an increased potency relative to a naturally occurring form of the protein.
  • HDx homologs can be generated by the present combinatorial approach to selectively inhibit (antagonize) histone deacetylation.
  • mutagenesis can provide HDx homologs which are able to bind other regulatory proteins or cytoskeletal elements (or DNA) yet prevent acetylation of histones, e.g. die homologs can be dominant negative mutants.
  • a dominant negative mutant of an HDx protein is mutated at one or more residues of its catalytic site and/or specificity subsites.
  • the amino acid sequences for a population of HDx homologs or other related proteins are aligned, preferably to promote the highest homology possible.
  • a population of variants can include, for example, HDx homologs from one or more species.
  • Amino acids which appear at each position of the aligned sequences are selected to create a degenerate set of combinatorial sequences.
  • the variegated library of HDx variants is generated by combinatorial mutagenesis at die nucleic acid level, and is encoded by a variegated gene library.
  • a mixture of synthetic oligonucleotides can be enzymatically ligated into gene sequences such that the degenerate set of potential HDx sequences are expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g. for phage display) containing the set of HDx sequences therein.
  • the amino acid sequences of interest can be aligned relative, to sequence homology.
  • the presence or absence of amino acids from an aligned sequence of a particular variant is relative to a chosen consensus length of a reference sequence, which can be real or artificial.
  • Figure 5B includes the alignment of the v and ⁇ -motifs for several of the ⁇ Dx gene products. Analysis of the alignment of these sequences from the ⁇ Dx clones can give rise to the generation of a degenerate library of polypeptides comprising potential ⁇ Dx sequences.
  • a library of variants based on the HDl sequence, but degenerate across each of the v and ⁇ -motifs can be provided.
  • On such library can be represented by the general formula A-(v motif)-B-( ⁇ motif)-C, wherein the v motif is an amino acid sequence represented in the general formula
  • ⁇ motif is an amino acid sequence represented in the general formula
  • Xi represents He or Val
  • X 2 represents Phe or Ser
  • X 3 represents Phe or Leu
  • X 4 represents Gly or Ala
  • X 5 represents Pro or Gin
  • Xg represents Gin or Glu
  • X 7 represents Leu or Thr
  • X represents Val or Tyr
  • X 9 represents Thr or Ser
  • X 10 represents Leu or He
  • X j 1 represents Met or Val
  • X 12 represents He or Val.
  • Xi represents Gly, Ala, Val, He or Leu
  • X 2 represents Phe, Tyr, Thr or Ser
  • X 3 represents Phe, Tyr, Gly, Ala, Val, He or Leu
  • X represents Gly, Ala, Val, He or Leu
  • X5 represents Pro, Asn or Gin
  • X represents .Asn, Gin, Asp or Glu
  • X 7 represents Gly, Ala, Val, He, Leu, Ser or Thr
  • X 8 represents Gly, Ala, Val, He, Leu, Phe or Tyr
  • X 9 represents Thr, Cys, or Ser
  • X 10 represents Gly, Ala, Val, He or Leu
  • X11 represents Met, Cys, Gly, Ala, Val, He, Leu, Ser or Thr
  • X 12 represents Gly, Ala, Val, He or Leu.
  • each degenerate position can be any one of the naturally occurring amino acids.
  • a library of coding sequence fragments can be provided for an HDx clone in order to generate a variegated population of HDx fragments for screening and subsequent selection of bioactive fragments.
  • a variety of techniques are known in the art for generating such libraries, including chemical synthesis.
  • a library of coding sequence fragments can be generated by (i) treating a double stranded PCR fragment of an HDx coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule; (ii) denaturing d e double stranded DNA; (iii) renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products; (iv) removing single stranded portions from reformed duplexes by treatment with S 1 nuclease; and (v) ligating the resulting fragment library into an expression vector.
  • an expression library can be derived which codes for N-terminal, C-terminal and internal fragments of various sizes.
  • a wide range of techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a certain property. Such techniques will be generally adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of HDx homologs.
  • the most widely used techniques for screening large gene libraries typically comprises cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates relatively easy isolation of the vector encoding the gene whose product was detected.
  • the library of HDx variants is expressed as a fusion protein on the surface of a viral particle.
  • foreign peptide sequences can be expressed on the surface of infectious phage, thereby conferring two significant benefits.
  • coli filamentous phages M13, fd., and fi are most often used in phage display libraries, as either of the phage gill or gVIII coat proteins can be used to generate fusion proteins widiout disrupting the ultimate packaging of the viral particle (Ladner et al. PCT publication WO 90/02909; Garrard et al., PCT publication WO 92/09690; Marks et al.
  • the recombinant phage antibody system (RPAS, Pharmacia Catalog number 27-9400-01) can be easily modified for use in expressing and screening HDx combinatorial libraries by panning on glutadiione unmobilized histones/GST fusion proteins or RbAp48/GST fusion protein to enrich for HDx homologs which retain an ability to bind a substrate or regulatory protein.
  • Each of these HDx homologs can subsequently be screened for further biological activities in order to differentiate agonists and antagonists.
  • histone-binding homologs isolated from the combinatorial library can be tested for their enzymatic activity directly, or for their effect on cellular proliferation relative to the wild-type form of the protein.
  • the invention also provides for reduction of the HDx or RbAp48 or histones proteins to generate mimetics, e.g. peptide or non-peptide agents, which are able to disrupt a biological activity of an HDx polypeptide of the present invention, e.g. as catalytic inhibitor or an inhibitor of protein-protein interactions.
  • mimetics e.g. peptide or non-peptide agents
  • Such mutagenic techniques as described above are also useful to map the determinants of the HDx proteins which participate in protein-protein or protein-DNA interactions involved in, for example, interaction of the subject HDx polypeptide with histones, RbAp48 or cytoskeletal elements.
  • the critical residues of a subject HDx polypeptide which are involved in molecular recognition of histones can be determined and used to generate HDx-derived peptidomimetics which competitively inhibit binding of the authentic HDx protein with that moiety.
  • residues of a histone or of RbAp48 involved in binding to HDx proteins can be identified, and peptides or peptidomimetics based on such residues can also be used as competitive inhibitors of die interaction of an HDx protein with either of those proteins.
  • peptidomimetic compounds can be generated which mimic those residues which facilitate the interaction.
  • non-hydrolyzable peptide analogs of such residues can be generated using benzodiazepine (e.g., see Freidinger et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), azepine (e.g., see Huffman et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), substituted gamma lactam rings (Garvey et al. in Peptides: Chemistry and Biology, G.R.
  • Another aspect of the invention pertains to an antibody specifically reactive with an HDx protein.
  • immunogens derived from an HDx protein e.g. based on the cDNA sequences
  • anti-protein/anti-peptide antisera or monoclonal antibodies can be made by standard protocols (See, for example, Antibodies: A Laboratory Manual ed. by Harlow and Lane (Cold Spring Harbor Press: 1988)).
  • a mammal, such as a mouse, a hamster or rabbit can be immunized with an immunogenic form of the peptide (e.g., an HDx polypeptide or an antigenic fragment which is capable of eliciting an antibody response).
  • an immunogenic portion of an HDx protein can be administered in d e presence of adjuvant.
  • the progress of immunization can be monitored by detection of antibody titers in plasma or serum.
  • Standard ELISA or other immunoassays can be used with the immunogen as antigen to assess the levels of antibodies.
  • the subject antibodies are immunospecific for antigenic determinants of an HDx protein of a organism, such as a mammal, e.g. antigenic determinants of a protein represented by one of SEQ DD Nos: 5-8 or closely related homologs (e.g.
  • the anti-HDx polypeptide antibodies do not substantially cross react (i.e. does not react specifically) with a protein which is, for example, less than 85%, 90% or 95% homologous with the selected HDx.
  • the antibody has a binding affinity for a non-homologous protein which is at least one order of magnitude, more preferably at least 2 orders of magnitude, and even more preferably at least 3 orders of magnitude less than the binding affinity of the antibody for the intended target HDx.
  • anti-HDx antisera can be obtained and, if desired, polyclonal anti-HDx antibodies isolated from the serum.
  • antibody-producing cells lymphocytes
  • myeloma cells can be harvested from an immunized animal and fused by standard somatic cell fusion procedures with immortalizing cells such as myeloma cells to yield hybridoma cells.
  • Hybridoma cells can be screened immunochemically for production of antibodies specifically reactive with an HDx polypeptide of the present invention and monoclonal antibodies isolated from a culture comprising such hybridoma cells.
  • antibody as used herein is intended to include fragments thereof which are also specifically reactive with one of the subject HDx polypeptides.
  • Antibodies can be fragmented using conventional techniques and d e fragments screened for utility in the same manner as described above for whole antibodies.
  • F(ab) fragments can be generated by treating antibody with pepsin.
  • the resulting F(ab) 2 fragment can be treated to reduce disulfide bridges to produce Fab fragments.
  • the antibody of the present invention is further intended to include bispecific and chimeric molecules having affinity for an HDx protein conferred by at least one CDR region of the antibody.
  • Both monoclonal and polyclonal antibodies (Ab) directed against authentic HDx polypeptides, or HDx variants, and antibody fragments such as Fab, F(ab) 2 , Fv and scFv can be used to block the action of one or more HDx proteins and allow the study of the role of these proteins in, for example, differentiation of tissue. Experiments of this nature can aid in deciphering the role of HDx proteins that may be involved in control of proliferation versus differentiation, e.g., in patterning and tissue formation.
  • Antibodies which specifically bind HDx epitopes can also be used in immunohistochemical staining of tissue samples in order to evaluate the abundance and pattern of expression of each of the subject HDx polypeptides.
  • Anti-HDx antibodies can be used diagnostically in immuno-precipitation and immuno-blotting to detect and evaluate HDx protein levels in tissue as part of a clinical testing procedure. For instance, such measurements can be useful in predictive valuations of the onset or progression of proliferative or differentiative disorders.
  • the ability to monitor HDx protein levels in an individual can allow determination of the efficacy of a given treatment regimen for an individual afflicted with such a disorder.
  • the level of HDx polypeptides may be measured from cells in bodily fluid, such as in samples of cerebral spinal fluid or amniotic fluid, or can be measured in tissue, such as produced by biopsy.
  • Diagnostic assays using anti-HDx antibodies can include, for example, immunoassays designed to aid in early diagnosis of a disorder, particularly ones which are manifest at birth. Diagnostic assays using anti-HDx polypeptide antibodies can also include immunoassays designed to aid in early diagnosis and phenotyping neoplastic or hype ⁇ lastic disorders.
  • anti-HDx antibodies of the present invention is in the immunological screening of cDNA libraries constructed in expression vectors such as ⁇ gtl l, ⁇ gtl8-23, ⁇ ZAP, and ⁇ ORF8.
  • Messenger libraries of this type having coding sequences inserted in the correct reading frame and orientation, can produce fusion proteins.
  • ⁇ gtl l will produce fusion proteins whose amino termini consist of ⁇ -galactosidase amino acid sequences and whose carboxy termini consist of a foreign polypeptide.
  • Antigenic epitopes of an HDx protein e.g.
  • HDx homologs can be detected and cloned from other animals, as can alternate isoforms (including splicing variants) from humans.
  • nucleotide sequences determined from the cloning of HDx genes from organisms will further allow for the generation of probes and primers designed for use in identifying and/or cloning HDx homologs in other cell types, e.g.
  • the present invention also provides a probe/primer comprising a substantially purified oligonucleotide, which oligonucleotide comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least 10 consecutive nucleotides of sense or anti-sense sequence selected from the group consisting of SEQ DD Nos: 1-4 or naturally occurring mutants thereof
  • primers based on the nucleic acid represented in SEQ ID Nos: 1-4 can be used in PCR reactions to clone HDx homologs.
  • probes based on the subject HDx sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins.
  • the probe further comprises a label group attached thereto and able to be detected, e.g. the label group is selected from amongst radioisotopes, fluorescent compounds, enzymes, and enzyme co-factors.
  • the label group is selected from amongst radioisotopes, fluorescent compounds, enzymes, and enzyme co-factors.
  • Such probes can also be used as a part of a diagnostic test kit for identifying cells or tissue which misexpress an HDx protein, such as by measuring a level of an HDx- encoding nucleic acid in a sample of cells from a patient; e.g. detecting HDx mRNA levels or determining whether a genomic HDx gene has been mutated or deleted.
  • nucleotide probes can be generated from the subject HDx genes which facilitate histological screening of intact tissue and tissue samples for the presence (or absence) of HDx-encoding transcripts. Similar to the diagnostic uses of anti-HDx antibodies, the use of probes directed to HDx messages, or to genomic HDx sequences, can be used for both predictive and therapeutic evaluation of allelic mutations which might be manifest in, for example, neoplastic or hype ⁇ lastic disorders (e.g. unwanted cell growth) or abnormal differentiation of tissue. Used in conjunction with immunoassays as described above, the oligonucleotide probes can help facilitate the determination of the molecular basis for a developmental disorder which may involve some abnormality associated with expression (or lack thereof) of an HDx protein. For instance, variation in polypeptide synthesis can be differentiated from a mutation in a coding sequence.
  • the present method provides a method for determining if a subject is at risk for a disorder characterized by aberrant cell proliferation and or differentiation.
  • method can be generally characterized as comprising detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion characterized by at least one of (i) an alteration affecting the integrity of a gene encoding an HDx-protein, or (ii) the mis-expression of the HDx gene.
  • such genetic lesions can be detected by ascertaining the existence of at least one of (i) a deletion of one or more nucleotides from an HDx gene, (ii) an addition of one or more nucleotides to an HDx gene, (iii) a substitution of one or more nucleotides of an HDx gene, (iv) a gross chromosomal rearrangement of an HDx gene, (v) a gross alteration in the level of a messenger RNA transcript of an HDx gene, (vii) aberrant modification of an HDx gene, such as of the methylation pattern of the genomic DNA, (vii) the presence of a non-wild type splicing pattern of a messenger RNA transcript of an HDx gene, (viii) a non-wild type level of an HDx-protein, and (ix) inappropriate post-translational modification of an HDx-protein.
  • tiiere is provided a nucleic acid composition comprising a (purified) oligonucleotide probe including a region of nucleotide sequence which is capable of hybridizing to a sense or antisense sequence of an HDx gene, such as represented by any of SEQ ID Nos: 1-4, or naturally occurring mutants thereof, or 5' or 3' flanking sequences or intronic sequences naturally associated with the subject HDx genes or naturally occurring mutants thereof.
  • the nucleic acid of a cell is rendered accessible for hybridization, the probe is exposed to nucleic acid of the sample, and the hybridization of the probe to the sample nucleic acid is detected.
  • detection of the lesion comprises utilizing the probe/primer in a polymerase chain reaction (PCR) (see, e.g. U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al.
  • PCR polymerase chain reaction
  • LCR ligation chain reaction
  • the method includes the steps of (i) collecting a sample of cells from a patient, (ii) isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, (iii) contacting the nucleic acid sample with one or more primers which specifically hybridize to an HDx gene under conditions such that hybridization and amplification of the HDx gene (if present) occurs, and (iv) detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample.
  • nucleic acid e.g., genomic, mRNA or both
  • the level of an HDx-protein can be detected by immunoassay.
  • the cells of a biopsy sample can be lysed, and the level of an HDx-protein present in the cell can be quantitated by standard immunoassay techniques.
  • aberrant methylation patterns of an HDx gene can be detected by digesting genomic DNA from a patient sample with one or more restriction endonucleases that are sensitive to methylation and for which recognition sites exist in the HDx gene (including in the flanking and intronic sequences). See, for example, Buiting et al. (1994) Human Mol Genet 3:893-895.
  • Digested DNA is separated by gel electrophoresis, and hybridized with probes derived from, for example, genomic or cDNA sequences.
  • the methylation status of the HDx gene can be determined by comparison of the restriction pattern generated from the sample DNA with that for a standard of known methylation.
  • the subject HDx polypeptides can be used to generate a "two hybrid” assay or an "interaction trap” assay (see, for example, U.S. Patent No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J Biol Chem 268:12046-12054; Bartel et al.
  • HDx-binding proteins for isolating coding sequences for other cellular proteins which bind HDxs.
  • HDx-binding proteins or "HDx-bp"
  • Such HDx-binding proteins would likely be involved in the regulation of HDx, e.g., as regulatory subunits or transducers, or be substrates which are regulated by an HDx.
  • the interaction trap relies on reconstituting in vivo a functional transcriptional activator protein from two separate fusion proteins.
  • the method makes use of chimeric genes which express hybrid proteins.
  • a first hybrid gene comprises die coding sequence for a DNA-binding domain of a transcriptional activator fused in frame to the coding sequence for an HDx polypeptide.
  • the second hybrid protein encodes a transcriptional activation domain fused in frame to a sample gene from a cDNA library. If die bait and sample hybrid proteins are able to interact, e.g., form an HDx-dependent complex, they bring into close proximity the two domains of the transcriptional activator.
  • the present invention facilitates the development of assays which can be used to screen for drugs, including HDx homologs, which are either agonists or antagonists of the normal cellular function of the subject HDx polypeptides, or of their role in the pathogenesis of cellular differentiation and/or proliferation and disorders related thereto.
  • the present invention further provides drug screening assays for detecting agents which modulate the bioactivity of HDx-related proteins.
  • agents when directed to, for example, fungal HDx-related proteins, can be used in the treatment of various infections.
  • the assay evaluates the ability of a compound to modulate binding between an HDx polypeptide and a molecule, be it protein or DNA, that interacts with the HDx polypeptide. It will be apparent from the following description of exemplary assays that, in place of a human (or other mammalian) HDx protein, the assay can be derived with an HDx-related protein such as RPD3.
  • HDx-binding proteins can be used, e.g., other human proteins.
  • exemplary compounds which can be screened include peptides, nucleic acids, carbohydrates, small organic molecules, and natural product extract libraries, such as isolated from animals, plants, fungus and/or microbes.
  • any of the novel interactions described herein could be exploited in a drug screening assay.
  • the interaction between an HDx protein and RbAp48 can be detected in the presence and the absence of a test compound.
  • the ability of a compound to modulate the binding of an HDx protein, or HDx-related protein such as die yeast RPD3, with histones can be assessed.
  • the identification of a test compound which influences, for example, ⁇ D1 catalyzed deacetylation of histones would be useful in the modulation of ⁇ D1 activity in mammalian cells, while the identification of a test compound which selectively inhibits the yeast RPD3 deacetylase activity would be useful as an antifungal agent.
  • test compound in other embodiments the effect of a test compound on the binding of an HDx protein to other molecules, such as cytoskeletal components, or other proteins identified by the HDx-dependent ITS set out above, could be tested.
  • assay formats will suffice and, in light of the present inventions, will be comprehended by a skilled artisan.
  • assays which employ the subject mammalian HDx proteins can be used to identify compounds that have therapeutic indexes more favorable than sodium butyrate, trapoxin, trichostatin or the like.
  • trapoxin-like drugs can be identified by the present invention which have enhanced tissue-type or cell-type specificity relative to trapoxin.
  • the subject assays can be used to generate compounds which preferentially inhibit IL-2 mediated proliferation activation of lymphocytes, or inhibit proliferation of certain tumor cells, without substantially interfering with other tissues, e.g. hepatocytes.
  • similar assays can be used to identify drugs which inhibit proliferation of yeast cells or other lower eukaryotes, but which have a substantially reduced effect on mammalian cells, thereby improving therapeutic index of the drug as an anti-mycotic agent.
  • the identification of such compounds is made possible by the use of differential screening assays which detect and compare drug-mediated inhibition of deacetylase activity between two or more different HDx-like enzymes, or compare drug- mediated inhibition of formation of complexes involving two or more different types of HDx-like proteins.
  • the assay can be designed for side-by-side comparison of the effect of a test compound on the deacetylase activity or protein interactions of tissue-type specific HDx proteins. Given the apparent diversity of HDx proteins, it is probable that different functional HDx activities, or HDx complexes exist and, in certain mstances, are localized to particular tissue or cell types.
  • test compounds can be screened for agents able to inhibit die tissue-specific formation of only a subset of the possible repertoire of HDx/regulatory protein complexes, or which preferentially inhibit certain HDx enzymes.
  • an interaction trap assay can be derived using two or more different human HDx "bait" proteins, while the "fish" protein is constant in each, e.g. a human RbAp48 construct. Running the interaction trap side- by-side permits the detection of agents which have a greater effect (e.g. statistically significant) on the formation of one of the HDx/RbAp48 complexes than on the formation of the other HDx complexes.
  • differential screening assays can be used to exploit the difference in protein interactions and/or catalytic mechanism of mammalian HDx proteins and yeast RPD3 proteins in order to identify agents which display a statistically significant increase in specificity for inhibiting the yeast enzyme relative to the mammalian enzyme.
  • lead compounds which act specifically on pathogens such as fungus involved in mycotic infections, can be developed.
  • the present assays can be used to screen for agents which may ultimately be useful for inhibiting at least one fungus implicated in such mycosis as candidiasis, aspergillosis, mucormycosis, blastomycosis, geotrichosis, cryptococcosis, chromoblastomycosis, coccidioidomycosis, conidiosporosis, histoplasmosis, maduromycosis, rhinosporidosis, nocaidiosis, para-actinomycosis, penicilliosis, monoliasis, or sporotrichosis.
  • mycosis as candidiasis, aspergillosis, mucormycosis, blastomycosis, geotrichosis, cryptococcosis, chromoblastomycosis, coccidioidomycosis, conidiosporosis, histoplasmosis, maduromycosis, rhinosporidosis,
  • the present assay can comprise comparing the relative effectiveness of a test compound on inhibiting the deacetylase activity of a mammalian HDx protein with its effectiveness towards inhibiting the deacetylase activity of an RPD3 homolog cloned from yeast selected from the group consisting of Candida albicans, Candida stellatoidea, Candida tropicalis, Can ⁇ da parapsilosis, Candida krusei, Candida pseudotropicalis, Candida quillermondii, or Candida rugosa.
  • the present assay can be used to identify anti-fungal agents which may have therapeutic value in the treatment of aspergillosis by selectively targeting RPD3 homologs cloned from yeast such as Aspergillus fumigatus, Aspergillus flavus, Aspergillus niger, Aspergillus nidulans, or Aspergillus terreus.
  • yeast such as Aspergillus fumigatus, Aspergillus flavus, Aspergillus niger, Aspergillus nidulans, or Aspergillus terreus.
  • the mycotic infection is mucormycosis
  • the RPD3 deacetylase can be derived from yeast such as Rhizopus arrhizus, Rhizopus oryzae, Absidia corymbifera, Absidia ramosa, or Mucor pusillus.
  • Sources of other RPD3 activities for comparison with a mammalian HDx activity includes the pathogen Pneum
  • anti-fungal agents developed with such differential screening assays can be used, for example, as preservatives in foodstuff, feed supplement for promoting weight gain in livestock, or in disinfectant formulations for treatment of non-living matter, e.g., for decontaminating hospital equipment and rooms.
  • the present invention expressly contemplates the use and formulations of the subject HDx therapeutics in insecticides, such as for use in management of insects like the fruit fly.
  • certain of the subject HDx inhibitors can be selected on the basis of inhibitory specificity for plant HDx-related activities relative to the mammalian enzyme.
  • a plant HDx-related protein can be disposed in a differential screen with one or more of the human enzymes to select those compounds of greatest selectivity for inhibiting the plant enzyme.
  • the present invention specifically contemplates formulations of the subject HDx inhibitors for agricultural applications, such as in the form of a defoliant or the like. In many drug screening programs which test libraries of compounds and natural extracts, high throughput assays are desirable in order to maximize the number of compounds surveyed in a given period of time.
  • Assays which are performed in cell-free systems such as may be derived with purified or semi-purified proteins, are often preferred as "primary" screens in that they can be generated to permit rapid development and relatively easy detection of an alteration in a molecular target which is mediated by a test compound.
  • the effects of cellular toxicity and/or bioavailability of the test compound can be generally ignored in the in vitro system, the assay instead being focused primarily on the effect of the drug on the molecular target as may be manifest in an alteration of binding affinity with upstream or downstream elements.
  • a reaction mixture is generated to include an HDx polypeptide, compound(s) of interest, and a "target polypeptide", e.g., a protein, which interacts with the HDx polypeptide, whether as a substrate or by some other protein-protein interaction.
  • target polypeptides include histones, RbAp48 polypeptides, Sin3 polypeptides, and or combinantions thereof or with other transciptional regulatory proteins (such as myc, max, etc, see Example 3)).
  • Detection and quantification of complexes containing the HDx protein provide a means for deterrnining a compound's efficacy at inhibiting (or potentiating) complex formation between the HDx and the target polypeptide.
  • the efficacy of the compound can be assessed by generating dose response curves from data obtained using various concentrations of the test compound.
  • a control assay can also be performed to provide a baseline for comparison. In the control assay, isolated and purified HDx polypeptide is added to a composition containing the target polypeptide and the formation of a complex is quantitated in the absence of the test compound.
  • Complex formation between the HDx polypeptide and the target polypeptide may be detected by a variety of techniques. Modulation of the formation of complexes can be quantitated using, for example, detectably labeled proteins such as radiolabeled, fluorescently labeled, or enzymatically labeled HDx polypeptides, by immunoassay, by chromatographic detection, or by detecting the intrinsic activity of the acetylase.
  • detectably labeled proteins such as radiolabeled, fluorescently labeled, or enzymatically labeled HDx polypeptides
  • immunoassay by immunoassay
  • chromatographic detection or by detecting the intrinsic activity of the acetylase.
  • a fusion protein can be provided which adds a domain that allows the protein to be bound to a matrix.
  • glutathione-S-transferase/HDx (GST/HDx) fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St.
  • the beads are washed to remove any unbound label, and the matrix immobilized and radiolabel determined directly (e.g. beads placed in scintillant), or in the supernatant after the complexes are subsequently dissociated.
  • the complexes can be dissociated from the matrix, separated by SDS-PAGE, and the level of HDx-binding protein found in the bead fraction quantitated from the gel using standard electrophoretic techniques such as described in the appended examples.
  • HDx or target polypeptide can be immobilized utilizing conjugation of biotin and streptavidin.
  • biotinylated HDx molecules can be prepared from biotin-N ⁇ S (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • antibodies reactive with HDx can be derivatized to the wells of the plate, and HDx trapped in the wells by antibody conjugation.
  • preparations of an target polypeptide and a test compound are incubated in the HDx-presenting wells of the plate, and the amount of complex trapped in the well can be quantitated.
  • Exemplary methods for detecting such complexes include immunodetection of complexes using antibodies reactive with the target polypeptide, or which are reactive with HDx protein and compete with the target polypeptide; as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the target polypeptide, either intrinsic or extrinsic activity.
  • the enzyme can be chemically conjugated or provided as a fusion protein with the target polypeptide.
  • the target polypeptide can be chemically cross-linked or genetically fused with horseradish peroxidase, and the amount of polypeptide trapped in the complex can be assessed with a chromogenic substrate of the enzyme, e.g. 3,3'-diamino-benzadine terahydrochloride or 4-chloro-l-napthol.
  • a fusion protein comprising the polypeptide and glutathione-S-transferase can be provided, and complex formation quantitated by detecting the GST activity using 1- chloro-2,4-dinitrobenzene ( ⁇ abig et al (1974) J Biol Chem 249:7130).
  • the protein to be detected in the complex can be "epitope tagged" in the form of a fusion protein which includes, in addition to the HDx sequence, a second polypeptide for which antibodies are readily available (e.g. from commercial sources).
  • the GST fusion proteins described above can also be used for quantification of binding using antibodies against the GST moiety.
  • Other useful epitope tags include myc-epitopes (e.g., see Ellison et al.
  • a two hybrid assay can be generated with an HDx and HDx-binding protein. Drug dependent inhibition or potentiation of the interaction can be scored.
  • the HDx proteins themselves, or in complexes with other proteins are capable of binding DNA and modifying transcription of a gene
  • a transcriptional based assay using, for example, an transcriptional regulatory sequences responsive to HDx complexes operably linked to a detectable marker gene. For illustration, see Example 3.
  • each of the assay systems set out above can be generated in a
  • the assay format can provide information regarding specificity as well as potency. For instance, side-by-side comparison of a test compound's effect on different HDxs can provide information on selectivity, and permit the identification of compounds which selectively modulate the bioactivity of only a subset of the HDx family.
  • inhibitors of the enzymatic activity of each of the subject HDx proteins can be identified using assays derived from measuring the ability of an agent to inhibit catalytic conversion of a substrate by the subject proteins.
  • the ability of the subject HDx proteins to deacetylate a histone substrate, such as histone ⁇ 4 (see examples), in the presence and absence of a candidate inhibitor can be determined using standard enzymatic assays.
  • the assay will employ a labeled acetyl group linked to appropriate histone lysine residues as substrates.
  • a histone substrate peptide can be labeled with a group whose signal is dependent on the simultaneous presence or absence of an acetyl group, e.g., the label can be a fluorogenic group whose fluorescence is modulated (either quenched or potentiated) by the presence of the acetyl moiety.
  • the ability of a test agent to cause a statistically significant change in substrate conversion by a histone deacetylase can be measured, and as desirable, inhibition constants, e.g., Kj values, can be calculated.
  • the histone substrate can be provided as a purified or semi-purified polypeptide or as part of a cell lysate.
  • the histone deacetylase can be provided to the reaction mixture as a purified or semi-purified polypeptide or as a cell lysate.
  • the reaction mixtures of the subject method can range from reconstituted protein mixtures derived with purified preparations of histones and deacetylases, to rnixtures of cell lysates, e.g., by admixing baculovirus lysates containing recombinant histones and deacetylases.
  • the histone substrate for the subject assay is provided by isolation of radiolabeled histones from metabolically labelled cells.
  • HeLa cells can be labelled in culture by addition of [ 3 H] acetate (New England Nuclear) to the culture media.
  • Radiolabelled histones can be isolated from the cells by extraction with HsSO 4 (Marushige et al. (1966) J Mol Biol 15:160-174). Briefly, cells are homogenized in buffer, centrifuged to isolate a nuclear pellet, the subsequently homogenized nuclear pellet centrifuged through sucrose, and die resulting chromatin pellet extracted by addition of H-SO 4 to yield [ 3 H]acetyl-labelled histones.
  • nucleosome preparations containing [ 3 H]acetyl-labelled histones can be isolated from the labelled cells.
  • nucleosomes can be isolated from cell preparations by sucrose gradient centrifugation (Hay et al. (1983) J Biol Chem 258:3726-3734; and Noll (1967) Nature 215:360-363), and polynucleosomes can be prepared by NaCl precipitation from micrococcal nuclease digested cells (Hay et al., supra). Similar procedures for isolating labelled histones from odier cells types, including yeast, have been described. See, for example, Alonso et al. (1986) Biochem Biophys Acta 866:161-169; and Kreiger et al. (1974) JBiol Chem 249:332-334.
  • the histone is generated by recombinant gene expression, and includes an exogenous tag (e.g., an HA epitope, a poly(his) sequence or the like) which facilitates in purification from cell extracts.
  • an exogenous tag e.g., an HA epitope, a poly(his) sequence or the like
  • whole nuclei can be isolated from metabolically labelled cells by micrococcal nuclease digestion (Hay et al., supra)
  • the deacetylase substrate can be provided as an acetylated peptide including a sequence corresponding to the sequence about the specific lysyl residues acetylated on histone, e.g., a peptidyl portions of the core histones H A, H2B, H3 or H4.
  • Such fragments can be produced by cleavage of acetylated histones derived from metabolically labelled cells, e.g., such as by treatment with proteolytic enzymes or cyanogen bromide (Kreiger et al., supra).
  • the acetylated peptide can be provided by standard solid phase synthesis using acetylated lysine residues (Kreiger et al., supra).
  • the activity of a histone deacetylase in the subject assays is detected by measuring release of [ 3 H]acetate by standard scintillant techniques.
  • a reaction mixture is provided which comprises a recombinant HDx protein suspended in buffer, along with a sample of [ 3 H]acetyl-labelled histones and (optionally) a test compound.
  • the reaction mixture is maintained at a desired temperature and pH, such as 22°C at pH7.8, for several hours, and the reaction terminated by boiling or other form of denaturation.
  • Released [ 3 H]acetate is extracted and counted.
  • the quenched reaction mixture can be acidified with concentrated HCl, and used to create a biphasic mixture with ethyl acetate.
  • the resulting 2 phase system is thoroughly mixed, centrifuged, and the ethyl acetate phase collected and counted by standard scintillation methods. Other methods for detecting acetate release will be easily recognized by those skilled in the art.
  • the drug screening assay is derived to include a whole cell recombinantly expressing one or more of a target protein or HDx protein.
  • the ability of a test agent to alter the activity of the HDx protein can be detected by analysis of the recombinant cell.
  • agonists and antagonists of the HDx biological activity can by detected by scoring for alterations in growth or differentiation (phenotype) of the cell.
  • General techniques for detecting each are well known, and will vary with respect to the source of the particular reagent cell utilized in any given assay. For example, quantification of proliferation of cells in the presence and absence of a candidate agent can be measured with a number of techniques well known in the art, including simple measurement of population growth curves.
  • turbidimetric techniques i.e. absorbence/ transmittance of light of a given wavelength through the sample
  • the reagent cell is a yeast cell
  • measurement of absorbence of light at a wavelength between 540 and 600nm can provide a conveniently fast measure of cell growth.
  • ability to form colonies in solid medium e.g. agar
  • an HDx substrate protein such as a histone
  • growth in solid media such as agar can further aid in establishing whether a mammalian cell is transformed.
  • visual inspection of the mo ⁇ hology of the reagent cell can be used to determine whether the biological activity of the targeted HDx protein has been affected by the added agent.
  • the ability of an agent to influence an apoptotic phenotype which is mediated in some way by a recombinant HDx protein can be assessed by visual microscopy.
  • the formation of certain cellular structures as part of differentiation such as the formation of neuritic process, can be visualized under a light microscope.
  • test agent on reagent cell can be assessed by measuring levels of expression of specific genes, e.g., by reverse transcription-PCR.
  • Another method of scoring for effect on Hdx activity is by detecting cell-type specific marker expression through immunofluorescent staining.
  • markers are known in the art, and antibodies are readily available.
  • the presence of chondroitin sulphate proteoglycans as well as type-II collagen are correlated with cartilage production in chondrocytes, and each can be detected by immunostaining.
  • the human kidney differentiation antigen gpl60, human aminopeptidase A is a marker of kidney induction
  • the cytoskeletal protein troponin I is a marker of heart induction.
  • the alteration of expression of a reporter gene construct provided in the reagent cell provides a means of detecting the effect on HDx activity.
  • reporter gene constructs derived using the transcriptional regulatory sequences, e.g. the promoters, for developmentally regulated genes can be used to drive the expression of a detectable marker, such as a luciferase gene.
  • the construct is derived using the promoter sequence from a gene expressed in a particular differentiative phenotype.
  • the recombinant HDx cells of the present assay can be generated so as to comprise heterologous HDx proteins (i.e. cross-species expression).
  • heterologous HDx proteins i.e. cross-species expression
  • HDx proteins from one species can be expressed in the cells of another under conditions wherein the heterologous protein is able to rescue loss-of-function mutations in the host cell.
  • the reagent cell can be a yeast cell in which a human MDx protein (e.g. exogenously expressed) is the intended target for development of an anti-proliferative agent.
  • the M778 strain MATa ura3-52 trpl ⁇ l his3-200 leu2-l trkl ⁇ rpd3 ⁇ ::HIS3, described by Vidal et al. (1991) Mol Cell Biol 6317-6327, which lacks a functional endogenous RPD3 gene can be transfected with an expression plasmid including a mammalian HDx gene in order to complement the RPD3 loss-of-function.
  • the coding sequence for HDl can be cloned into a pRS integrative plasmid containing a selectable marker (Sikorski et al. (1989) Genetics 122:19-27), and resulting construct used to transform the M778 strain.
  • the resulting cells should produce a mammalian HDl protein which may be capable performing at least some of the functions of the yeast RPD3 protein.
  • the HDx transformed yeast cells can be easier to manipulate than mammalian cells, and can provide access to certain assay formats, such as turbidity detection methods, which may not be obtainable with mammalian cells.
  • the combination of the "mammalianized" strain with the strain M537 can provide an extraordinarly sensitive cell-based assay for detecting agent which specifically inhibit, for example, the yeast RPD3 deacetylase.
  • an HDx inhibitor compound of the invention can be represented by the formula A-B-C, in which A is a specificity element for selective binding to an HDx, B is a linker element, and C is an electrophilic moiety capable of reacting with a nucleophilic moiety of an HDx; with the proviso that the compound is not butyrate, trapoxin, or trichostatin.
  • the invention provides an affinity matrix for binding or purifying an HDx.
  • the affinity matrix can be represented by the formula S-A-B-C, in which S is a solid or insoluble support, and A, B, and C are as described above.
  • the solid or insoluble support S can be any of a variety of supports, many of which are known in the art, for synthesis of, or immobilization of, compounds, e.g., peptides, benzodiazepines, and the like.
  • solid-supported synthesis see, e.g., ⁇ odge et ⁇ l., Polymer-supported Reactions in Organic Synthesis, John Wiley & Sons, New York, 1980.
  • the HDx inhibitor moiety A-B-C can be bonded directly to the support S, or can be bonded to the support S through a linking or spacing moiety, as is known in the art.
  • the invention provides a method of inhibiting an HDx.
  • the method comprises contacting the HDx with a compound capable of inhibiting HDx activity, under conditions such that HDx activity is inhibited.
  • the compounds can be represented by the formula A-B-C, in which A, B, and C are as described above; with the proviso that the compound is not butyrate, trapoxin, or trichostatin.
  • the invention provides a method of purifying an HDx.
  • the method includes contacting a reaction mixture comprising an HDx with an affinity matrix capable of selectively binding to an HDx, and separating at least one odier component of the reaction mixture from the HDx.
  • the affinity matrix can be represented by the formula S-A-B-C, in which S, A, B, and C are as described above.
  • the elements A, B, and C of the inhibitor compounds are selected to permit selective binding to, and inhibition of, at least one HDx.
  • the elements A, B, and C can be selected to provide specificity for particular HDxs.
  • a series of candidate HDx inhibitor compounds can be synthesized, e.g., according to the combinatorial methods described infra, and the library of candidate compounds screened against one or more HDxs to determine the compound or compounds with optimal activity and specificity for a particular HDx.
  • the specificity element A is selected such that the HDx inhibitor compound binds selectively to an HDx.
  • the specificity element A will be selected according to factors such as the binding specificity of the HDx or HDxs to which the inhibitor compound should bind, ease of synthesis, stability in vivo or in vitro, and the like.
  • the specificity element A is a cyclotetrapeptidyl moiety.
  • A is a substituted or unsubstituted aryl moiety.
  • A is a nonaromatic carbocycle.
  • A is an amino acyl moiety (e.g., a natural or non-natural amino acyl moiety).
  • A is a heterocyclyl moiety.
  • B is selected from the group consisting of substituted and unsubstituted C -C alkylidene, C -C alkenylidene, C 4 -C alkynylidene, and D-E-F, in which D and F are independently absent or C -C 7 alkylidene, C -C 7 alkenylidene, or C2-C7 alkynylidene, and E is O, S, or NR', in which R' is ⁇ , lower alkyl, lower alkenyl, lower alkynyl, aralkyl, aryl, or heterocyclyl.
  • the element B should be selected to permit the specificity element A to interact with an HDx such that specific binding occurs, while poising the electrophilic moiety C for reaction with a nucleophilic moiety of the HDx.
  • C is an electrophilic moiety that is approximately isosteric with an N-acetyl group (i.e., C has approximately the same steric bulk as an N- acetyl group)
  • the element C is capable of reacting, covalently or non-covalently, with a nucleophilic moiety of an HDx.
  • the element C is capable of binding (e.g., by chelation) to a metal ion, e.g., a divalent metal ion, e.g., zinc or calcium.
  • C is selected from die group consisting of ⁇ , ⁇ -epoxyketones, ⁇ , ⁇ -epoxythioketones, ⁇ , ⁇ -epoxysuIfoxides, hydroxamic acids, ⁇ -haloketones, ⁇ -halothioketones, ⁇ -diazoketones, ⁇ - diazothioketones, vinyl epoxides, trifluoromethylketone, trifluoromediylthioketone, enones (e.g., of ketones or thioketones), ynones (e.g., of ketones or thioketones), ⁇ , ⁇ - aziridinoketones, hydrazones, boronic acids, carboxylates, amides (e.g., -C(O)-amino), sulfones, aldehyde, alkyl halides, epoxides, and the like.
  • enones
  • the moieties A, B, and C can illustratively be represented by the formulas depicted in Figure 6, in which Ri represents one or more substituents selected from the group consisting of amino, halogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, heterocyclyl, azido, carboxyl, alkoxycarbonyl, hydroxyl, alkoxy, cyano, trifluoromethyl, and the like; R" is Cj-C 8 alkylidene, C 2 -C 8 alkenylidene, or C 2 -C 8 alkynylidene; R 5 is hydrogen, alkyl, alkoxycarbonyl, aryloxycarbonyl, alkylsulfonyl, arylsulfonyl or aryl; R ⁇ is hydrogen, alkyl, aryl, alkoxy, aryloxy, halogen, and the like; R' 6 is hydrogen, alkyl, alkyl, alk
  • an HDx inhibitor compound can be represented by the formula A-B-C, in which A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclotetrapeptides; B is selected from the group consisting of substituted and unsubstituted C -C alkylidene, C -C 8 alkenylidene, C 4 -C 8 alkynylidene, C -C 8 enyne, and D-E-F, in which D and F are independently absent or a C-C 7 alkylidene, an C 2 -C 7 alkenylidene, or an C 2 -C alkynylidene, and E is O, S, or NR', in which R' represents ⁇ , a lower alkyl, a lower alkenyl, a lower alkynyl, an aralkyl, aryl
  • R'g is hydrogen.
  • B is not a
  • C 4 -C alkylidene if B is a C 4 -C alkylidene, C is not a boronic acid. In other preferred embodiments, the inhibitor compound is not trapoxin.
  • an HDx inhibitor compound can be represented by the formula A-B-C, in which A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclotetrapeptides; B is selected from the group consisting of substituted and unsubstituted C 4 -C 8 alkylidene, C -C 8 alkenylidene, C 4 -C 8 alkynylidene, C -C 8 enyne, and D-E-F, in which D and F are independently absent or C1-C7 alkylidene, C2-C7 alkenylidene, or C2-C7 alkynylidene, and E is O, S, or NR', in which R' represents H, a lower alkyl, a lower alkenyl, a lower alkynyl, an aralkyl, an aryl, or a
  • B is not a C -C 8 alkylidene.
  • the inhibitor compound is not trichostatin.
  • an HDx inhibitor compound can be represented by the formula A-B-C, in which A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclotetrapeptides; B is selected from the group consisting of substituted and unsubstituted C 4 -C alkylidene, C 4 -C alkenylidene, C 4 -C 8 alkynylidene, C 4 -C enyne, and D-E-F, in which D and F are independently absent or a C1-C 7 alkylidene, a C 2 -G7 alkenylidene, or a C 2 -C 7 alkynylidene, and E is O, S, or NR', in which R' is H, lower
  • HDx inhibitor compounds of the present invention may exist in particular geometric or stereoisomeric forms.
  • amino acids can contain at least one chiral center.
  • the present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
  • a particular enantiomer of a compound of die present invention may be prepared by asymmetric synthesis, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomer.
  • diastereomeric salts can be formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers.
  • alkyl refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups.
  • a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C1-C 30 for straight chain, C 3 -C 30 for branched chain), and more preferably 20 or fewer.
  • preferred cycloalkyls have from 4-10 carbon atoms in their ring structure, and more preferably have 5, 6 or 7 carbons in the ring structure.
  • lower alkyl as used herein means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six carton atoms in its backbone structure. Likewise, “lower alkenyl” and “lower alkynyl” have similar chain lengths. Preferred alkyl groups are lower alkyls. In preferred embodiments, a substituent designated herein as alkyl is a lower alkyl.
  • alkyl (or “lower alkyl) as used throughout the specification and claims is intended to include both “unsubstituted alkyls” and “substituted alkyls”, the latter of which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone.
  • substituents can include, for example, halogen, hydroxyl, carbonyl (such as a carboxylate, alkoxycarbonyl, aryloxycarbonyl, alkylcarbonyl, arylcarbonyl, aldehyde, and the like), thiocarbonyl (such as a thioacid, alkoxycarbonyl, and the like), an alkoxyl, unsubstituted amino, mono- or disubstituted amino, amido, amidine, imine, nitro, azido, sulfhydryl, alkylthio, cyano, trifluoromethyl, sulfonato, sulfamoyl, sulfonamido, heterocyclyl, aralkyl, or an aromatic or heteroaromatic moiety.
  • carbonyl such as a carboxylate, alkoxycarbonyl, aryloxycarbonyl, alkylcarbonyl, arylcarbonyl, aldeh
  • moieties substituted on the hydrocarbon chain can themselves be substituted, as described above, if appropriate.
  • exemplary substituted alkyls are described below.
  • Cycloalkyls can be further substituted with, e.g., alkyls, alkenyls, alkoxys, alkylthios, aminoalkyls, carbonyl-substituted alkyls, - CF 3 , -CN, and the like.
  • alkenyl and “alkynyl” refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
  • alkylidene refers to an unsaturated aliphatic moiety having at least one double bond and one triple bond.
  • alkylidene alkenylidene
  • alkynylidene alkynylidene
  • aryl as used herein includes 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, phenyl, pyrrolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, thiazolyl, triazolyl, pyrazolyl, pyridyl, pyrazinyl, pyridazinyl and pyrimidyl, and the like.
  • Those aryl groups having heteroatoms in the ring structure may also be referred to as “aryl heterocycles" or "heteroaromatics”.
  • the aromatic ring can be substituted at one or more ring positions with such substituents as described above, as for example, halogen, azido, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, -CF3, -CN, or the like.
  • substituents as described above, as for example, halogen, azido, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, carbonyl, carboxyl, silyl,
  • aralkyl refers to an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group).
  • aryl group e.g., an aromatic or heteroaromatic group.
  • heterocyclyl or “heterocyclic group” refer to non-aromatic 4- to 10- membered ring structures, more preferably 4- to 7-membered rings, which ring structures include one to four heteroatoms (e.g., O, N, S, P and the like).
  • Heterocyclyl groups include, for example, pyrrolidine, oxolane, thiolane, imidazole, oxazole, piperidine, piperazine, mo ⁇ holine, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like.
  • the heterocyclic ring can be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, alkoxycarbonyl, aryloxycarbonyl, carboxyl, silyl, ether, alkylthio, alkylsulfonyl, arylsulfonyl, ketone (e.g., -C(O)-alkyl or -C(O)-aryl), aldehyde, heterocyclyl, an aryl or heteroaryl moiety, -CF3, -CN, or the like.
  • substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro,
  • halogen e.g., organotin, boronate, aryllithium, cuprate, Grignard reagent, etc.
  • a moieties e.g., specificity elements
  • synthons e.g., by substituting vinylorganometallic compounds for the organometallic aryl compound of the Scheme (followed by further treatment, e.g., reduction, of the vinyl group, if desired, to yield an alkyl A moiety).
  • the carbonyl group can be used for elaboration, e.g., by reduction of the carbonyl group to an alcohol, conversion of the alcohol to a tosylate, and nucleophilic displacement of the tosylate by an acyl compound (e.g., a ketone or ester) to provide a chain-lengthened product (Route A), which can be converted to a C(O)X functionality (e.g., by hydrolysis of an ester and conversion of die resulting carboxylic acid to an acid chloride).
  • an acyl compound e.g., a ketone or ester
  • Route A chain-lengthened product
  • C(O)X functionality e.g., by hydrolysis of an ester and conversion of die resulting carboxylic acid to an acid chloride
  • the carbonyl group of I can be used for olefination (Route B), e.g., Homer-Emmons olefination, to provide an elaborated alkenyl compound.
  • the carbonyl group can be converted to an alkynyl functionality, e.g., via the Corey-Fuchs procedure, to provide an elaborated alkynyl compound.
  • Route B olefination
  • the carbonyl group can be converted to an alkynyl functionality, e.g., via the Corey-Fuchs procedure, to provide an elaborated alkynyl compound.
  • B moieties i.e., linking moieties
  • the C(O)X functionality (e.g., an acid chloride where X is Cl) can be converted to functional groups such as amide, hydrazido, trifluormethylketone, enone, epoxide, aziridine, and the like, through methods conventional in the art.
  • the synthetic pathways shown in the Scheme provide access to compounds having a variety of C moieties (e.g., reactive moieties) suitable for substitution in the subject HDx inhibitors.
  • In vitro chemical synthesis provides a method for generating libraries of compounds that can be screened for ability to bind to or inhibit a target protein, e.g., an
  • HDx Although in vitro methods have previously been used in the pharmaceutical industry to identify potential drugs, recently developed methods have focused on rapidly and efficiently generating and screening large numbers of compounds and are amenable to generating HDx inhibitor compound libraries for use in the subject method. The various approaches to simultaneous preparation and analysis of large numbers of compounds
  • Geysen and co-workers introduced a method for generating compounds by a parallel synthesis on polyacrylic acid-grated polyethylene pins arrayed in the microtitre plate format.
  • about 50 nmol of a single compound was covalently linked to the spherical head of each pin, and interactions of each compound with a receptor or antibody could be determined in a direct binding assay.
  • the Geysen technique can be used to synthesize and screen thousands of compounds per week using the multipin method, and the tethered compounds may be reused in many assays.
  • a variegated library of HDx inhibitor compounds is provided on a set of beads utilizing the strategy of divide-couple-recombine (see, e.g., ⁇ oughten (1985) PNAS 82:5131-5135; and U.S. Patents 4,631,211; 5,440,016; 5,480,971).
  • degeneracy e.g., a plurality of different moieties
  • the beads are divided into as many separate groups to correspond to the number of different residues (e.g., functional groups or other moieties) to be added at that position, the different residues coupled in separate reactions, and the beads recombined into one pool for the next step.
  • the divide-couple-recombine strategy can be carried out using the so-called "tea bag” method first developed by Houghten, where synthesis occurs on resin that is sealed inside porous polypropylene bags (Houghten et al. (1986) PNAS 82:5131-5135). Residues are coupled to the resins by placing the bags in solutions of the appropriate individual activated monomers, while all common steps such as resin washing and deprotection (if appropriate) are performed simultaneously in one reaction vessel. At the end of the synthesis, each bag contains a single compound, and the compounds may be liberated from the resins using a multiple cleavage apparatus (Houghten et al. (1986) Int J Pept Protein Res 27:673-678).
  • a scheme of combinatorial synthesis in which the identity of a compound is given by its locations on a synthesis substrate is termed a spatially-addressable synthesis.
  • the combinatorial process is carried out by controlling the addition of a chemical reagent to specific locations on a solid support (Dower et al. (1991) Annu Rep Med Chem 26:271-280; Fodor, S.P.A. (1991) Science 251:767; Pirrung et al. (1992) U.S. Patent No. 5,143,854; Jacobs et al. (1994) Trends Biotechnol 12:19-26).
  • the technique combines two well-developed technologies: solid-phase synthesis chemistry and photolithography.
  • a synthesis substrate is prepared for compound syntiiesis through die covalent attachment of photolabile nitroveratryloxycarbonyl (NVOC) protected amino linkers.
  • Light is used to selectively activate a specified region of the synthesis support for coupling. Removal of the photolabile protecting groups by lights (deprotection) results in activation of selected areas. After activation, the first of a set of residues, each bearing a photolabile protecting group, is exposed to the entire surface. Coupling only occurs in regions that were addressed by light in the preceding step. The reagent solution is removed, and the substrate is again illuminated through a second mask, activating a different region for reaction with a second protected building block.
  • NVOC photolabile nitroveratryloxycarbonyl
  • the pattern of masks and the sequence of reactants define the products and their locations. Since this process utilizes photolithography techniques, the number of compounds that can be synthesized is limited only by the number of synthesis sites that can be addressed with appropriate resolution. The position of each compound is precisely known; hence, its interactions with other molecules can be directly assessed.
  • the target can be labeled with a fluorescent reporter group to facilitate the identification of specific interactions with individual members of the matrix.
  • the subject mediod provides an HDx inhibitor compound library provided with an encoded tagging system.
  • a recent improvement in the identification of active compounds from combinatorial libraries employs chemical indexing systems using tags that uniquely encode the reaction steps a given bead has undergone and, by inference, the structure it carries.
  • this approach mimics phage display libraries, where activity derives from expressed peptides, but the structures of the active peptides are deduced from the corresponding genomic DNA sequence.
  • the first encoding of synthetic combinatorial libraries employed DNA as die code. Two forms of encoding have been reported: encoding with sequenceable bio-oligomers (e.g., oligonucleotides and peptides), and binary encoding with non-sequenceable tags.
  • a combinatorial library of nominally 7 7 ( 823,543) peptides composed of all combinations of Arg, Gin, Phe, Lys, Val, D-Val and Thr (three-letter amino acid code), each of which was encoded by a specific dinucleotide (TA, TC, CT, AT, TT, CA and AC, respectively), was prepared by a series of alternating rounds of peptide and oligonucleotide synthesis on solid support.
  • the amine linking functionality on die bead was specifically differentiated toward peptide or oligonucleotide synthesis by simultaneously preincubating the beads with reagents that generate protected O ⁇ groups for oligonucleotide synthesis and protected N ⁇ 2 groups for peptide synthesis (here, in a ratio of 1:20).
  • the tags each consisted of 69-mers, 14 units of which carried the code.
  • the bead-bound library was incubated with a fluorescently labeled antibody, and beads containing bound antibody that fluoresced strongly were harvested by fluorescence-activated cell sorting (FACS).
  • FACS fluorescence-activated cell sorting
  • the DNA tags were amplified by PCR and sequenced, and the predicted peptides were synthesized. Following such techniques, HDx inhibitor compound libraries can be derived and screened using HDxs of the subject invention.
  • oligonucleotide tags permits extremelyly sensitive tag analysis. Even so, the method requires careful choice of orthogonal sets of protecting groups required for alternating co-synthesis of the tag and the library member. Furthermore, the chemical lability of the tag, particularly the phosphate and sugar anomeric linkages, may limit the choice of reagents and conditions that can be employed for the synthesis on non- oligomeric libraries.
  • the libraries employ linkers permitting selective detachment of the test HDx inhibitor compound library member for bioassay, in part (as described infra) because assays employing beads limit the choice of targets, and in part because the tags are potentially susceptible to biodegradation.
  • branched linkers are employed so that the coding unit and the test peptide are both attached to the same functional group on the resin.
  • a linker can be placed between the branch point and the bead so that cleavage releases a molecule containing both code and ligand (Ptek et al. (1991) Tetrahedron Lett 32:3891-3894).
  • the linker can be placed so that the test peptide can be selectively separated from the bead, leaving the code behind. This last construct is particularly valuable because it permits screening of the test peptide without potential interference, or biodegradation, of the coding groups. Examples in the art of independent cleavage and sequencing of peptide library members and their corresponding tags has confirmed that the tags can accurately predict the peptide structure.
  • peptide tags are more resistant to decomposition during ligand synthesis than are oligonucleotide tags, but they must be employed in molar ratios nearly equal to those of the ligand on typical 130 ⁇ m beads in order to be successfully sequenced.
  • oligonucleotide encoding the use of peptides as tags requires complex protection/deprotection chemistries.
  • Non-sequenceable tagging binary encoding
  • An alternative form of encoding the test peptide library employs a set of non- sequenceable tagging molecules (e.g., molecules having electrophoric moieties) that are used as a binary code (Ohlmeyer et al. (1993) PNAS 90:10922-10926).
  • exemplary tags are haloaromatic alkyl ethers that are detectable as their trimethylsilyl ethers at less than femtomolar levels by electron capture gas chromatography (ECGC). Variations in the length of the alkyl chain, as -well as the nature and position of the aromatic halide substituents, permit the synthesis of at least 40 such tags, which in principle can encode 2 40 (e.g., upwards of IO 12 ) different molecules.
  • ECGC electron capture gas chromatography
  • the tags were bound to about 1% of the available amine groups of a peptide library via a photocleavable O-nitrobenzyl linker.
  • This approach is convenient when preparing combinatorial libraries of peptides or other amme-containing molecules.
  • a more versatile system has, however, been developed that permits encoding of essentially any combinatorial library.
  • the ligand is attached to the solid support via the photocleavable linker and d e tag is attached through a catechol ether linker via carbene insertion into the bead matrix (Nestler et al. (1994) J Org Chem 59:4723-4724).
  • This orthogonal attachment strategy permits the selective detachment of library members for bioassay in solution and subsequent decoding by ECGC after oxidative detachment of the tag sets.
  • Binary encoding with tags e.g., electrophoric tags
  • tags has been particularly useful in defining selective interactions of substrates with synthetic receptors (Borchardt et al. (1994) J Am Chem Soc 116:373-374), and model systems for understanding the binding and catalysis of biomolecules. Even using detailed molecular modeling, the identification of the selectivity preferences for synthetic receptors has required the manual synthesis of dozens of potential substrates. The use of encoded libraries makes it possible to rapidly examine all the members of a potential binding set.
  • Successive photoelution also permits a very high throughput iterative screening strategy: first, multiple beads are placed in 96-well microtiter plates; second, ligands are partially detached and transferred to assay plates; third, a bioassay identifies the active wells; fourth, the corresponding beads are rearrayed singly into new microtiter plates; fifth, single active compounds are identified; and sixth, the structures are decoded.
  • HDx inhibitor compounds can be synthesized on solid support by appropriate functionalization for attachment to a solid matrix, or altematively, by solution-phase synthesis followed by immobilization through an appropriate functional group.
  • an HDx inhibitor compound which is analogous to trichostatin, can be synthesized on a solid support by attachment through an amino group of the specificity element A, as shown in Figure 7.
  • the solid support is preferably capable of withstanding synthetic conditions required to synthesize the requisite compounds.
  • the compound can preferably be released from the solid support, e.g., by selective cleavage of an amide bond.
  • the synthetic steps employed to synthesize compounds on solid support are preferably selected to allow a wide variety of residues (e.g., building blocks) to be coupled to the immobilized moieties, preferably under mild conditions.
  • Suitable reaction chemistries include well-known carbon-carbon bond forming reactions such as the Stille and Suzuki couplings, as well as ⁇ orner-Emmons reactions, Ni/Cr mediated couplings, and the like.
  • Particularly preferred coupling reactions can be performed in the presence of water and do not require harsh conditions or expensive reagents.
  • (tributyltin)anilines (in which Rj represents one or more substitutions, e.g., hydrogen, halogen, alkyl, alkoxy, and the like) are coupled in a plurality of reaction vessels to beads of a solid support (e.g., Affigel).
  • the beads are further divided into a plurality of reaction vessels, and suspended in a solvent such as DMF, and one acid chloride building block (corresponding to linking element B) is introduced into each vessel (R 2 and R 3 represent, e.g., hydrogen, halogen, alkyl, and the like; and the broken line represents an optional double bond).
  • the reactions are stirred under an inert gas (e.g.
  • a palladium catalyst e.g., Pd(PPh 3 ) 4
  • Pd(PPh 3 ) 4 a palladium catalyst
  • the reaction is stirred for 1-24 hours.
  • the beads are washed, and placed in a plurality of vessels.
  • the aldehyde moiety is deprotected by mild acid treatment (e.g., PPTS in MeO ⁇ ), and the beads are again washed and placed in a plurality of reaction vessels, and the beads are suspended in dry acetonitrile.
  • One building block (corresponding to the reactive element C) is then added to each reaction vessel.
  • a plurality of phosphonates can be employed (R 4 represents, e.g., alkyl, alkenyl, alkynyl, alkoxy, and the like).
  • a ⁇ orner-Emmons reaction is performed by addition of LiCl (1.1 equiv.) and diisopropylethylamine (DD?EA) or DBU (1.2 equiv).
  • the beads are washed with water and acetonitrile, and then dried to yield a library of candidate HDx inhibitor compounds on solid support.
  • the compounds can then be released from the solid support into solution; or the compounds can be screened while attached to the solid support.
  • the above combinatorial synthesis can be performed in an encoded mode, e.g., the binary tagging method described supra, by addition of the appropriate tag for each monomer.
  • this mode after each reaction has been performed and the corresponding tag attached, the beads from all reactions can be recombined and then divided into aliquots for further derivatization.
  • This method provides the advantage of ease of handling when large libraries are to be synthesized.
  • the combinatorial library can be screened for activity according to known methods (see, e.g., Gordon et al., supra).
  • the present invention provides pharmaceutically acceptable compositions which comprise a therapeutically-effective amount of one or more of the compounds described above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents.
  • the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example; drenches (aqueous or non-aqueous solutions or suspensions), tablets, boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (3) topical application, for example, as a cream, ointment or spray applied to the skin; or (4) intravaginally or intrarectally, for example, as a pessary, cream or foam.
  • terapéuticaally-effective amount means that amount of a compound, material, or composition comprising a deacetylase inhibitor of the present invention which is effective for producing some desired therapeutic effect by inhibiting histone deacetylation in at least a sub-population of cells in an animal and thereby blocking the biological consequences of that event in the treated cells, at a reasonable benefit/risk ratio applicable to any medical treatment.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact wid the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically-acceptable carrier means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject deacetylase inhibitor agent from one organ, or portion of d e body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically- acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as co starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil and soybean oil; (10) glycois, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl iaurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydrox
  • certain embodiments of the present deacetylase inhibitors may contain a basic functional group, such as amino or alkylamino, and are, thus, capable of forming pharmaceutically-acceptable salts with pharmaceutically-acceptable acids.
  • pharmaceutically-acceptable salts refers to the relatively non-toxic, inorganic and organic acid addition salts of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by separately reacting a purified compound of the invention in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, Iaurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like.
  • sulfate bisulfate
  • phosphate nitrate
  • acetate valerate
  • oleate palmitate
  • stearate Iaurate
  • benzoate lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like.
  • the deacetylase inhibitory compounds of die present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically-acceptable salts with pharmaceutically-acceptable bases.
  • pharmaceutically-acceptable salts refers to the relatively non-toxic, inorganic and organic base addition salts of compounds of the present invention. These salts can likewise be prepared in situ during the final isolation and purification of the compounds, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically- acceptable metal cation, with ammonia, or with a pharmaceutically-acceptable organic primary, secondary or tertiary amine.
  • a suitable base such as the hydroxide, carbonate or bicarbonate of a pharmaceutically- acceptable metal cation, with ammonia, or with a pharmaceutically-acceptable organic primary, secondary or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like.
  • Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like. (See, for example, Berge et al., supra)
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • Formulations of the present invention include those suitable for oral, nasal, topical
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the deacetylase inhibitor which produces a tiierapeutic effect. Generally, out of one hundred per cent, tiiis amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
  • Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention widi the carrier and, optionally, one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association a deacetylase inhibitor of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient.
  • a deacetylase inhibitor of the present invention may also be administered as a bolus, electuary or paste.
  • the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) abso ⁇ tion accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as,
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycois and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered deacetylase inhibitor moistened with an inert liquid diluent.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention may optionally be scored or prepared with coatings and shells, such as enteric coatings and odier coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres.
  • compositions may be sterilized by, for example, filtration through a bacteria-retaining filter, or by inco ⁇ orating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions which can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration of the deacetylase inhibitors of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycois and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents, so
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • Suspensions in addition to the active deacetylase inhibitor, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active deacetylase inhibitor.
  • suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active deacetylase inhibitor.
  • Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of a deacetylase inhibitor of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams and gels may contain, in addition to an active deacetylase inhibitor of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycois, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycois, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body.
  • dosage forms can be made by dissolving or dispersing the deacetylase inhibitor in the proper medium.
  • Abso ⁇ tion enhancers can also be used to increase the flux of the deacetylase inhibitor across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the deacetylase inhibitor in a polymer matrix or gel.
  • Ophthalmic formulations are also contemplated as being within the scope of this invention.
  • compositions of this invention suitable for parenteral administration comprise one or more deacetylase inhibitors of the invention in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged abso ⁇ tion of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay abso ⁇ tion such as aluminum monostearate and gelatin.
  • adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as
  • a dmg in order to prolong the effect of a dmg, it is desirable to slow the abso ⁇ tion of the dmg from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amo ⁇ hous material having poor water solubility. The rate of abso ⁇ tion of die dmg then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Altematively, delayed abso ⁇ tion of a parenterally-administered dmg form is accomplished by dissolving or suspending the dmg in an oil vehicle.
  • Injectable depot forms are made by forming microencapsule matrices of the subject deacetylase inhibitors in biodegradable polymers such as polylactide- polyglycolide. Depending on the ratio of dmg to polymer, and the nature of the particular polymer employed, the rate of dmg release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the dmg in liposomes or microemulsions which are compatible with body tissue.
  • the compounds of the present invention When the compounds of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • the preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given by forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infiision or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administration is preferred.
  • deacetylase inhibitor may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally and topically, as by powders, ointments or drops, including buccally and sublingually.
  • routes of administration including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally and topically, as by powders, ointments or drops, including buccally and sublingually.
  • the compounds of the present invention which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of factors including the activity of the particular deacetylase inhibitor employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular deacetylase inhibitor employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve d e desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • Another aspect of the present invention relates to a method of inducing and/or maintaining a differentiated state, enhancing survival, and/or inhibiting (or altematively potentiating) proliferation of a cell, by contacting the cells with an agent which modulates HDx-dependent transcription.
  • an agent which modulates HDx-dependent transcription for instance, it is contemplated by the invention that, in light of the present finding of an apparently broad involvement of HDx proteins in the control of chromatin stmcture and, thus, transcription and replication, the subject method could be used to generate and/or maintain an array of different tissue both in vitro and in vivo.
  • An "HDx therapeutic,” whether inhibitory or potentiating with respect to modulating histone deacetylation, can be, as appropriate, any of the preparations described above, including isolated polypeptides, gene therapy constmcts, antisense molecules, peptidomimetics or agents identified in the dmg assays provided herein.
  • the HDx compounds of the present invention are likely to play an important role in the modulation of cellular proliferation.
  • pathological cell proliferative conditions for which HDx therapeutics of the present invention may be used in treatment.
  • such agents can provide therapeutic benefits where the general strategy being the inhibition of an anomalous cell proliferation.
  • Diseases that might benefit from this methodology include, but are not limited to various cancers and leukemias, psoriasis, bone diseases, fibroproliferative disorders such as involving connective tissues, atherosclerosis and other smooth muscle proliferative disorders, as well as chronic inflammation.
  • the present invention contemplates the use of HDx therapeutics for the treatment of differentiative disorders which result from, for example, de-differentiation of tissue which may (optionally) be accompanied by abortive reentry into mitosis, e.g. apoptosis.
  • degenerative disorders include chronic neurodegenerative diseases of the nervous system, including Alzheimer's disease, Parkinson's disease, Huntingdon's chorea, amylotrophic lateral sclerosis and the like, as well as spinocerebellar degenerations.
  • disorders associated with connective tissue such as may occur due to de- differentiation of chondrocytes or osteocytes, as well as vascular disorders which involve de-differentiation of endothelial tissue and smooth muscle cells, gastric ulcers characterized by degenerative changes in glandular cells, and renal conditions marked by failure to differentiate, e.g. Wilm's tumors.
  • connective tissue such as may occur due to de- differentiation of chondrocytes or osteocytes
  • vascular disorders which involve de-differentiation of endothelial tissue and smooth muscle cells, gastric ulcers characterized by degenerative changes in glandular cells, and renal conditions marked by failure to differentiate, e.g. Wilm's tumors.
  • the subject HDx therapeutics can be used to reform injured tissue, or to improve grafting and mo ⁇ hology of transplanted tissue.
  • HDx antagonists and agonists can be employed in a differential manner to regulate different stages of organ repair after physical, chemical or pathological insult.
  • such regimens can be utilized in repair of cartilage, increasing bone density, liver repair subsequent to a partial hepatectomy, or to promote regeneration of lung tissue in the treatment of emphysema.
  • the present method is also applicable to cell culture techniques.
  • the HDx therapeutic of the present invention can be used to induce differentiation of uncommitted progenitor cells and thereby give rise to a committed progenitor cell, or to cause further restriction of the developmental fate of a committed progenitor cell towards becoming a terminally-differentiated cell.
  • the present method can be used in vitro or in vivo to induce and/or maintain the differentiation of hematopoietic cells into erythrocytes and other cells of the hematopoietic system.
  • the effect of erythropioetin (EPO) on the growth of EPO-responsive erythroid precursor cells is increased to influence their differentiation into red blood cells.
  • the HDx therapeutics of the present invention can be administered alone or in conjunction with EPO and in a suitable carrier to vertebrates to promote erythropoiesis.
  • cells could be treated ex vivo.
  • Such treatment is contemplated in die treatment of a variety of disease states, including in individuals who require bone marrow transplants (e.g. patients with aplastic anemia, acute leukemias, recurrent lymphomas, or solid tumors).
  • a recipient prior to receiving a bone marrow transplant, a recipient is prepared by ablating or removing endogenous hematopoietic stem cells. Such treatment is usually carried out by total body irradiation or delivery of a high dose of an alkylating agent or other chemotherapeutic, cytotoxic agent, Anklesaria, et al. (1987) PNAS 84:7681-7685). Following preparation of the recipient, donor bone marrow cells are injected intravenously.
  • the HDx therapeutics of the present invention could be contacted with the cells ex vivo or administered to the subject with the reimplanted cells.
  • the present invention contemplates the use of agents which modulate histone deacetylase activity to specifically inhibit or activate certain cell types.
  • T cell proliferation could be preferentially inhibited in order to induce tolerance by using a procedure similar to that for inducing tolerance using sodium butyrate (see, for example, PCT7US93/03045).
  • the HDx therapeutics of the present invention may be used to induce antigen- specific tolerance in any situation in which it is desirable to induce tolerance, such as autoimmune diseases, in allogeneic or xenogeneic transplant recipients, or in graft versus host (GV ⁇ ) reactions.
  • tolerance will typically be induced by presenting the tolerizing compound (e.g., an HDx inhibitor) substantially contemporaneously with the antigen, i.e. reasonably close together in time with the antigen.
  • the HDx therapeutic will be administered after presentation of the antigen, so that they will have their effect after the particular repertoire of Th cells begins to undergo clonal expansion.
  • compositions comprising HDx therapeutics can also be utilized for both cell culture and therapeutic methods involving generation and maintenance of tissue.
  • the subject HDx therapeutics will be useful in increasing the amount of protein produced by a cell or recombinant cell.
  • the cell may include any primary cell isolated from any animal, cultured cells, immortalized cells, and established cell lines.
  • the animal cells used in the present invention include cells which intrinsically have an ability to produce a desired protein; cells which are induced to have an ability to produce a desired protein, for example, by stimulation with a cytokine such as an interferon, an interleukin; genetically engineered cells into which a gene for a desired protein is introduced.
  • the protein produced by the process could include any peptides or proteins, including peptide hormone or proteinaceous hormones such as any useful hormone, cytokine, interleukin, or protein which it may be desirable to have in purified form and/or in large quantity.
  • Another aspect of the invention features transgenic non-human animals which express a heterologous HDx gene of the present invention, or which have had one or more genomic HDx genes dismpted in at least one of the tissue or cell-types of the animal.
  • the invention features an animal model for developmental diseases, which animal has one or more HDx allele which is mis-expressed.
  • a mouse can be bred which has one or more HDx alleles deleted or otherwise rendered inactive.
  • Such a mouse model can then be used to study disorders arising from mis-expressed HDx genes, as well as for evaluating potential therapies for similar disorders.
  • transgenic animals which are comprised of cells (of that animal) which contain a transgene of the present invention and which preferably (though optionally) express an exogenous HDx protein in one or more cells in the animal.
  • An HDx transgene can encode the wild-type form of the protein, or can encode homologs thereof, including both agonists and antagonists, as well as antisense constructs.
  • the expression of the transgene is restricted to specific subsets of cells, tissues or developmental stages utilizing, for example, cis-acting sequences that control expression in the desired pattern.
  • such mosaic expression of an HDx protein can be essential for many forms of lineage analysis and can additionally provide a means to assess the effects of, for example, lack of HDx expression which might grossly alter development in small patches of tissue within an otherwise normal embryo.
  • tissue-specific regulatory sequences and conditional regulatory sequences can be used to control expression of the transgene in certain spatial pattems.
  • temporal pattems of expression can be provided by, for example, conditional recombination systems or prokaryotic transcriptional regulatory sequences.
  • target sequence refers to a nucleotide sequence that is genetically recombined by a recombinase.
  • the target sequence is flanked by recombinase recognition sequences and is generally either excised or inverted in cells expressing recombinase activity.
  • Recombinase catalyzed recombination events can be designed such diat recombination of the target sequence results in either the activation or repression of expression of one of the subject HDx proteins.
  • excision of a target sequence which interferes with the expression of a recombinant HDx gene can be designed to activate expression of that gene.
  • This interference with expression of the protein can result from a variety of mechanisms, such as spatial separation of die HDx gene from the promoter element or an internal stop codon.
  • the transgene can be made wherein d e coding sequence of the gene is flanked by recombinase recognition sequences and is initially transfected into cells in a 3' to 5' orientation with respect to the promoter element.
  • inversion of the target sequence will reorient the subject gene by placing the 5' end of the coding sequence in an orientation with respect to the promoter element which allow for promoter driven transcriptional activation.
  • cre/loxP recombinase system of bacteriophage PI (Lakso et al. (1992) PNAS 89:6232-6236; Orban et al. (1992) PNAS 89:6861-6865) or the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al. (1991) Science 251 :1351-1355; PCT publication WO 92/15694) can be used to generate in vivo site-specific genetic recombination systems. Cre recombinase catalyzes the site-specific recombination of an intervening target sequence located between loxP sequences.
  • loxP sequences are 34 base pair nucleotide repeat sequences to which the Cre recombinase binds and are required for Cre recombinase mediated genetic recombination.
  • the orientation of loxP sequences determines whether the intervening target sequence is excised or inverted when Cre recombinase is present (Abremski et al. (1984) J. Biol. Chem. 259:1509-1514); catalyzing the excision of the target sequence when the loxP sequences are oriented as direct repeats and catalyzes inversion of the target sequence when loxP sequences are oriented as inverted repeats.
  • genetic recombination of the target sequence is dependent on expression of the Cre recombinase.
  • Expression of the recombinase can be regulated by promoter elements which are subject to regulatory control, e.g., tissue-specific, developmental stage-specific, inducible or repressible by externally added agents. This regulated control will result in genetic recombination of the target sequence only in cells where recombinase expression is mediated by the promoter element.
  • the activation expression of a recombinant HDx protein can be regulated via control of recombinase expression.
  • cre loxP recombinase system to regulate expression of a recombinant HDx protein requires the constmction of a transgenic animal containing transgenes encoding both d e Cre recombinase and the subject protein. Animals containing both the Cre recombinase and a recombinant HDx gene can be provided through the constmction of "double" transgenic animals.
  • a convenient method for providing such animals is to mate two transgenic animals each containing a transgene, e.g., an HDx gene and recombinase gene.
  • One advantage derived from initially constmcting transgenic animals containing an HDx transgene in a recombinase-mediated expressible format derives from the likelihood that the subject protein, whether agonistic or antagonistic, can be deleterious upon expression in the transgenic animal.
  • a founder population in which the subject transgene is silent in all tissues, can be propagated and maintained. Individuals of this founder population can be crossed with animals expressing the recombinase in, for example, one or more tissues and/or a desired temporal pattern.
  • prokaryotic promoter sequences which require prokaryotic proteins to be simultaneous expressed in order to facilitate expression of the HDx transgene.
  • Exemplary promoters and the corresponding trans-activating prokaryotic proteins are given in U.S. Patent No. 4,833,080.
  • conditional transgenes can be induced by gene therapy-like methods wherein a gene encoding the trans-activating protein, e.g. a recombinase or a prokaryotic protein, is delivered to the tissue and caused to be expressed, such as in a cell-type specific manner.
  • a gene encoding the trans-activating protein e.g. a recombinase or a prokaryotic protein
  • the "transgenic non-human animals" of the invention are produced by introducing transgenes into the germline of the non-human animal.
  • Embryonic target cells at various developmental stages can be used to introduce transgenes. Different methods are used depending on the stage of development of the embryonic target cell.
  • the zygote is the best target for micro-injection. In the mouse, the male pronucleus reaches the size of approximately 20 micrometers in diameter which allows reproducible injection of l-2pl of DNA solution.
  • the use of zygotes as a target for gene transfer has a major advantage in that in most cases the injected DNA will be inco ⁇ orated into the host gene before the first cleavage (Brinster et al.
  • Retroviral infection can also be used to introduce HDx transgenes into a non- human animal.
  • the developing non-human embryo can be cultured in vitro to the blastocyst stage. During this time, the blastomeres can be targets for retroviral infection (Jaenich, R. (1976) PNAS 73 : 1260-1264).
  • Efficient infection of the blastomeres is obtained by enzymatic treatment to remove the zona pellucida (Manipulating the Mouse Embryo, ⁇ ogan eds. (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1986).
  • the viral vector system used to introduce the transgene is typically a replication-defective retrovirus carrying the transgene (Jahner et al. (1985) PNAS 82:6927-6931; Van der Putten et al. (1985) PNAS 82:6148-6152).
  • Transfection is easily and efficiently obtained by culturing the blastomeres on a monolayer of vims-producing cells (Van der Putten, supra; Stewart et al. (1987) EMBO J.
  • infection can be performed at a later stage.
  • Vims or vims-producing cells can be injected into the blastocoele (Jahner et al. (1982) Nature 298:623-628).
  • Most of the founders will be mosaic for the transgene since inco ⁇ oration occurs only in a subset of the cells which formed the transgenic non-human animal. Further, the founder may contain various retroviral insertions of the transgene at different positions in the genome which generally will segregate in the offspring.
  • a third type of target cell for transgene introduction is die embryonic stem cell
  • ES cells are obtained from pre-implantation embryos cultured in vitro and fused with embryos (Evans et al. (1981) Nature 292:154-156; Bradley et al. (1984) Nature 309:255-258; Gossler et al. (1986) PNAS 83: 9065-9069; and Robertson et al. (1986) Nature 322:445-448).
  • Transgenes can be efficiently introduced into the ES cells by DNA transfection or by retrovims-mediated transduction.
  • Such transformed ES cells can thereafter be combined with blastocysts from a non-human animal. The ES cells thereafter colonize the embryo and contribute to the germ line of the resulting chimeric animal.
  • Jaenisch, R. (1988) Science 240:1468-1474 For review see Jaenisch, R. (1988) Science 240:1468-1474.
  • HDx knock-out or dismption transgenic animals are also generally known. See, for example, Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986). Recombinase dependent knockouts can also be generated, e.g. by homologous recombination to insert recombinase target sequences flanking portions of an endogenous HDx gene, such that tissue specific and/or temporal control of inactivation of an HDx allele can be controlled as above. Exemplification
  • Trapoxin is a microbially derived cyclotetrapeptide that inhibits histone deacetylation in vivo and causes mammalian cells to arrest in the cell cycle.
  • a trapoxin affinity matrix was used to isolate two nuclear proteins that copurified with histone deacetylase activity. Both proteins were identified by peptide microsequencing, and a cDNA encoding die histone deacetylase catalytic subunit ⁇ HDl) was cloned from a Jurkat T cell library.
  • the predicted protein is highly similar to the yeast transcriptional regulator RPD3, this study supports a role for histone deacetylase as a key regulator of eukaryotic transcription.
  • Trapoxin is an "irreversible" inhibitor of histone deacetylase activity and its molecular stmcture offers clues as to how it could form a covalent bond with a nucleophilic active site residue.
  • trapoxin contains an electrophilic epoxyketone that is essential for biological activity (5).
  • the aliphatic epoxyketone side chain is approximately isosteric with N-acetyl lysine ( Figure IA). Trapoxin likely acts as a substrate mimic, with epoxyketone poised to alkylate an active site nucleophile. We therefore regarded trapoxin as a tool that could reveal the molecular identity of histone deacetylase, so that its role in transcriptional regulation and cell cycle progression could be elucidated.
  • Tritium-Iabeled trapoxin was prepared by total synthesis and used to identify trapoxin binding protein in crude extracts from bovine thymus. We used a charcoal precipitation assay to detect a specific trapoxin binding activity primarily in the nuclear fraction of the extracts (6). The binding activity was saturable with nanomolar concentrations of [ 3 H]trapoxin and was completed by the simultaneous addition of unlabled trapoxin. Trichostatin also competed with [ 3 H]trapoxin (f° r synthesis, see Example 2), suggesting that both of these compounds exert their cellular effects by targeting the same molecule.
  • thymocytes (-12 g) prepared from fresh bovine thymus were homogenized in hypotonic lysis buffer [20 mM tris (pH 7.8), 20 mM NaCl, 1 mM EDTA, 10% glycerol, ImM PMSF, ImM benzamidine, 10 ⁇ g/ml each of pepstatin, aprotinin, and leupeptin] by mechanical dismption and the nuclei were isolated by centrifugation at 3000g. Nuclei were resuspended in lysis buffer and the proteins were extracted with 0.4 M ammonium sulfate. The viscous lysate was sonicated and clarified by centrifugation at 100,000g for one hour.
  • hypotonic lysis buffer 20 mM tris (pH 7.8), 20 mM NaCl, 1 mM EDTA, 10% glycerol, ImM PMSF, ImM benzamidine, 10 ⁇ g/ml each of pep
  • Proteins were then precipitated with 90% saturated ammonium sulfate and recovered by centrifugation (100,000g, one hour). After through dialysis against Q buffer (25 mM tris pH 8, 10 mM NH Cl, 0.25 mM EDTA, 10% glycerol), a portion of the nuclear proteins (-12 mg total protein) was loaded onto a HR 10/10 Mono Q column (Pharmacia). The column was washed with 25ml Q buffer and eluted with a 50 ml linear gradient of 10 to 500 mM NH4 Cl. The column was further washed with 25 ml 500 mM Nl and 25 ml 1 M histone deacetylase activities or further purified with the K-trap affinity matrix. All procedures were done at 4°C.
  • Q buffer 25 mM tris pH 8, 10 mM NH Cl, 0.25 mM EDTA, 10% glycerol
  • K-trap was deprotected with Pd(Ph 3 P) 4 and coupled to an activated agarose matrix ( Figure 2 A).
  • Mono Q fractions containing nuclear proteins from bovine thymus were incubated with the K-trap affinity matrix and then tested for both trapoxin binding and histone deacetylase activity. Both activities were depleted (90%) by treatment with the K-trap matrix, yet a control matrix capped with ethanolamine had no effect on either activity (8).
  • Bound polypeptides were eluted by boiling the matrix in 1% SDS buffer and separated b polyacrylamide gel electrophoresis.
  • bovine proteins led to the resolution of two major bands of -46 and -50 kD in the final preparative electrophoresis step, both of which were submitted for microsequencing.
  • nuclear ammonium sulfate pellets from 15 bovine thymuses were prepared as described above. Sedimented proteins were resuspended in and dialyzed against buffer A [20 mM bistris (pH 7.2), 20 mM NaCl, 10% glycerol] for 12 hours, and brought to pH 5.8 by dialyzing against bugger A (pH 5.8) for 30 minutes.
  • the dialysate (-650 mg protein) was loaded onto a Q Sepharose FF column (2.6 x 10 cm; Pharmacia) and the column was washed with 120 ml buffer A (pH 5.8). Proteins wee eluted with a 400 ml linear gradient of 20 to 600 mM NaCl in buffer A. Fractions (10 ml; each fraction contained 1 ml of 1 M tris pH 8 to neutralize the acidic buffer A) were assayed for trapoxin binding activity. Tween-20 was added to active fractions at a final concentration of 0.05%, and these fractions were incubated with K-trap affinity matrix for 16 hours (25 ⁇ l per ml Q fraction).
  • the bovine protein of larger molecular size corresponds to a known protein, RbAp48 (11), that consists of seven WD repeat domains (12). Originally identified as a protein that binds to the retinoblastoma gene product (pRb), RbAp48 may constitute an adaptor subunit that targets the histone deacetylase to specific chromatin domains.
  • the -46 kD bovine protein is highly related to the protein encoded by the yeast RPD3 gene, which has been implicated by several genetic screens as a transcriptional regulator, but whose biochemical function is unknown (13).
  • Partial cDNA sequences for the human gene were identified in the expressed sequence tag database (dbEST) and were used to design polymerase chain reaction (PCR) primers. Briefly, after noting sequence similarity between peptides derived from the purified bovine trapoxin binding protein and yeast RPD3, we checked dbEST to see whether any partial sequences for the human homologue had been reported.
  • ESTs Two ESTs (Genbank accession numbers: D31480 and F07807) were identified whose predicted translation products aligned widi high sequence similarity to NH 2 - and COOH-terminal regions of HDl, respectively, PCR primers were designed based on these tags and a one kilobase PCR product was obtained from a Jurkat cDNA library (Stratagene). A 32 P labeled probe prepared by random priming was used to screen the Jurkat library, and ten positive clones were isolated. One of the clones was fully sequenced and found to contain a putative full-length open reading frame (Figure 3A).
  • HDl like RbAp48 (11), is detected predominantly in the nucleus by immunostaining with the aforementioned antibodies (8).
  • HDl and RbAp48 are the major proteins eluted from the K-trap matrix ( Figure 2B), it is likely that they interact directly with one another.
  • HDl is 60% identical to the protein encoded by the yeast RPD3 gene, which was isolated in four independent mutant suppressor screens designed to identify transcriptional repressors (13, 16, 17, 18, 19). No biochemical function for the yeast protein has previously been postulated.
  • a negative regulator of the TRK2 gene, RPD3 is necessary for the transcriptional repression of several genes whose expression is regulated according to specific environmental conditions. Loss of RPD3 also leads to decreased transcriptional activation of certain genes, but this effect may be indirect (13, 17).
  • the SIR3 and SIR4 proteins have been shown to interact with a bacterially expressed histone H4 NH -terminal domain in vitro (25), and it is possible that deacetylation of one or more lysine residues is required for this interaction in vivo.
  • Our results further support a role for histone deacetylase as a transcriptional regulator and establish a biochemical connection to the genetic studies that originally characterized RPD3.
  • 3H-Trapoxin was prepared from (S.S)-threitol acetonide (9) by total synthesis, as outlined in Figures 8A-8C.
  • (S,S)-threitol acetonide (9) was monoprotected by treatment with triisopropylsilylchloride (TLPSC1) and sodium hydride in tetrahydrofuran (THF). The free alcohol was then subjected to Swem oxidation. Wittig reaction of die resulting aldehyde gave compound 10 in good yield for the three steps. Compound 10 was then hydrogenated with deprotection of the primary alcohol, which was then converted to the bromide 11 in excellent yield. Bromide 11 was converted to the organocuprate and reacted with (S)-serine ⁇ -lactone to yield the benzyloxycarbonyl- (Cbz) protected amino acid 12.
  • TLPSC1 triisopropylsilylchloride
  • THF tetrahydrofuran
  • Cyclotetrapeptide 18 was tritiated, as shown in Figure 8C, by oxidation of the primary alcohol with the Dess-Martin reagent, and the aldehyde was reduced with tritiated sodium borohydride to provide tritiated 18, which was converted to [ 3 H]Trapoxin B by tosylation of the primary alcohol, deprotection of the diol, epoxide ring closure, and oxidation of the secondary alcohol to yield the desired compound.
  • Non-radiolabelled 18 was converted to [ 3 H]Trapoxin B, via tosylate 19, in 68% overall yield.
  • K-Trap was prepared from (S,S)-threitol acetonide (9) by total synthesis, as outlined in Figures 9A-9C.
  • Figure 9A monoprotection and Swem oxidation of 9 yielded the aldehyde as above.
  • Wittig homologation yielded carboxylic acid 20, which was converted to the mixed anhydride and treated with lithiated oxazolidinone 21 to provide 22 in excellent yield.
  • Deprotection of the primary alcohol and conversion to the tosylate were followed by treatment of the potassium enolate with trisylazide according to the method of Evans to effect electrophilic azide transfer in good overall yield and stereoselectivity, providing compound 23.
  • compound 28 was converted to K-trap (29) by deprotection of the diol, base-promoted epoxide closure, and oxidation of the secondary alcohol to provide K-trap (29) in good overall yield.
  • the K-trap affinity matrix 30 was provided by palladium-catalyzed removal of the allyloxycarbonyl (Alloc) group from the lysine residue of 29, and immobilization on Affigel 10.
  • Histone Deacetylase Activity is Required for Full Transcriptional Repression by mSin3 A
  • BHLHZip basic region-Helix-Loop-Helix-Leucine Zipper
  • the second PAH motif in mSin3A, mSin3B and Sin3p interacts with the mSin3 interaction domain or SID in the amino terminus of the four Mad family members (Ayer et al., 1995; Schreiber-Agus et al., 1995; Hurlin et al., 1995a; Kasten et al., 1996). Madl, Max and mSin3A form ternary complexes capable of binding DNA (Ayer et al., 1995).
  • IBD AC 1 is approximately 60% identical to the S. cerevisiae RPD3 protein, which is a component of a yeast histone deacetylase complex (Rundlett et al., 1996). Single mutations in either RPD3 or SIN3 give the same phenotypes as RPD3/SIN3 double, mutants suggesting that they function in the same pathway (Stillman et al., 1994).
  • Mad family proteins use mSin3A as a corepressor and Madl can repress transcription in wild-type yeast but not yeast having a null mutation in SIN3 (Kasten et al., 1996) or RPD3 (D.J. Stillman, personal comm.), it is likely that the mechanism of transcriptional repression by Mad proteins may be conserved between yeast and higher eukaryotes. Consistent with this hypothesis, the DNA-binding transcription factor YY1 interacts with a mammalian RPD3 homolog to repress transcription of a heterologous reporter gene (Yang et. at, 1996). These results demonstrate that mammalian RPD3-like activity functions in transcriptional regulation.
  • histone hyperacetylation correlates with transcriptionally active or poised genes; conversely, hypoacetylation correlates with transcriptionally repressed heterochromatin (for reviews see: Turner, 1993; Loidl, 1994; Wolffe, 1996).
  • mSin3A is present in cells as a large stable multiprotein complex.
  • HDACl and RbAp48 are components of the mSin3A complex. Because SIN3 and RPD3 appear to function in the same pathway in yeast and two components of the mSin3A complex, p50 and p55, are similar in apparent molecular weight to HDACl we hypothesized that HDACl or related proteins might be components of the mSin3A repressor complex. To test this hypothesis, the proteins bound to mSin3A immunecomplexes were eluted with ionic detergents and reprecipitated with affinity purified antibodies specific for an internal peptide of HDACl.
  • HDACl immunoprecip-itates contain several proteins that were specifically competed with the immunizing peptide ( Figure 1 IB).
  • Figure 1 IB A polypeptide doublet that comigrated with mSin3A was detected in the HDACl immunocomplexes ( Figure I IB and 11C).
  • Figure 11C To confirm that the doublet coprecipitating with HDACl is mSin3A, the HDACl immunocomplex was eluted and reprecipitated with antisemm specific for mSin3A ( Figure 11C). The two proteins in this precipitate comigrated with mSin3A, confirming that mSin3A and HDACl are associated in vivo.
  • the low-stringency mSin3A immunoprecipitation shown in Figure 1 IC also contained a protein that comigrated with RbAp48 (marked with an asterisk) that was not readily visible on the shorter exposures of low stringency mSin3 immunoprecipitations ( Figure 11 A).
  • RbAp48 in mSin3A immunoprecipitates from cell extracts of nontransfected cells by western blotting further demonstrating that mSin3A and RbAp48 associate in vivo ( Figure 12 A).
  • HDACl -F FLAG-epitope tagged HDACl
  • mSin3A-H histidine-tagged mSin3A
  • HDAC1-F was also precipitated by Ni 2+ -NTA agarose in a manner that was dependent on coexpression of mSin3A-H ( Figure 12B), demonstrating that a complex between HDAC 1 and mSin3A is formed in insect cells using exogenously expressed human proteins.
  • mSin3 As the primary candidate for the negative transcriptional function of the DNA binding transcription factor Mad (Ayer et al., 1995; Schreiber-Agus et al. 1995; Ayer et al., 1996).
  • Mad DNA binding transcription factor
  • the mSin3A compressor is part of a high molecular weight, multicomponent complex(es) that contains active histone deacetylase, thereby implicating histone deacetylation as a potential mechanism for mSin3A-mediated repression.
  • the MadN35GALVP16 chimera functioned as a repressor by a mechanism that was dependent on the binding of mSin3 A and that was sensitive to trapoxin.
  • the same mutations that inactivate MadN35GALVP16 as a transcriptional repressor i.e. Mad(Pro)N35GALVP16
  • Mad(Pro)N35GALVP16) also block interaction between Madl and mSin3A in vitro and Madl function in vivo. Therefore, it is likely that Mad:Max heterocomplexes repress transcription in a manner dependent on an mSin3A- associated histone deacetylase.
  • the heterogeneous nature of the mSin3 A complex potentially reflects a diverse array of repressors, histone deacetylases and different targeting molecules that facilitate mSin3A-dependent alterations in gene expression. At least five additional polypeptides are stably associated with mSin3A ( Figures
  • yeast mutation of acetyl-accepting lysines in histone H4 reduces the activity of the GALI, PHOS and CUPl promoters in vivo (Durrin et al., 1991).
  • the transcriptionally silent regions in yeast, HML and HMR, are hypoacetylated and their activation is correlated with acetylation of histone H4 (Braunstein et al uncomfortable 1993).
  • biochemical studies showed that certain transcription factors have higher affinity for their binding sites when those sites are embedded in chromatin assembled from hyperacetylated histones (Lee et al., 1993; Vettes-Dadey al., 1996).
  • the acetylalion status of a particular chromatin region represents a balance between competing, acetylation and deacetylation reactions.
  • MadN35GALVPI6 recmits mSin3A-HDAC complexes to specific sites on DNA and shifts this equilibrium towards deacetylation and subsequent transcription repression by creating a high effective molarity of the histone deacetylase.
  • deacetylase inhibitors can inhibit MyoD- (Johnston et al., 1992) and steroid receptor-activated transcription (McKnight et al., 1990; Bresnick et al., 1990). While it remains to be shown that the effects of RPD3 on silencing are direct, this evidence suggests that histone deacetylation can elicit both positive and negative effects on gene expression. Determining the factors diat govern the functional outcome of histone deacetylation will provide fertile ground for further experimentation.
  • Antibodies, cell culture, and Immunoprecipitations To generate antisemm specific for mSin3A a GST fusion protein encoding, amino acids 251 through 405 of mSin3A was used to immunize a New Zealand White rabbit. The cmde semm was passed over a GST column to remove the anti-GST antibodies. U937 cells were grown in RPMI supplemented with 10% calf semm (Hyclone), glutamine and penicillin- streptomycin. Low and high stringency immunoprecipitations were performed essentially as described (Ayer and Eisenman, 1993). To elute proteins from low stringency immunoprecipitates, they were incubated for 60 minutes at room temperature in antibody buffer and reprecipitated under high stringency conditions.
  • Luciferase assays 293 cells were seeded in triplicate onto 60 mm dishes at 3x10 s cells in 4 ml DME with 10% calf semm (Hyclone). Six hours after seeding, cells were transfected with 50 ng luciferase reporter, 50 ng CMV- ⁇ -gal, 50 ng expression constmct, and 2.85 ⁇ g carrier DNA using the BBS/CaPO method. 10 nM trapoxin was added to the media 8 hours prior to the luciferase assays. Cell lysates were prepared 20-24 hours following transfections, and luciferase and ⁇ -galactosidase activities was assayed according to manufacturer directions (Promega, Tropix). Luciferase values (relative light units) were normalized for transfection efficiency by dividing by ⁇ -gal activity.
  • Histone deacetylase assays In vitro histone deacetylase activity was assayed essentially as described with either 50 ⁇ l of cmde cell extract (approximately 5 X IO 6 Cells) or immunopurified cell extracts (approximately 2 X IO 7 cells) for 2.5 hrs at 37 C (Taunton et al., 1996b). Pretreatment of cmde or immunopurified extract with synthetic trapoxin was performed for 30 minutes at 4 C prior to addition of peptide substrate. TAg Jurkat and 293 cell extracts for histone deacetylase assays were prepared as in Taunton et al., 1996.
  • Anti-HDACl and anti-mSin3A immunoprecipitations were performed as described above and in Figure 11.
  • the Protein-A conjugated immunoprecipitates were washed three times in J-buffer plus I mM EDTA and resuspended in J-buffer without Triton-X-100, and histone deacetylase activity was measured as described.
  • Baculovimses cDNAs encoding Flag-tagged HDACl, HA-tagged RbAp48 and His-tagged mSin3A were cloned into the transfer vector pVL 1392 (specific details on the constmction of these vectors is available upon request). Recombinant vims was generated using Baculogold DNA according to the manufactures instmctions (Pharminigen). Sf9 or High 5 cells were infected at high multiplicity, extracts prepared 48 hours post infection and immunoprecipitations performed as described above. Ni 2+ - NTA agarose and anti-Flag antibody were purchased from Qiagen and Kodak-IBI, respectively.
  • Mad-Max transcriptional repression is mediated by ternary complex formation with mammalian homologs of yeast repressor Sin3. Cell 80, 767-776.
  • Transcriptional silencing in yeast is associated with reduced nucleosome acetylation.
  • Glucocorticoid receptor-dependent disruption of a specific nucleosome on the mouse mammary tumor vims promoter is prevented by sodium butyrate. Proc. Natl. Acad Sci.
  • Mad3 and Mad4 Novel Max-interacting transcriptional repressors that suppress c-Myc-dependent transformation and are expressed during, neural and epidermal differentiation. EMBO J 14, 5646-5659.
  • the transcriptional activators p3OO and CBP are histone acetylases- Cell 87, 953-959, Parthun, M. R., Windom, J., and Gottschling, D. E. (1996).
  • the major cytoplasmic histone acetyltransferase in yeast links to chromatin replication and histone metabolism Cell 87, 85-94.
  • HDA1 and RPD3 are members of distinct yeast histone deacetylase complexes that regulate silencing and transcription. Proc. Natl. Aca Sci. USA 93, 14503-14508.
  • Histone H4 isoforms acetylated at specific lysine residues define individual chromosomes and chromatin domains in Drosophila polytene nuclei. Cell 69, 375-84.
  • the p55 subunit of Drosophila chromatin assembly factor 1 is homologous to a histone deacetylase-associated protein. Mol. Cell. Biol. 16, 6149-6159,
  • RPD3 encodes a second factor required to achieve maximum positive and negative transcriptional states in Saccharomyces cerevisiae. Mol. Cell. Biol. 11, 6317-27.
  • Saccharomyces cerevisiae SIN3 gene a negative regulator of HO, contains four paired amphipathic helix motifs. Mol. Cell. Biol. 10, 5927-36.
  • each clone includes a v motiff represented in the general formula of SEQ LD No. 12, and a ⁇ motif represented in die general formula SEQ ID No. 14.
  • ADDRESSEE FOLEY, HOAG & ELIOT, LLP
  • TTCCAGCCGG TTATCAACCA GGTAGTGGAC TTCTACCAAC CCACGTGCAT TGTGCTCCAG 120 TGTGGAGCTG ACTCTCTGGG CTGTGATCGA TTGGGCTGCT TTAACCTCAG CATCCGAGGG 180
  • GAAGAGGCCA TTAGTGAGGA GCTTCCCTAT AGTGAATACT TCGAGTACTT TGCCCCAGAC 360

Abstract

The present invention concerns proteins encoded by a family of genes, termed here HDx-related genes, which are involved in the control of chromatin structure and, thus in transcription and translation. The present invention makes available compositions and methods that can be utilized, for example, to control cell proliferation and differentiation in vitro and $i(in vivo.)

Description

Histone Deacetylases, and Uses Related Thereto
Government Funding
Work described herein was supported in part by funding from the National Institute of Health. The United States Government has certain rights in the invention.
Background of the Invention
The organization of regulatory DNA elements into precise chromatin structures is important for both DNA replication and transcription in vivo (Lee et al. (1993) Cell 72:73-84; Felsenfeld (1992) Nature. 355:219). In eukaryotic cells, nuclear DNA exists as a hierarchy of chromatin structures, resulting in the compaction of nuclear DNA about 10,000 fold (Davie and Hendzel (1994) J. Cell. Biochem. 55:98). The repeating structural unit in the extended 10 nm fibre form of chromatin is the nucleosome (van Holde (1988) Chromatin. New York: Springer-Verlag). The nucleosome consists of 146 bp of DNA wrapped around a protein core of the histones H2A, H2B, H3, and H4, known as the core histones. These histones are arranged as an (H3-H4)2 tetramer and two H2A-H2B dimers positioned on each face of the tetramer. The DNA joining the nucleosomes is called linker DNA; it is to the linker DNA to which the HI or linker histones bind. The 10 nm fibre is compacted further into the 30 nm fibre. Linker histones and amino-terminal regions ("tails") of the core histones maintain the higher order folding of chromatin (Garcia Ramirez et al.. (1992) J. Biol Chem 267:19587). This chromatin structure must be relaxed when DNA is transcribed or translated.
Histones of the nucleosome core particle are subject to reversible acetylation at the ε-amino group of lysines present in their amino terminus (Csordas et al. (1990) Biochem J 265:23-38). Transcriptionally silent regions of the genome are enriched in underacetyiated histone H4 (Turner (1993) Cell 75:5-8), and histone hyperacetylation facilitates the ability of transcription factor TFHIA to bind to chromatin templates (Lee et al. (1993) Cell 72:73-84). Recent genetic, biochemical and immunological approaches have provided substantial evidence indicating that histones associated with actively transcribed genes are more highly acetylated than those from nontranscribed regions. While not wishing to be bound by any particular theory, histone acetylation may influence transcription at several stages, for example, by causing transcription factors to bind or by inducing structural transitions in chromatin, or by facilitating histone displacement and repositioning during polymerase elongation.
The acetylation and deacetylation are catalyzed by specific enzymes, histone acetyltransferase and deacetylase, respectively, and the net level of the acetylation is controlled by the equilibrium between these enzymes. The steady state level of acetylation and the rates at which acetate groups are turned over vary both between and within different cell types, with half-lives that vary from a few minutes to several hours. Although a histone acetyltransferase gene (HAT1) has been identified in yeast (KelfF et al. (1995) J. Biol. Chem. 270:24674-24677), the molecular entities responsible for histone deacetylation were heretofore unknown in the art.
The identification of the mechanism by which histones are deacetylated would be of great benefit in the control of gene transcription and the cell cycle.
Summary of the Invention The present invention relates to the discovery of a novel family of genes, and gene products, expressed in mammals, which genes are referred to hereinafter as the "histone deacetylase" genes or "HDx" gene family, the products of which are referred to as histone deacetylases or HDx proteins.
In genera], the invention features isolated HDx polypeptides, preferably substantially pure preparations of one or more of the subject HDx polypeptides. The invention also provides recombinantly produced HDx polypeptides. In preferred embodiments the polypeptide has a biological activity including an ability to deacetylate an acetylated histone substrate, preferably a substrate analog of histone H3 and/or H4. In other embodiments the HDx polypeptides of the present invention bind to trapoxin or to trichostatin, such binding resulting in the inhibition a deacetylase activity of the HDx polypeptide. However, HDx polypeptides which specifically antagonize such activities, such as may be provided by dominant negative mutants, are also specifically contemplated.
The HDx polypeptides disclosed herein are capable of modulating proliferation, survival and/or differentiation of cells, because of their ability to alter chromatin structure by deacetylating histones such as H3 or H4. Moreover, in preferred embodiments, the subject HDx proteins have the ability to modulate cell growth by influencing cell cycle progression or to modulate gene transcription. In one embodiment, the polypeptide is identical with or homologous to an HDx protein. Exemplary HDx polypeptide include amino acid sequences represented in any one of SEQ ID Nos 5-8. Related members of the HDx family are also contemplated, for instance, an HDx polypeptide preferably has an amino acid sequence at least 85% homologous to a polypeptide represented by one or more of the polypeptides designated SEQ ID Nos: 5-8, though polypeptides with higher sequence homologies of, for example, 88, 90% and 95% or are also contemplated. In one embodiment, the HDx polypeptide is encoded by a nucleic acid which hybridizes under stringent conditions with a nucleic acid sequence represented in one or more of SEQ ID Nos. 1-4. Homologs of the subject HDx proteins also include versions of the protein which are resistant to post-translation modification, as for example, due to mutations which alter modification sites (such as tyrosine, threonine, serine or aspargine residues), or which inactivate an enzymatic activity associated with the protein.
The HDx polypeptide can comprise a full length protein, such as represented in SEQ ID No. 5, or it can comprise a fragment corresponding to particular motifs/domains, or to arbitrary sizes, e.g., at least 5, 10, 25, 50, 100, 150 or 200 amino acids in length. In preferred embodiments, the polypeptide, or fragment thereof, specifically deacetylates histone H4. In other preferred embodiments, the HDx polypeptide includes both a v motif (SEQ ID No. 12) and a χ motif (SEQ ID No. 14), preferably a v motif represented in the general formula SEQ ID No. 13, and a χ motif represented in the general formula SEQ ID No. 15.
In certain preferred embodiments, the invention features a purified or recombinant HDx polypeptide having a molecular weight in the range of 40kd to 60kd. For instance, preferred HDx polypeptides, have molecular weights in the range of 50kd to about 60kd, even more preferably in the range of 53-58kd. It will be understood that certain post¬ translational modifications, e.g., phosphorylation, prenylation and the like, can increase the apparent molecular weight of the HDx protein relative to the unmodified polypeptide chain.
The subject proteins can also be provided as chimeric molecules, such as in the form of fusion proteins. For instance, die HDx protein can be provided as a recombinant fusion protein which includes a second polypeptide portion, e.g., a second polypeptide having an amino acid sequence unrelated (heterologous) to the HDx polypeptide, e.g. the second polypeptide portion is glutathione-S-transferase, e.g. the second polypeptide portion is an enzymatic activity such as alkaline phosphatase, e.g. the second polypeptide portion is an epitope tag. In yet another embodiment, the invention features a nucleic acid encoding a an HDx polypeptide, or polypeptide homologous thereto, which polypeptide has the ability to modulate, e.g., either mimic or antagonize, at least a portion of the activity of a wild¬ type HDx polypeptide. Exemplary HDx-encoding nucleic acid sequences are represented by SEQ ID Nos: 1-4.
In another embodiment, the nucleic acid of the present invention includes a coding sequence which hybridizes under stringent conditions with one or more of the nucleic acid sequences in SEQ ID Nos: 1-4. The coding sequence of the nucleic acid can comprise a sequence which is identical to a coding sequence represented in one of SEQ ID Nos: 1-4, or it can merely be homologous to one or more of those sequences. In preferred embodiments, the nucleic acid encodes a polypeptide which specifically modulates, by acting as either an agonist or antagonist, the enzymatic activity of an HDx polypeptide.
Furthermore, in certain preferred embodiments, the subject HDx nucleic acid will include a transcriptional regulatory sequence, e.g. at least one of a transcriptional promoter or transcriptional enhancer sequence, which regulatory sequence is operably linked to the HDx gene sequence. Such regulatory sequences can be used in to render the HDx gene sequence suitable for use as an expression vector. This invention also contemplates the cells transfected with said expression vector whether prokaryotic or eukaryotic and a method for producing HDx proteins by employing said expression vectors.
In yet another embodiment, the nucleic acid hybridizes under stringent conditions to a nucleic acid probe corresponding to at least 12 consecutive nucleotides of either sense or antisense sequence of one or more of SEQ ID Nos: 1-4; though preferably to at least 25 consecutive nucleotides; and more preferably to at least 40, 50 or 75 consecutive nucleotides of either sense or antisense sequence of one or more of SEQ ID Nos: 1-4.
Yet another aspect of the present invention concerns an immunogen comprising an HDx polypeptide in an immunogenic preparation, the immunogen being capable of eliciting an immune response specific for an HDx polypeptide; e.g. a humoral response, e.g. an antibody response; e.g. a cellular response. In preferred embodiments, the immunogen comprising an antigenic determinant, e.g. a unique determinant, from a protein represented by one of SEQ ID Nos. 5-8.
A still further aspect of the present invention features antibodies and antibody preparations specifically reactive with an epitope of the HDx immunogen. The invention also features transgenic non-human animals, e.g. mice, rats, rabbits, chickens, frogs or pigs, having a transgene, e.g., animals which include (and preferably express) a heterologous form of an HDx gene described herein, or which misexpress an endogenous HDx gene, e.g., an animal in which expression of one or more of the subject HDx proteins is disrupted. Such a transgenic animal can serve as an animal model for studying cellular and tissue disorders comprising mutated or mis-expressed HDx alleles or for use in drug screening.
The invention also provides a probe/primer comprising a substantially purified oligonucleotide, wherein the oligonucleotide comprises a region of nucleotide sequence which hybridizes under stringent conditions to at least 12 consecutive nucleotides of sense or antisense sequence of SEQ DD Nos: 1-4, or naturally occurring mutants thereof. Nucleic acid probes which are specific for each of the HDx proteins are contemplated by the present invention, e.g. probes which can discern between nucleic acid encoding a human or bovine HD. In preferred embodiments, the probe/primer further includes a label group attached thereto and able to be detected. The label group can be selected, e.g., from a group consisting of radioisotopes, fluorescent compounds, enzymes, and enzyme co-factors. Probes of the invention can be used as a part of a diagnostic test kit for identifying dysfunctions associated with mis-expression of an HDx protein, such as for detecting in a sample of cells isolated from a patient, a level of a nucleic acid encoding a subject HDx protein; e.g. measuring an HDx mRNA level in a cell, or determining whether a genomic HDx gene has been mutated or deleted. These so called "probes/primers" of the invention can also be used as a part of "antisense" therapy which refers to administration or in situ generation of oligonucleotide probes or their derivatives which specifically hybridize (e.g. bind) under cellular conditions, with the cellular mRNA and or genomic DNA encoding one or more of the subject HDx proteins so as to inhibit expression of that protein, e.g. by inhibiting transcription and/or translation. Preferably, die oligonucleotide is at least 12 nucleotides in length, though primers of 25, 40, 50, or 75 nucleotides in length are also contemplated.
In yet another aspect, the invention provides an assay for screening test compounds for inhibitors, or alternatively, potentiators, of an interaction between an HDx protein and an HDx binding protein or nucleic acid sequence. An exemplary method includes the steps of (i) combining an HDx polypeptide or fragment thereof, one or more HDx target polypeptide (such as a histone, SIN3, RpAp48 or other protein which participates in HDx complexes, e.g., one or more proteins having molecular weights of 250 kDa, 180 kDa, 55 kDa, 50 kDa, 42 kDa, 33-36 kDa and 30 kDa, see also Example 3), and a test compound, e.g., under conditions wherein, but for the test compound, the HDx protein and target polypeptide(s) are able to interact; and (ii) detecting the formation of a complex which includes the HDx protein and target polypeptide(s) either by directly quantitating the complex, the deacetylase activity of the HDx protein, or by measuring inductive effects of the HDx protein. A statistically significant change, such as a decrease, in the formation of the complex in the presence of a test compound (relative to what is seen in the absence of the test compound) is indicative of a modulation, e.g., inhibition, of the interaction between the HDx protein and its target polypeptide. Furthermore, the present invention contemplates the use of other homologs of the
HDx polypeptides or bioactive fragments thereof to generate similar assay formats. In one embodiment, the drug screening assay can be derived with a fungal homolog of an HDx protein, such as RPD3, in order to identify agents which inhibit histone deacetylation in a yeast cell. Yet another aspect of the present invention concerns a method for modulating one or more of growth, differentiation, or survival of a mammalian cell by modulating HDx bioactivity, e.g., by inhibiting the deacetylase activity of HDx proteins, or disrupting certain protein-protein interactions. In general, whether carried out in vivo, in vitro, or in situ, the method comprises treating the cell with an effective amount of an HDx therapeutic so as to alter, relative to the cell in the absence of treatment, at least one of (i) rate of growth, (ii) differentiation, or (iii) survival of the cell. Accordingly, the method can be carried out with HDx therapeutics such as peptide and peptidomimetics or other molecules identified in the above-referenced drug screens which antagonize the effects of a naturally-occurring HDx protein on said cell. Other HDx therapeutics include antisense constructs for inhibiting expression of HDx proteins, and dominant negative mutants of HDx proteins which competitively inhibit protein-substrate and/or protein- protein interactions upstream and downstream of the wild-type HDx protein.
In an exemplary embodiment the subject method is used to treat tumor cells by antagonizing HDx activity and blocking cell cycle progression. In one embodiment, the subject method includes the treatment of testicular cells, so as modulate spermatogenesis. In another embodiment, the subject method is used to modulate osteogenesis, comprising the treatment of osteogenic cells with an HDx polypeptide. Likewise, where the treated cell is a chondrogenic cell, the present method is used to modulate chondrogenesis. In still another embodiment, HDx polypeptides can be used to modulate the differentiation of progenitor cells, e.g., the method can be used to cause differentiation of a hematopoietic cells, neuronal cells, or other stem/progenitor cell populations, to maintain a that cell in a differentiated state, and/or to enhance the survival of a differentiated cell, e.g., to prevent apoptosis or other forms of cell death.
In addition to such HDx therapeutic uses, anti-fungal agents developed with such screening assays as described herein can be used, for example, as preservatives in foodstuff, feed supplement for promoting weight gain in livestock, or in disinfectant formulations for treatment of non-living matter, e.g., for decontaminating hospital equipment and rooms. In similar fashion, assays provided herein will permit selection of deacetylase inhibitors which discriminate between the human and insect deacetylase enzymes. Accordingly, d e present invention expressly contemplates the use and formulations of the deacetylase inhibitors in insecticides, such as for use in management of insects like the fruit fly. Moreover, certain of the inhibitors can be selected on the basis of inhibitory specificity for plant HΩx-related activities relative to the mammalian enzymes. Thus, the present invention specifically contemplates formulations of deacetylase inhibitors for agricultural applications, such as in the form of a defoliant or the like.
The present method is applicable, for example, to cell culture technique, such as in the culturing of hematopoietic cells and other cells whose survival or differentiative state is dependent on HDx function. Moreover, HDx agonists and antagonists can be used for therapeutic intervention, such as to enhance survival and maintenance of cells, as well as to influence organogenic pathways, such as tissue patterning and other differentiation processes. In an exemplary embodiment, the method is practiced for modulating, in an animal, cell growth, cell differentiation or cell survival, and comprises administering a therapeutically effective amount of an HDx polypeptide to alter, relative the absence of HDx treatment, at least one of (i) rate of growth, (ii) differentiation, or (iii) survival of one or more cell-types in the animal.
Another aspect of the present invention provides a method of determining if a subject, e.g. a human patient, is at risk for a disorder characterized by unwanted cell proliferation or aberrant control of differentiation. The method includes detecting, in a tissue of the subject, the presence or absence of a genetic lesion characterized by at least one of (i) a mutation of a gene encoding an HDx protein, e.g. represented in one of SEQ ID Nos: 1-4, or a homolog thereof; or (ii) the mis-expression of an HDx gene. In preferred embodiments, detecting the genetic lesion includes ascertaining the existence of at least one of: a deletion of one or more nucleotides from an HDx gene; an addition of one or more nucleotides to the gene, a substitution of one or more nucleotides of the gene, a gross chromosomal rearrangement of the gene; an alteration in the level of a messenger RNA transcript of the gene; the presence of a non- wild type splicing pattern of a messenger RNA transcript of the gene; or a non-wild type level of the protein.
For example, detecting the genetic lesion can include (i) providing a probe/primer including an oligonucleotide containing a region of nucleotide sequence which hybridizes to a sense or antisense sequence of an HDx gene, e.g. a nucleic acid represented in one of SEQ ID Nos: 1-4, or naturally occurring mutants thereof, or 5' or 3' flanking sequences naturally associated with the HDx gene; (ii) exposing the probe primer to nucleic acid of the tissue; and (iii) detecting, by hybridization of the probe/primer to the nucleic acid, the presence or absence of the genetic lesion; e.g. wherein detecting the lesion comprises utilizing the probe/primer to determine the nucleotide sequence of the HDx gene and, optionally, of the flanking nucleic acid sequences. For instance, the probe/primer can be employed in a polymerase chain reaction (PCR) or in a ligation chain reaction (LCR). In alternate embodiments, d e level of an HDx protein is detected in an immunoassay using an antibody which is specifically immunoreactive with the HDx protein.
In another aspect, the invention provides compounds useful for inhibition of HDxs. In a preferred embodiment, an HDx inhibitor compound of the invention can be represented by the formula A-B-C, in which A is a specificity element for selective binding to an HDx, B is a linker element, and C is an electrophilic moiety capable of reacting with a nucleophilic moiety of an HDx; with the proviso that the compound is not butyrate, trapoxin, or trichostatin.
For instance, in one embodiment, there is provided a composition for inhibiting a histone deacetylase comprising a compound represented by the general formula A-B-C, wherein A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclopeptides;
B is selected from d e group consisting of substituted and unsubstituted C4-C8 alkylidenes, C -Cg alkenylidenes, C -C8 alkynylidenes, and -(D-E-F)-, in which D and F are, independently, absent or represent a C2-C7 alkylidene, a C2-C7 alkenylidene or a C2- C7 alkynylidene, and E represents O, S, or NR', in which R' represents H, a lower alkyl, a lower alkenyl, a lower alkynyl, an aralkyl, aryl, or a heterocyclyl; and
C is selected from the group consisting of
Figure imgf000011_0001
, and a boronic acid; in which Z represents O, S, or NR5, and Y; R5 represents a hydrogen, an alkyl, an alkoxycarbonyl, an aryloxycarbonyl, an alkylsulfonyl, an arylsulfonyl or an aryl; R'6 represents hydrogen, an alkyl, an alkenyl, an alkynyl or an aryl; and R represents a hydrogen, an alkyl, an aryl, an alkoxy, an aryloxy, an amino, a hydroxylamino, an alkoxylamino or a halogen; with the proviso that the compound is not trapoxin.
In another preferred embodiment, the compound represented by the general formula A-B-C, wherein
A is selected from the group consisting of cycloalk ls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclopeptides;
B is selected from the group consisting of substituted and unsubstituted C -C8 alkylidenes, C -C8 alkenylidenes, C4-C8 alkynylidenes, and -(D-E-F)-, in which D and F are, independently, absent or represent C2-C alkylidenes, C2-C7 alkenylidenes or C2-C7 alkynylidenes, and E represents O, S, or NR', in which R' represents H, a lower alkyl, a lower alkenyl, a lower alkynyl, an aralkyl, an aryl, or a heterocyclyl; and
C is selected from the group consisting of
Figure imgf000011_0002
, in which R9 represents a hydrogen, an alkyl, an aryl, a hydroxyl, an alkoxy, an aryloxy or an amino, with the proviso that the inhibitor compound is not trichostatin. In still another preferred embodiment, the compound is represented by the general formula A-B-C, wherein
A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclopeptides;
B is selected from the group consisting of substituted and unsubstituted C -C8 alkylidenes, C -C8 alkenylidenes, C -C alkynylidenes, and -(D-E-F)-, in which D and F are, independently, absent or a C2-C7 alkylidene, a C2-C alkenylidene, or a C2-C7 alkynylidene, and E represents O, S, or NR', in which R' is H, lower alkyl, lower alkenyl, lower alkynyl, aralkyl, aryl, or heterocyclyl; and
Figure imgf000012_0001
C represents ; in which Y is O or S, and R7 represents a hydrogen, an alkyl, an aryl, an alkoxy, an aryloxy, an amino, a hydroxylamino, an alkoxylamino or a halogen.
The present invention also contemplates pharmaceutical preparations of such compounds, e.g., in an amount effective for inhibiting proliferation of a cell, formulated in a pharmaceutically acceptable diluent.
Moreover, such compounds can be used for modulating one or more of growth, differentiation, or survival of a mammalian cell responsive to HDx-mediated histone deacetylation, by treating the cell with an effective amount of the deacetylase inhibitor so as to modulate the deacetylase activity and alter, relative to the cell in the absence of the agent, at least one of (i) the rate of growth, (ii) the differentiation state, or (iii) the rate of survival of the cell.
The practice of the present invention will employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See, for example, Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989); DNA Clomng, Volumes I and II (D. N. Glover ed., 1985); Oligonucleotide Synthesis (M. J. Gait ed., 1984); Mullis et al. U.S. Patent No: 4,683,195; Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins eds. 1984); Transcription And Translation (B. D. Hames & S. J. Higgins eds. 1984); Culture Of Animal Cells (R. I. Freshney, Alan R. Liss, Inc., 1987); Immobilized Cells And Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide To Molecular Cloning (1984); the treatise, Methods In Enzymology (Academic Press, Inc., N.Y.); Gene Transfer Vectors For Mammalian Cells (J. H. Miller and M. P. Calos eds., 1987, Cold Spring Harbor Laboratory); Methods In Enzymology, Vols. 154 and 155 (Wu et al. eds.), Immunochemical Methods In Cell And Molecular Biology (Mayer and Walker, eds., Academic Press, London, 1987); Handbook Of Experimental Immunology, Volumes I- IV (D. M. Weir and C. C. Blackwell, eds., 1986); Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986). Other features and advantages of the invention will be apparent from the following detailed description, and from the claims.
Brief Description of the Drawings Figure IA illustrates the chemical structures of trapoxin and trichostatin, natural products diat inhibit the enzymatic deacetylation of lysine residues near the NH2-terminus of histones. The epoxyketone side chain of trapoxin is approximately isosteric with N- acetyl lysine and likely alkylates an active site nucleophile.
Figure IB illustrates the copurification of trapoxin binding and histone deacetylase activities. Nuclear proteins from bovine thymus were precipitated with ammonium sulfate and fractionated on a Mono Q column. Trapoxin binding was assayed by charcoal precipitation with [3H]trapoxin. For the histone deacetylase assay, a peptide corresponding to bovine histone H4 (1-24) was synthesized. The peptide was chemically acetylated with sodium [3H]acetate (5.3 Ci/mmol, New England Nuclear) BOP reagent (Aldrich) and purified by reverse phase HPLC. Two microliters of
[3H]peptide(~40,000dpm) were used per 200 μl assay. After incubation at 37°C for one hour, the reaction was quenched with 1 M HC1/0.16 M acetic acid (50 μl). Released
[3Hjacetic acid was extracted with 600 μl of ethyl acetate and quantified by scintillation counting. Pretreatment of crude or partially purified enzyme with trapoxin or trichostatin (20nM) for 30 min. at 4°C abolished deacetylase activity. A28o= absorbance at 280 nm.
Figure 2A shows the synthesis of K-trap and the K-trap affinity matrix. K-trap contains a protected lysine residue in place of the phenylalanine at position two in trapoxin. Alloc = allyloxycarbonyl.
Figure 2B is a silver stained gel showing bovine and human trapoxin binding proteins. Proteins bound to the K-trap affinity matrix in the presence or absence of trapoxin or trichostatin were eluted by boiling in SDS loading buffer and analyzed by SDS-PAGE (9% gel). Nuclear proteins from human Jurkat T cells were prepared identically to those from bovine thymus (Figure IB). Molecular size standards (in kilodaltons) are indicated to the right. Figure 3 A is the predicted amino acid sequence of human HD An in-frame stop codon was found upstream of the starting methionine. Regions equivalent to microsequenced tryptic peptides from the purified bovine protein are boxed. Underlined amino acids 319-334 and 467-482 denote the sequences of synthetic peptides that were conjugated to KLH and used to generate polyclonal antisera. Abbreviations for the amino acid residues are: A, Ala; C, Cys; D, Asp; E, Glu; F, Phe; G, Gly; H, His; I, He; K, Lys; L, Leu; M, Met; N, Asn; P, Pro; Q, Gin; R, Arg; S, Ser; T, Thr; V, Val; W, Trp; and Y, Tyr.
Figure 3B is a protein immunoblot analogous to d e silver stained gel in Figure 2B, showing the relationship between bovine p46-p49 and human p55 (top panels) and confirming the identity of p50 (bovine and human) as RbAp48 (bottom panels). Proteins eluted from the K-trap affinity matrix (Figure 2) were separated by SDS-PAGE and transferred to Immobilon-P (Millipore). Blots were probed with polyclonal anti-HDl (319-336) or monoclonal anti-RbAp48 and bound antibodies were detected with enhanced chemilurninescence (Amersham).
Figure 4A is an immunoprecipitation of endogenous histone deacetylase activity with affinity purified anti-HDl (467-482) antibodies. Anti-HDl (467-482) immunoprecipitates from equivalent amounts of Jurkat nuclear extract (1 mg nuclear protein supplemented with 0.5 M NaCl, 1% BSA, and 0.1% NP-40) were isolated in the presence or absence of HD 1(467-482) peptide competitor. After resuspending the immunoprecipitates in HDx buffer [20 mM tris (pΗ 8), 150 mM NaCl, 10% glycerol], inhibitors were added as indicated, and histone deacetylase activity was measured as described in Figure 1 A.
Figure 4B shows the coprecipitation of HD1 and RbAp48, as detected by protein immunoblot analysis.
Figure 4C demonstrates the histone deacetylase activity of recombinant HD1-F. Tag Jurkat cells (Clipstone et al. (1992) Nature 357, 695-7) were transfected with pFJ5 (vector alone) or pBJ5/HDl-F (encoding COOΗ-terminal FLAG epitope tagged HD1) by electroporation and detergent lysates were prepared [0.5% Triton X-100, 50 mM tris (pΗ 8), 100 mM NaCl, 10% glycerol]. Anti-FLAG antibodies conjugated to agarose beads (IBI) were used to immunoprecipitate recombinant HD1 in the presence or absence of FLAG peptide competitor, and histone deacetylase activity was measured as described above.
Figure 4D shows die interaction between recombinant ΗD1-F and the K-trap affinity matrix. Lysates from Jurkat cells transfected with pBJ5/ΗDl-F were incubated with the K-trap affinity matrix in the presence or absence of inhibitors. Immunoblots of the eluted proteins were probed with the anti-FLAG M2 monoclonal antibody (IBI).
Figures 5 A and 5B are sequence alignments for various HDx and HDx-related cDNAs and proteins, respectively. Figure 6 depicts exemplary specificity elements (A), linker elements (B), and electrophilic moieties (C) for generating compounds which are capable of reacting with a nucleophilic moiety of an HDx protein.
Figure 7 illustrates an exemplary synthesis of trichostatin analogs. Figures 8A-8C illustrate a synthesis of tritiated Trapoxin B.
Figures 9A-9C depict a synthesis of the K-trap and K-trap affinity matrix .
Figures 10A-10B: mSin3A is present in cells as a large stable multiprotein complex. Nuclear lysates were prepared from U937 cells metabolically labeled with [35]S-methionine and low stringency immunoprecipitations performed with antiserum specific for mSin3A. "+ block" shows proteins immunoprecipitated when the anti- mSin3 A was preincubated with purified GST-PAΗ2 (A). In (B), low stringency mSin3A immunoprecipitates were washed for an additional 60 minutes using the salt and detergent conditions indicated at the top of die Figure. In (A) and (B), the immunoprecipitates were analyzed by SDS-PAGE and autoradiography. Apparent molecular weight of the coprecipitating proteins and the sizes of die molecular weight markers are given in kilodaltons.
Figures 11 A-D: mSin3A and EMACI associate in vivo. Immunoprecipitations were performed using nuclear extracts from [35]S-methionine labeled U937 cells. (A) The left lane shows proteins from an anti-mSin3A immunoprecipitate. The right lane shows proteins eluted from an anti-mSin3A immunoprecipitate and reprecipitated with anti¬ serum specific for HD1. In (B) and (C), low stringency immunoprecipitations were performed using antiserum specific for the carboxy-terminus of HD1. "+ block" indicates diat the HD1 antiserum was preincubated with the immunizing peptide. In (C), proteins immunoprecipitated with anti-mS3A are shown for reference, proteiηs eluted from a low stringency anti-HDl immunoprecipitate and reprecipitated with anti-mSin3A are shown in die right most lane. In (A), (B), and (C), autoradiographs of SDS-PAGE gels are shown. Apparent molecular weight of the coprecipitating, proteins and die sizes of the molecular weight markers are given in kilodaltons. In (D), in vitro histone deacetylase activity in anti-mSin3A immunoprecipitates is shown. Human Jurkat cell extracts (12 mg) were immunoprecipitated using anti-mSin3A polyclonal antibodies, "+block" indicates that the anti-mSin3A antibody was preincubated with GST-PAH2, "+10 nM trapoxin" indicates diat the immunoprecipitated proteins were pretreated with 10 nM trapoxin for 30 minutes at 4°C prior to being assayed for histone deacetylase activity. Figures 12 A-C: RbAp48 is associated with mSin3A in vivo and recombinant HDl, RbAp48 and mSin3A copurify from insect cell extracts. (A) TAg Jurkat cell lysates were immunoprecipitated using antibodies specific to C-terminalal portion of HDl (left) or antibodies specific to PAH2 of mSin3A (right). Parallel immunoprecipitations were blocked as described in figure 11. Immunopurified proteins were analyzed by SDS- PAGE and immunoblotted with (x-RbAp48 monoclonal antibody 12B1. (B & C) Equal amounts of baculovirus coinfected Sf9 cell extracts were affinity purified using Ni2+- agarose "Ni" or or x-FLAG-M2-agarose "F". Purified recombinant proteins were analyzed by SDS-PAGE, transferred to Immobilon-P (Millipore) and immunoblotted with FLAG to detect HD1-F (B) or (x-Flu (12CA.5) to detect p48-HA (C). We observe a reduction in expression of HDI-F and p48-HA when coexpressed with mSin3A.
Figures 13A-C: Trapoxin reverses transcriptional repression by mSin3A. (A) The structure of the minimal reporter gene derived from the myelomonocytic growth factor gene and the expression vectors. Mad(Pro)N35GALVPI6 has leucine at position 12 and alanine at position 16 mutated to proline as indicated. These point mutations prevent association between mSin3A and Mad (Ayer et al., 1995). The transcriptional activity of MadN35GALVP16 and Mad(Pro)N35GALVP16 was determined by measuring luciferase activity (Relative Light Units, RLU) of transfected 293 cells following an 8 hour treatment with 0 (solid bars) or 10 nM trapoxin (striped bars) (B). To control for differences in transfection efficiency, the RLU values were normalized to the β-galactosidase activity produced by a cotransfected CMV-βGAL construct. Shown is data from representative experiment and the error is reported as die standard error of the mean (s.e.m). This experiment has been done a minimum of five times in triplicate with similar results. An 8 hour treatment of 293 cells with 10 nM trapoxin is witiiin the linear range of the response of the reporter gene. Furthermore, trapoxin treatment did not prevent association between mSin3A and HDl (data not shown). (C), trapoxin inhibits histone deacetylase activity of human 293 cells in vivo. 2 x IO8 cells were cultured for 8 hours in the absence, "O", or in the presence of 10 nM trapoxin. Cells were harvested and crude extracts from approximately 1 x IO7 cells (solid bars) or anti- HDl immunoprecipitations of extracts from approximately 4 x IO7 cells (white bars) were assayed for histone deacetylase activity in vitro. Detailed Description of the Invention
The positioning of nucleosomes relative to particular regulatory elements in genomic DNA has emerged as a mechanism for managing the association of sequence- specific DNA-binding proteins with promoters, enhancers and odier transcriptional regulatory sequences. Two modifications to nucleosomes have been observed to influence the association of DNA-binding proteins with chromatin. Depletion of histones H2A/H2B from the nucleosome facilitates the binding of RNA polymerase II (Baer et al. (1983) Nature 301:482-488) and TFIHA (Hayes et al. (1992) PNAS 89:1229-1233). Likewise, acetylation of die core histones apparently destabilizes the nucleosome and is thought to modulate the accessibility of transcription factors to their respective enhancer and promoter elements (Oliva et al. (1990) Nuc Acid Res 18:2739-2747; and Walker et al. (1990) J Biol Chem 265:5622-5746). In both cases, overall histone-DNA contacts are altered.
In one aspect, die present invention concerns die discovery of a family of genes in mammals, the gene products of which are referred to herein as "histone deacetylases" or "HDxV. Experimental evidence indicates a functional role for the HDx gene products as catalysts of the deacetylation of histones in mammalian cells, and accordingly play a role in determining tissue fate and maintenance. For instance, the results provided below indicate that proteins encoded by the HDx genes may participate, under various circumstances, in the control of proliferation, differentiation and cell death.
The family of HDx gene apparently encode at least three different sub-families, e.g., paralogs, and have been identified from the cells of various mammals. The HDx proteins were first isolated from bovine thymus nuclei by use of a binding assay which exploited the ability of trapoxin, an inhibitor of histone deacetylase activity, to isolate proteins which co-purified widi a histone acetylase activity. The partial identity of the isolated proteins were determined by peptide microsequencing, and primers based on the peptide sequences were used to clone human cDNAs from a T cell library. One of the HDx gene products described below is referred to herein as HDl, and is represented in SEQ ID No. 1 (nucleotide) and SEQ ID No. 5 (amino acid). A search of expressed sequence tag (EST) libraries turned up partial sequences for human HDx transcripts, and revealed the existence of at least two other human HDx genes related to HDl, these other paralogs referred to herein as HD2 and HD3. Nucleotide and amino acid sequences for partial clones of other human HDx homologs are provided by SEQ LD Nos. 2-4 and 6-8, respectively. Analysis of die HDx sequences indicated no obvious similarities with any previously identified domains or motifs. However, the fact that each full-length clone lacks a signal sequence, along with the observation that proteins can be detected in the nucleus, indicates that die HDx genes encode intracellular proteins. Careful inspection of the HDx clones suggests at least two novel motifs, one or both of which may be characteristic of at least subfamilies of the mammalian HDx family. The first apparently conserved structural element of the HDx family occurs in the N- terminal portion of the molecule, and is designated herein as the "v motif'. With reference to human HDl, d e v motif corresponds to amino acid residues Asp 130- Phe 198. By alignment of the human HDx sequences, the element is represented by the consensus sequence:
DXXX TXXC<ΪLHHAKKXEASGFCy^^ VMTXSF, (SEQ ID No. 12) more preferably by the consensus sequence: DDVX1iNWAGGLHHAKKX2EASGFCY t roiV?C3X4 7TD-RVMTVSF (SEQ ID No. 13) wherein each of Xn represents any single amino acid, though more preferably represents an amino acid residue in the corresponding human HDx sequences of the appended sequence listing. A second motif, herein designated the χ motif is represented by die consensus sequence:
CVXXXKXPXXPXXXXGGGGYTXRNVARXWXXET (SEQ ID No. 14) more preferably by the consensus sequence:
C VEX8VKX9F X10PLLXi ιLGGGGYT 12RlWARCWTYET (SEQ ID No: 15) wherein each of Xn represents any single amino acid, diough more preferably represents an amino acid residue in the corresponding human HDx sequences of the appended sequence listing. The χ motif can be found in the human HDl sequence at C284-Thr316
The family of HDx proteins apparently ranges in size from about 40kd to about
60kd for the unmodified polypeptide chain. For instance, the bovine HD7 protein migrates on an SDS-PAGE (9%) gel with an apparent molecular weight of 46kD. The human HDl amino acid sequence predicts a molecular weight for the polypeptide chain of 55kD. Accordingly, certain aspects of the present invention relate to nucleic acids encoding HDx proteins, the HDx proteins themselves, antibodies immunoreactive with HDx proteins, and preparations of such compositions. Moreover, the present invention provides diagnostic and therapeutic assays and reagents for detecting and treating disorders involving, for example, aberrant expression (or loss thereof) of HDx homologs. In addition, drug discovery assays are provided for identifying agents which can modulate the biological function of HDx proteins, such as by altering the binding of HDx molecules to either proteins or nucleic acids. Such agents can be useful therapeutically to alter the growth and/or differentiation of a cell. Other aspects of the invention are described below or will be apparent to those skilled in the art in light of the present disclosure.
Analysis of die human HDx sequences, while not revealing any obvious similarities to known domains or motifs, did indicate similarities with previously identified proteins from both Saccharomyces cerevisiae and Xenopus laevis. Those genes, RPD3 (SEQ ID No. 9) and Xe-RPD3 (SEQ ID No. 10), respectively, had not previously been ascribed any specific function. However, based on our observations for the function of HDl, it is now apparent that each of these other proteins are also deacetylases, and represent potential therapeutic targets. Accordingly, drug discovery assays are provided for identifying agents which can modulate the biological function of "HDx-related" proteins, such as RPD3 homologs, by altering the enzymatic activity of the deacetylase, or its binding to odier cellular components including homologs of RbAp48 (described infra). Such agents can be useful therapeutically to alter the growth and/or differentiation of non-human cells, such as in the treatment of mycotic infections, or as additives to livestock feed, e.g., to promote weight gain, or as topical antiseptics for sterilizing medical equipment. In addition we isolated another bovine protein having an approximate molecular size of 50kD which apparently binds HDx proteins isolated by the trapoxin matrix, and microsequencing of that protein demonstrated that it was related to d e protein referred to in the art as RbAp48 (Qian et al. (1993) Nature 364:648; SEQ ID No. 11). RbAp48 was originally identified as a protein that binds to the retinoblastoma (Rb) gene product. The retinoblastoma (RB) gene product plays a role in tumor suppression (Weinberg, R.A., (Sept 1988) Scientific Amer.pp 44-51; Hansen et al. (1988) Trends Genet 4:125- 128). The role of RB as a tumor-suppressor protein in cell-cycle control is believed to be similar to that of another tumor-suppressor, p53 (Green (1989) Cell 56:1-3; Mowat et al (1985 Nature 314:633-636). Inactivation or mutation of the second RB allele in one of the somatic cells of these susceptible individuals appears to be the molecular event that leads to tumor formation (Caveneee et al. (1983) Nature 305:799-784; Friend et al. (1987) PNAS 84:9059-9063).
The growth suppression function of die retinoblastoma protein is thought to be mediated by Rb binding to cellular proteins. RbAp48 is one of the major proteins that binds to a putative functional domain at the carboxy terminus of the Rb protein. Complex formation between RbAp48 and Rb occurs in vitro and in vivo, and apparently involves direct interaction between the proteins. Like Rb, RbAp48 is a ubiquitously expressed nuclear protein. RbAp48 share sequence homology with MS11, a negative regulator of the Ras-cyclic AMP pathway in the yeast Saccharomyces cerevisiae. Furthermore, like MS11, human RbAp48 suppresses the heat-shock sensitivity of the yeast iral strains and RAS2Vall9 strains. Interaction with RbAp48 may be one of the mechanisms for suppression of growth mediated by Rb. Accordingly, d e interaction of RbAp48 with HDx proteins further implicates the HDx proteins in cell-cycle regulation.
The RpAp48 interaction with HDx and HDx-related proteins represents yet another therapeutic target. Accordingly, drug discovery assays are provided for identifying agents which can modulate the interaction of RbAp48 proteins and the like with HDx-related proteins. Such assays can be derived to detect the ability of a test agent to alter protein-protein contacts, or to alter the enzymatic activity of the deacetylase in complexes including an RbAp48 protein (e.g., were such complexes allosterically modulate the HDx enzymatic activity). As above, such agents can be useful therapeutically to alter the growth and/or differentiation of cells.
Members of the Mad family of BΗLΗZip proteins heterodimerize with Max to repress transcription in a sequence-specific manner. Transcriptional repression by Mad:Max heterodimers is mediated by ternary complex formation with either of the corepressors mSin3A or mSin3B. Example 3 demonstrates that Sin3 proteins are an in vivo component of large, heterogeneous multiprotein complexes and is tightly and specifically associated with at least seven polypeptides. Two of the Sin3-associated proteins, p50 and p55, are members of the histone deacetylase family described herein. Sin3 immunecomplexes possess histone deacetylase activity that is sensitive to the specific deacetylase inhibitor trapoxin. Sin3 targeted repression of a reporter gene is reduced by trapoxin treatment, suggesting that histone deacetylation mediates transcriptional repression through Mad-Max-Sin3 A multimeric complexes.
The Sin3 interaction with HDx and HDx-related proteins represents still another therapeutic target. Thus, in one aspect of the present invention there is provided drug discovery assays for identifying agents which can modulate the interaction of Sin3 proteins and the like with HDx-related proteins. Such assays can be derived to detect the ability of a test agent to alter protein-protein contacts, or to alter the enzymatic activity of the deacetylase in complexes including Sin3 or other transcriptional regulatory proteins. As above, such agents can be useful therapeutically to alter the growth and/or differentiation of cells.
For convenience, certain terms employed in the specification, examples, and appended claims are collected here.
As used herein, the term "nucleic acid" refers to polynucleotides such as deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA). The term should also be understood to include, as equivalents, analogs of either RNA or
DNA made from nucleotide analogs, and, as applicable to the embodiment being described, single (sense or antisense) and double-stranded polynucleotides.
As used herein, the term "gene" or "recombinant gene" refers to a nucleic acid comprising an open reading frame encoding one of the HDx polypeptides of the present invention, including both exon and (optionally) intron sequences. A "recombinant gene" refers to nucleic acid encoding an HDx polypeptide and comprising HDx-encoding exon sequences, though it may optionally include intron sequences which are either derived from a chromosomal HDx gene or from an unrelated chromosomal gene. Exemplary recombinant genes encoding the subject HDx polypeptide are represented in the appended Sequence Listing. The term "intron" refers to a DNA sequence present in a given HDx gene which is not translated into protein and is generally found between exons.
As used herein, the term "transfection" means the introduction of a nucleic acid, e.g., an expression vector, into a recipient cell by nucleic acid-mediated gene transfer. "Transformation", as used herein, refers to a process in which a cell's genotype is changed as a result of the cellular uptake of exogenous DNA or RNA, and, for example, die transformed cell expresses a recombinant form of an HDx polypeptide or, where anti¬ sense expression occurs from die transferred gene, the expression of a naturally- occurring form of die HDx protein is disrupted.
As used herein, the term "specifically hybridizes" refers to the ability of the probe/primer of die invention to hybridize to at least 15 consecutive nucleotides of an HDx gene, such as an HDx sequence designated in one of SEQ ID Nos: 1-4, or a sequence complementary thereto, or naturally occurring mutants thereof, such that it has less than 15%, preferably less than 10%, and more preferably less than 5% background hybridization to a cellular nucleic acid (e.g., mRNA or genomic DNA) encoding a protein other than an HDx protein, as defined herein. In preferred embodiments, the oligonucleotide probe specifically detects only one of the subject HDx paralogs, e.g., does not substantially hybridize to transcripts for other HDx homologs in the same species.
As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of preferred vector is an episome, i.e., a nucleic acid capable of extra-chromosomal replication. Preferred vectors are diose capable of autonomous replication and/expression of nucleic acids to which they are linked. Vectors capable of directing the expression of genes to which they are operatively linked are referred to herein as "expression vectors". In general, expression vectors of utility in recombinant DNA techniques are often in the form of "plasmids" which refer generally to circular double stranded DNA loops which, in their vector form are not bound to the chromosome. In the present specification, "plasmid" and "vector" are used interchangeably as d e plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors which serve equivalent functions and which become known in the art subsequently hereto. "Transcriptional regulatory sequence" is a generic term used throughout the specification to refer to DNA sequences, such as initiation signals, enhancers, and promoters, which induce or control transcription of protein coding sequences widi which they are operably linked. In preferred embodiments, transcription of one of the recombinant HDx genes is under the control of a promoter sequence (or other transcriptional regulatory sequence) which controls the expression of the recombinant gene in a cell-type in which expression is intended. It will also be understood that the recombinant gene can be under the control of transcriptional regulatory sequences which are the same or which are different from those sequences which control transcription of the naturally-occurring forms of HDx genes. As used herein, the term "tissue-specific promoter" means a DNA sequence that serves as a promoter, i.e., regulates expression of a selected DNA sequence operably linked to the promoter, and which effects expression of the selected DNA sequence in specific cells of a tissue, such as cells of hepatic, pancreatic, neuronal or hematopoietic origin. The term also covers so-called "leaky" promoters, which regulate expression of a selected DNA primarily in one tissue, but can cause at least low level expression in other tissues as well.
As used herein, a "transgenic animal" is any animal, preferably a non-human mammal, bird or an amphibian, in which one or more of the cells of the animal contain heterologous nucleic acid introduced by way of human intervention, such as by transgenic techniques well known in the art. The nucleic acid is introduced into the cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection wid a recombinant virus. The term genetic manipulation does not include classical cross-breeding, or in vitro fertilization, but rather is directed to the introduction of a recombinant DNA molecule. This molecule may be integrated within a chromosome, or it may be extrachromosomally replicating DNA. In the typical transgenic animals described herein, the transgene causes cells to express a recombinant form of one of the HDx proteins, e.g. either agonistic or antagonistic forms. However, transgenic animals in which the recombinant HDx gene is silent are also contemplated, as for example, the FLP or CRE recombinase dependent constructs described below. - Moreover, "transgenic animal" also includes those recombinant animals in which gene disruption of one or more HDx genes is caused by human intervention, including both recombination and antisense techniques.
The "non-human animals" of the invention include vertebrates such as rodents, non-human primates, sheep, dog, cow, chickens, amphibians, reptiles, etc. Preferred non- human animals are selected from the rodent family including rat and mouse, most preferably mouse, though transgenic amphibians, such as members of the Xenopus genus, and transgenic chickens can also provide important tools for understanding and identifying agents which can affect, for example, embryogenesis and tissue formation. The invention also contemplates transgenic insects, including those of the genus Drosophila, such as D. melanogaster. The term "chimeric animal" is used herein to refer to animals in which the recombinant gene is found, or in which the recombinant is expressed in some but not all cells of the animal. The term "tissue-specific chimeric animal" indicates that one of the recombinant HDx genes is present and/or expressed or disrupted in some tissues but not others.
As used herein, the term "transgene" means a nucleic acid sequence (encoding, e.g., one of the HDx polypeptides, or pending an antisense transcript thereto), which is partly or entirely heterologous, i.e., foreign, to the transgenic animal or cell into which it is introduced, or, is homologous to an endogenous gene of the transgenic animal or cell into which it is introduced, but which is designed to be inserted, or is inserted, into the animal's genome in such a way as to alter the genome of the cell into which it is inserted (e.g., it is inserted at a location which differs from that of the natural gene or its insertion results in a knockout). A transgene can include one or more transcriptional regulatory sequences and any other nucleic acid, such as introns, that may be necessary for optimal expression of a selected nucleic acid.
As is well known, genes for a particular polypeptide may exist in single or multiple copies within the genome of an individual. Such duplicate genes may be identical or may have certain modifications, including nucleotide substitutions, additions or deletions, which all still code for polypeptides having substantially the same activity. The term "DNA sequence encoding an HDx polypeptide" may thus refer to one or more genes within a particular individual. Moreover, certain differences in nucleotide sequences may exist between individuals of the same species, which are called alleles. Such allelic differences may or may not result in differences in amino acid sequence of the encoded polypeptide yet still encode a protein with the same biological activity. "Homology" refers to sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base or amino acid, then the molecules are homologous at that position. A degree of homology between sequences is a function of die number of matching or homologous positions shared by the sequences. An "unrelated" or "non-homologous" sequence shares less tiian 40 percent identity, though preferably less than 25 percent identity, with one of the HDx sequences of the present invention.
As used herein, an "HDx-related" protein refers to die HDx proteins described herein, and other human homologs of those HDx sequences, as well as orthologs and paralogs (homologs) of the HDx proteins in other species, ranging from yeast to other mammals, e.g., homologous histone deacetylase, The term "ortholog" refers to genes or proteins which are homologs via speciation, e.g., closely related and assumed to have common descent based on structural and functional considerations. Orthologous proteins function as recognizably the same activity in different species. The term "paralog" refers to genes or proteins which are homologs via gene duplication, e.g., duplicated variants of a gene within a genome. See also, Fritch, WM (1970) Syst Zool 19:99-113.
"Cells," "host cells" or "recombinant host cells" are terms used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein. A "chimeric protein" or "fusion protein" is a fusion of a first amino acid sequence encoding one of the subject HDx polypeptides with a second amino acid sequence defining a domain (e.g. polypeptide portion) foreign to and not substantially homologous with any domain of one of the HDx proteins. A chimeric protein may present a foreign domain which is found (albeit in a different protein) in an organism which also expresses the first protein, or it may be an "interspecies", "intergenic", etc. fusion of protein structures expressed by different kinds of organisms. In general, a fusion protein can be represented by the general formula X-HDx-Y, wherein HDx represents a portion of the protein which is derived from one of the HDx proteins, and X and Y are, independently, absent or represent amino acid sequences which are not related to one of the HDx sequences in an organism.
The term "isolated" as also used herein with respect to nucleic acids, such as DNA or RNA, refers to molecules separated from other DNAs, or RNAs, respectively, that are present in the natural source of the macromolecule. For example, an isolated nucleic acid encoding one of the subject HDx polypeptides preferably includes no more than 10 kilobases (kb) of nucleic acid sequence which naturally immediately flanks the HDx gene in genomic DNA, more preferably no more than 5kb of such naturally occurring flanking sequences, and most preferably less than 1.5kb of such naturally occurring flanking sequence. The term isolated as used herein also refers to a nucleic acid or peptide that is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or odier chemicals when chemically synthesized. Moreover, an "isolated nucleic acid" is meant to include nucleic acid fragments which are not naturally occurring as fragments and would not be found in the natural state.
As described below, one aspect of the invention pertains to isolated nucleic acids comprising nucleotide sequences encoding HDx polypeptides, and or equivalents of such nucleic acids. The term nucleic acid as used herein is intended to include fragments as equivalents. The term equivalent is understood to include nucleotide sequences encoding functionally equivalent HDx polypeptides or functionally equivalent peptides having an activity of an HDx protein such as described herein. Equivalent nucleotide sequences will include sequences that differ by one or more nucleotide substitutions, additions or deletions, such as allelic variants; and will, therefore, include sequences that differ from the nucleotide sequence of the HDx cDNA sequences shown in any of SEQ ID Nos: 1-4 due to the degeneracy of the genetic code. Equivalents will also include nucleotide sequences diat hybridize under stringent conditions (i.e., equivalent to about 20-27°C below the melting temperature (Tm) of the DNA duplex formed in about IM salt) to the nucleotide sequences represented in one or more of SEQ ID Nos: 1-4. In one embodiment, equivalents will further include nucleic acid sequences derived from and evolutionarily related to, a nucleotide sequences shown in any of SEQ ID Nos: 1-4.
Moreover, it will be generally appreciated that, under certain circumstances, it may be advantageous to provide homologs of one of the subject HDx polypeptides which function in a limited capacity as one of either an HDx agonist (mimetic) or an HDx antagonist, in order to promote or inhibit only a subset of the biological activities of the naturally-occurring form of the protein. Thus, specific biological effects can be elicited by treatment with a homolog of limited function, and with fewer side effects relative to treatment with agonists or antagonists which are directed to all of die biological activities of naturally occurring forms of HDx proteins.
Homologs of each of the subject HDx proteins can be generated by mutagenesis, such as by discrete point mutation(s), or by truncation. For instance, mutation can give rise to homologs which retain substantially the same, or merely a subset, of the biological activity of the HDx polypeptide from which it was derived. Alternatively, antagonistic forms of the protein can be generated which are able to inhibit the function of the naturally occurring form of the protein, such as by competitively binding to an HDx substrate or HDx associated protein, as for example competing with wild-type HDx in the binding of RbAp48 or a histone. In addition, agonistic forms of the protein may be generated which are constitutively active, or have an altered K^ or Km for deacetylation reactions. Thus, the HDx protein and homologs thereof provided by the subject invention may be either positive or negative regulators of transcription and/or replication.
In general, polypeptides referred to herein as having an activity of an HDx protein (e.g., are "bioactive") are defined as polypeptides which include an amino acid sequence corresponding (e.g., identical or homologous) to all or a portion of die arnino acid sequences of an HDx proteins shown in any one or more of SEQ ID Nos:5-8 and which mimic or antagonize all or a portion of the biological/biochemical activities of a naturally occurring HDx protein. Examples of such biological activity include the ability to modulate proliferation of cells. For example, inhibiting histone deacetylation causes cells to arrest in GI and G2 phases of the cell cycle. The biochemical activity associated with HDx proteins of the present invention can also characterized in terms of binding to and (optionally) catalyzing the deacetylation of an acetylated histone. Another biochemical property of certain of the subject HDx proteins involves binding to other cellular proteins, such as RbAp48 or Sin3A. Other biological activities of die subject HDx proteins are described herein or will be reasonably apparent to those skilled in the art. According to the present invention, a polypeptide has biological activity if it is a specific agonist or antagonist of a naturally- occurring form of an HDx protein.
Preferred nucleic acids encode an HDx polypeptide comprising an amino acid sequence at least 80% homologous, more preferably at least 85% homologous and most preferably at least 88% homologous with an amino acid sequence of a human HDx, e.g., such as selected from the group consisting of SEQ ID Nos: 5-8. Nucleic acids which encode polypeptides at least about 90%, more preferably at least about 95%, and most preferably at least about 98-99% homology with an amino acid sequence represented in one of SEQ ID Nos: 5-8 are of course also within the scope of the invention, as are nucleic acids identical in sequence with any of the enumerated HDx sequences of the sequence listing. In one embodiment, the nucleic acid is a cDNA encoding a polypeptide having at least one activity of the subject HDx polypeptide.
In certain preferred embodiments, the invention features a purified or recombinant HDx polypeptide having peptide chain with a molecular weight in the range of 40kd to 60kd, even more preferably in the range of 45-50 kd or 53-58kd. It will be understood diat certain post-translational modifications, e.g., phosphorylation and the like, can increase the apparent molecular weight of the HDx protein relative to the unmodified polypeptide chain, and cleavage of certain sequences, such as pro-sequences, can likewise decrease the apparent molecular weight.
In other preferred embodiments, the nucleic acid encodes an HDx polypeptide which includes both the v and χ motifs, and preferably possess a histone deacetylase activity. For example, preferred HDx proteins are represented by the general formula A- (v motif)-B-(χ motif)-C, wherein the v motif is an amino acid sequence represented in SEQ ID NO. 12, more preferably SEQ ID No. 13, the χ motif is an amino acid sequence represented in SEQ ID No. 14, more preferably SEQ ID No. 15, and A, B and C represent amino acid sequences which are correspond to HDx or HDx-related proteins.
Still other preferred nucleic acids of the present invention encode an HDx polypeptide which includes a polypeptide sequence corresponding to all or a portion of amino acid residues of any one of SEQ ID Nos: 5-8, e.g., at least 5, 10, 25, 50 or 100 amino acid residues of that region.
Another aspect of the invention provides a nucleic acid which hybridizes under high or low stringency conditions to the nucleic acid represented by SEQ ID No: 1. Appropriate stringency conditions which promote DNA hybridization, for example, 6.0 x sodium chloride/sodium citrate (SSC) at about 45°C, followed by a wash of 2.0 x SSC at 50°C, are known to those skilled in the art or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. For example, the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at 50°C to a high stringency of about 0.2 x SSC at 50°C. In addition, the temperature in the wash step can be increased from low stringency conditions at room temperature, about 22°C, to high stringency conditions at about 65°C.
Nucleic acids, having a sequence that differs from the nucleotide sequences shown in one of SEQ ID Nos: 1-4 due to degeneracy in the genetic code are also within the scope of the invention. Such nucleic acids encode functionally equivalent peptides (i.e., a peptide having a biological activity of an HDx polypeptide) but differ in sequence from the sequence shown in the sequence listing due to degeneracy in the genetic code. For example, a number of amino acids are designated by more than one triplet. Codons diat specify the same amino acid, or synonyms (for example, CAU and CAC each encode histidine) may result in "silent" mutations which do not affect die amino acid sequence of an HDx polypeptide. However, it is expected that DNA sequence polymorphisms that do lead to changes in the amino acid sequences of the subject HDx polypeptides will exist among, for example, humans. One skilled in the art will appreciate that these variations in one or more nucleotides (up to about 3-5% of the nucleotides) of the nucleic acids encoding polypeptides having an activity of an HDx polypeptide may exist among individuals of a given species due to natural allelic variation.
As used herein, an HDx gene fragment refers to a nucleic acid having fewer nucleotides than the nucleotide sequence encoding the entire mature form of an HDx protein yet which (preferably) encodes a polypeptide which retains some biological activity of the full length protein. Fragment sizes contemplated by the present invention include, for example, 5, 10, 25, 50, 75, 100, or 200 amino acids in length.
As indicated by the examples set out below, HDx protein-encoding nucleic acids can be obtained from mRNA present in any of a number of eukaryotic cells. It should also be possible to obtain nucleic acids encoding HDx polypeptides of the present invention from genomic DNA from both adults and embryos. For example, a gene encoding an HDx protein can be cloned from either a cDNA or a genomic library in accordance with protocols described herein, as well as those generally known to persons skilled in the art. A cDNA encoding an HDx protein can be obtained by isolating total mRNA from a cell, e.g. a mammalian cell, e.g. a human cell, including embryonic cells. Double stranded cDNAs can then be prepared from the total mRNA, and subsequently inserted into a suitable plasmid or bacteriophage vector using any one of a number of known techniques. The gene encoding an HDx protein can also be cloned using established polymerase chain reaction techniques in accordance with the nucleotide sequence information provided by the invention. The nucleic acid of the invention can be DNA or RNA. A preferred nucleic acid is a cDNA including a nucleotide sequence represented by one of SEQ ID Nos: 1-4.
Another aspect of the invention relates to the use of the isolated nucleic acid in "antisense" therapy. As used herein, "antisense" therapy refers to administration or in situ generation of oligonucleotide probes or their derivatives which specifically hybridize (e.g. binds) under cellular conditions, with the cellular mRNA and/or genomic DNA encoding one or more of the subject HDx proteins so as to inhibit expression of diat protein, e.g. by inhibiting transcription and/or translation. The binding may be by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interactions in the major groove of the double helix. In general, "antisense" therapy refers to the range of techniques generally employed in the art, and includes any therapy which relies on specific binding to oligonucleotide sequences.
An antisense construct of the present invention can be delivered, for example, as an expression plasmid which, when transcribed in the cell, produces RNA which is complementary to at least a unique portion of the cellular mRNA which encodes an HDx protein. Alternatively, the antisense construct is an oligonucleotide probe which is generated ex vivo and which, when introduced into the cell causes inhibition of expression by hybridizing with the mRNA and/or genomic sequences of an HDx gene. Such oligonucleotide probes are preferably modified oligonucleotides which are resistant to endogenous nucleases, e.g. exonucleases and/or endonucleases, and are therefore stable in vivo. Exemplary nucleic acid molecules for use as antisense oligonucleotides are phosphoramidate, phosphothioate and methylphosphonate analogs of DNA (see also U.S.
Patents 5,176,996; 5,264,564; and 5,256,775), or peptide nucleic acids (PNAs). Additionally, general approaches to constructing oligomers useful in antisense therapy have been reviewed, for example, by Van der Krol et al. (1988) Biotechmques 6:958- 976; and Stein et al. (1988) Cancer Res 48:2659-2668.
Accordingly, the modified oligomers of the invention are useful in therapeutic, diagnostic, and research contexts. In therapeutic applications, the oligomers are utilized in a manner appropriate for antisense therapy in general. For such therapy, the oligomers of the invention can be formulated for a variety of routes of administration, including systemic and topical or localized administration. Techniques and formulations generally may be found in Remmington's Pharmaceutical Sciences, Meade Publishing Co., Easton, PA. For systemic administration, injection is preferred, including intramuscular, intravenous, intraperitoneal, and subcutaneous. For injection, the oligomers of the invention can be formulated in liquid solutions, preferably in physiologically compatible buffers such as Hank's solution or Ringer's solution. In addition, the oligomers may be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms are also included. Systemic administration can also be by transmucosal or transdermal means, or the compounds can be administered orally. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration bile salts and fusidic acid derivatives. In addition, detergents may be used to facilitate permeation. Transmucosal administration may be through nasal sprays or using suppositories. For oral administration, the oligomers are formulated into conventional oral administration forms such as capsules, tablets, and tonics. For topical administration, the oligomers of the invention are formulated into ointments, salves, gels, or creams as generally known in the art. In addition to use in therapy, the oligomers of the invention may be used as diagnostic reagents to detect the presence or absence of the target DNA or RNA sequences to which they specifically bind. Such diagnostic tests are described in further detail below.
Likewise, die antisense constructs of the present invention, by antagonizing the normal biological activity of one of the HDx proteins, can be used in the manipulation of tissue, e.g. tissue differentiation or growth, both in vivo and ex vivo.
Furthermore, die anti-sense techniques (e.g. microinjection of antisense molecules, or transfection with plasmids whose transcripts are anti-sense with regard to an HDx mRNA or gene sequence) can be used to investigate role of HDx in developmental events, as well as the normal cellular function of HDx in adult tissue. Such techniques can be utilized in cell culture, but can also be used in d e creation of transgenic animals (described infra).
This invention also provides expression vectors containing a nucleic acid encoding an HDx polypeptide, operably linked to at least one transcriptional regulatory sequence. Operably linked is intended to mean that the nucleotide sequence is linked to a regulatory sequence in a manner which allows expression of the nucleotide sequence. Regulatory sequences are art-recognized and are selected to direct expression of the subject HDx proteins. Accordingly, the term transcriptional regulatory sequence includes promoters, enhancers and other expression control elements. Such regulatory sequences are described in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990). For instance, any of a wide variety of expression control sequences, sequences diat control the expression of a DNA sequence when operatively linked to it, may be used in these vectors to express DNA sequences encoding HDx polypeptides of this invention. Such useful expression control sequences, include, for example, a viral LTR, such as the LTR of the Moloney murine leukemia virus, the early and late promoters of SV40, adenovirus or cytomegalovirus immediate early promoter, the lac system, the trp system, the TAC or TRC system, T7 promoter whose expression is directed by T7 RNA polymerase, the major operator and promoter regions of phage λ, the control regions for fd coat protein, the promoter for 3- phosphogiycerate kinase or other glycolytic enzymes, the promoters of acid phosphatase, e.g., Pho5, the promoters of die yeast α-mating factors, the polyhedron promoter of the baculovirus system and other sequences known to control the expression of genes of prokaryotic or eukaryotic cells or their viruses, and various combinations thereof. It should be understood that the design of the expression vector may depend on such factors as the choice of the host cell to be transformed and/or the type of protein desired to be expressed. Moreover, the vector's copy number, the ability to control that copy number and the expression of any other proteins encoded by the vector, such as antibiotic markers, should also be considered. In one embodiment, the expression vector includes a recombinant gene encoding a peptide having an agonistic activity of a subject HDx polypeptide, or alternatively, encoding a peptide which is an antagonistic form of the HDx protein, such as a catalytically-inactive deacetylase. Such expression vectors can be used to transfect cells and thereby produce polypeptides, including fusion proteins, encoded by nucleic acids as described herein. Moreover, the gene constructs of the present invention can also be used as a part of a gene therapy protocol to deliver nucleic acids, e.g., encoding either an agonistic or antagonistic form of one of the subject HDx proteins or an antisense molecule described above. Thus, another aspect of the invention features expression vectors for in vivo or in vitro transfection and expression of an HDx polypeptide or antisense molecule in particular cell types so as to reconstitute the function of, or alternatively, abrogate the function of HDx-induced transcription in a tissue in which the naturally-occurring form of the protein is misexpressed; or to deliver a form of the protein which alters differentiation of tissue, or which inhibits neoplastic transformation. Expression constructs of the subject HDx polypeptides, as well as antisense constructs, may be administered in any biologically effective carrier, e.g. any formulation or composition capable of effectively delivering the recombinant gene to cells in vivo. Approaches include insertion of the subject gene in viral vectors including recombinant retroviruses, adenovirus, adeno-associated virus, and herpes simplex virus- 1, or recombinant bacterial or eukaryotic plasmids. Viral vectors transfect cells directly; plasmid DNA can be delivered with the help of, for example, cationic liposomes (lipofectin) or derivatized (e.g. antibody conjugated), polylysine conjugates, gramacidin S, artificial viral envelopes or other such intracellular carriers, as well as direct injection of the gene construct or CaPO precipitation carried out in vivo. It will be appreciated that because transduction of appropriate target cells represents the critical first step in gene therapy, choice of the particular gene delivery system will depend on such factors as the phenotype of the intended target and the route of administration, e.g. locally or systemically. Furthermore, it will be recognized that the particular gene construct provided for in vivo transduction of HDx expression are also useful for in vitro transduction of cells, such as for use in the ex vivo tissue culture systems described below.
A preferred approach for in vivo introduction of nucleic acid into a cell is by use of a viral vector containing nucleic acid, e.g. a cDNA encoding the particular HDx polypeptide desired. Infection of cells with a viral vector has the advantage that a large proportion of the targeted cells can receive the nucleic acid. Additionally, molecules encoded within the viral vector, e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells which have taken up viral vector nucleic acid. Retrovirus vectors, adenovirus vectors and adeno-associated virus vectors are exemplary recombinant gene delivery system for the transfer of exogenous genes in vivo, particularly into humans. These vectors provide efficient delivery of genes into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host.
In addition to viral transfer methods, such as those illustrated above, non-viral methods can also be employed to cause expression of a subject HDx polypeptide in the tissue of an animal. Most nonviral methods of gene transfer rely on normal mechanisms used by mammalian cells for the uptake and intracellular transport of macromolecules. In preferred embodiments, non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject HDx polypeptide gene by the targeted cell. Exemplary gene delivery systems of this type include liposomal derived systems, poly-lysine conjugates, and artificial viral envelopes.
In clinical settings, the gene delivery systems for the therapeutic HDx gene can be introduced into a patient by any of a number of methods, each of which is familiar in the art. For instance, a pharmaceutical preparation of the gene delivery system can be introduced systemically, e.g. by intravenous injection, and specific transduction of the protein in the target cells occurs predominantly from specificity of transfection provided by the gene delivery vehicle, cell-type or tissue-type expression due to the transcriptional regulatory sequences controlling expression of the receptor gene, or a combination thereof. In other embodiments, initial delivery of the recombinant gene is more limited with introduction into the animal being quite localized. For example, the gene delivery vehicle can be introduced by catheter (see U.S. Patent 5,328,470) or by stereotactic injection (e.g. Chen et al. (1994) PNAS 91: 3054-3057). AN HDx gene, such as any one of the clones represented in the group consisting of SEQ ID NO: 1-4, can be delivered in a gene therapy construct by electroporation using techniques described, for example, by Dev et al. ((1994) Cancer Treat Rev 20: 105-115). The pharmaceutical preparation of the gene therapy construct can consist essentially of the gene delivery system in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery system can be produced intact from recombinant cells, e.g. retroviral vectors, the pharmaceutical preparation can comprise one or more cells which produce the gene delivery system.
Another aspect of the present invention concerns recombinant forms of the HDx proteins. Recombinant polypeptides preferred by the present invention, in addition to native HDx proteins, are at least 80% homologous, more preferably at least 85% homologous and most preferably at least 88% homologous with an amino acid sequence represented by any of SEQ ID Nos: 5-8. Polypeptides which possess an activity of an HDx protein (i.e. either agonistic or antagonistic), and which are at least 90%, more preferably at least 95%, and most preferably at least about 98-99% homologous with a sequence selected from the group consisting of SEQ ID Nos: 5-8 are also within the scope of the invention. In other preferred embodiments, the HDx polypeptide includes both the v and χ motifs, and preferably possess a histone deacetylase activity.
The term "recombinant HDx protein" refers to a polypeptide which is produced by recombinant DNA techniques, wherein generally, DNA encoding an HDx polypeptide is inserted into a suitable expression vector which is in turn used to transform a host cell to produce the heterologous protein. Moreover, the phrase "derived from", with respect to a recombinant HDx gene, is meant to include within the meaning of "recombinant protein" those proteins having an amino acid sequence of a native HDx protein, or an amino acid sequence similar thereto which is generated by mutations including substitutions and deletions (including truncation) of a naturally occurring form of the protein.
The present invention further pertains to recombinant forms of the subject HDx polypeptides which are encoded by genes derived from a mammal (e.g. a human), and which have amino acid sequences evolutionarily related to the HDx proteins represented in SEQ ID Nos: 5-8. Such recombinant HDx polypeptides preferably are capable of functioning in one of either role of an agonist or antagonist of at least one biological activity of a wild-type ("authentic") HDx protein of the appended sequence listing. The term "evolutionarily related to", with respect to amino acid sequences of HDx proteins, refers to both polypeptides having amino acid sequences which have arisen naturally, and also to mutational variants of HDx polypeptides which are derived, for example, by combinatorial mutagenesis.
The present invention also provides methods of producing the subject HDx polypeptides. For example, a host cell transfected with a nucleic acid vector directing expression of a nucleotide sequence encoding the subject polypeptides can be cultured under appropriate conditions to allow expression of the peptide to occur. The cells may be harvested, lysed and the protein isolated. A cell culture includes host cells, media and other byproducts. Suitable media for cell culture are well known in the art. The recombinant HDx polypeptide can be isolated from cell culture medium, host cells, or both using techniques known in the art for purifying proteins including ion-exchange chromatography, gel filtration chromatography, ultrafiltration, electrophoresis, and immunoaffinity purification with antibodies specific for such peptide. In a preferred embodiment, the recombinant HDx polypeptide is a fusion protein containing a domain which facilitates its purification, such as GST fusion protein or poly(Ηis) fusion protein.
This invention also pertains to a host cell transfected to express recombinant forms of the subject HDx polypeptides. The host cell may be any prokaryotic or eukaryotic cell. Thus, a nucleotide sequence derived from the cloning of HDx proteins, encoding all or a selected portion of a full-length protein, can be used to produce a recombinant form of an HDx polypeptide via microbial or eukaryotic cellular processes. Ligating the polynucleotide sequence into a gene construct, such as an expression vector, and transforming or transfecting into hosts, either eukaryotic (yeast, avian, insect or mammalian) or prokaryotic (bacterial cells), are standard procedures used in producing other well-known proteins, e.g. MAP kinases, p53, WTl, PTP phosphatases, SRC, and the like. Similar procedures, or modifications thereof, can be employed to prepare recombinant HDx polypeptides by microbial means or tissue-culture technology in accord with the subject invention. The recombinant HDx genes can be produced by ligating nucleic acid encoding an
HDx protein, or a portion thereof, into a vector suitable for expression in either prokaryotic cells, eukaryotic cells, or both. Expression vectors for production of recombinant forms of the subject HDx polypeptides include plasmids and other vectors. For instance, suitable vectors for the expression of an HDx polypeptide include plasmids of die types: pBR322-derived plasmids, pEMBL-derived plasmids, pEX-derived plasmids, pBTac-derived plasmids and pUC-derived plasmids for expression in prokaryotic cells, such as E. coli.
A number of vectors exist for the expression of recombinant proteins in yeast. For instance, YEP24, YIP5, YEP51, YEP52, ρYES2, and YRP17 are cloning and expression vehicles useful in the introduction of genetic constructs into S. cerevisiae (see, for example, Broach et al. (1983) in Experimental Manipulation of Gene Expression, ed. M. Inouye Academic Press, p. 83, incoφorated by reference herein). These vectors can replicate in E. coli due the presence of the pBR322 ori, and in S. cerevisiae due to the replication determinant of the yeast 2 micron plasmid. In addition, drug resistance markers such as ampicillin can be used. In an illustrative embodiment, an HDx polypeptide is produced recombinantly utilizing an expression vector generated by sub¬ cloning the coding sequence of one of the HDx genes represented in SEQ ID Nos: 1-4.
The preferred mammalian expression vectors contain both prokaryotic sequences, to facilitate the propagation of the vector in bacteria, and one or more eukaryotic transcription units that are expressed in eukaryotic cells. The pcDNAI/amp, pcDNAI/neo, pRc/CMV, pSV2gpt, pSV2neo, pSV2-dhfr, pTk2, pRSVneo, pMSG, pSVT7, pko-neo and pHyg derived vectors are examples of mammalian expression vectors suitable for transfection of eukaryotic cells. Some of these vectors are modified with sequences from bacterial plasmids, such as pBR322, to facilitate replication and drug resistance selection in both prokaryotic and eukaryotic cells. Alternatively, derivatives of viruses such as the bovine papillomavirus (BPV-1), or Epstein-Barr virus (pHEBo, pREP-derived and p205) can be used for transient expression of proteins in eukaryotic cells. The various methods employed in the preparation of the plasmids and transformation of host organisms are well known in the art. For other suitable expression systems for both prokaryotic and eukaryotic cells, as well as general recombinant procedures, see Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989) Chapters 16 and 17.
In some instances, it may be desirable to express the recombinant HDx polypeptide by the use of a baculovirus expression system. Examples of such baculovirus expression systems include pVL-derived vectors (such as pVL1392, pVL1393 and pVL941), pAcUW-derived vectors (such as pAcUWl), and pBlueBac-derived vectors
(such as the β-gal containing pBlueBac III).
When it is desirable to express only a portion of an HDx protein, such as a form lacking a portion of the N-terminus, i.e. a truncation mutant which lacks the signal peptide, it may be necessary to add a start codon (ATG) to the oligonucleotide fragment containing the desired sequence to be expressed. It is well known in the art that a methionine at the N-terminal position can be enzymatically cleaved by the use of the enzyme methionine aminopeptidase (MAP). MAP has been cloned from E. coli (Ben- Bassat et al. (1987) J. Bacteriol. 169:751-757) and Salmonella typhimurium and its in vitro activity has been demonstrated on recombinant proteins (Miller et al. (1987) PNAS
84:2718-1722). Therefore, removal of an N-terminal methionine, if desired, can be achieved either in vivo by expressing HDx-derived polypeptides in a host which produces
MAP (e.g., E. coli or CM89 or S. cerevisiae), or in vitro by use of purified MAP (e.g., procedure of Miller et al., supra).
Alternatively, the coding sequences for the polypeptide can be incoφorated as a part of a fusion gene including a nucleotide sequence encoding a different polypeptide.
This type of expression system can be useful under conditions where it is desirable to produce an immunogenic fragment of an HDx protein. For example, the VP6 capsid protein of rotavirus can be used as an immunologic carrier protein for portions of the HDx polypeptide, either in the monomeric form or in the form of a viral particle. The nucleic acid sequences corresponding to the portion of a subject HDx protein to which antibodies are to be raised can be incoφorated into a fusion gene construct which includes coding sequences for a late vaccinia virus structural protein to produce a set of recombinant viruses expressing fusion proteins comprising HDx epitopes as part of the virion. It has been demonstrated with the use of immunogenic fusion proteins utilizing the Hepatitis B surface antigen fusion proteins that recombinant Hepatitis B virions can be utilized in this role as well. Similarly, chimeric constructs coding for fusion proteins containing a portion of an HDx protein and the poliovirus capsid protein can be created to enhance immunogenicity of the set of polypeptide antigens (see, for example, EP Publication No: 0259149; and Evans et al. (1989) Nature 339:385; Huang et al. (1988) J. Virol. 62:3855; and Schlienger et al. (1992) J. Virol. 66:2).
The Multiple Antigen Peptide system for peptide-based immunization can also be utilized to generate an immunogen, wherein a desired portion of an HDx polypeptide is obtained directly from organo-chemical synthesis of the peptide onto an oligomeric branching lysine core (see, for example, Posnett et al. (1988) JBC 263:1719 and Nardelli et al. (1992) J. Immunol. 148:914). Antigenic determinants of HDx proteins can also be expressed and presented by bacterial cells.
In addition to utilizing fusion proteins to enhance immunogenicity, it is widely appreciated that fusion proteins can also facilitate the expression of proteins, and accordingly, can be used in the expression of the HDx polypeptides of the present invention. For example, HDx polypeptides can be generated as glutathione-S-transferase (GST-fusion) proteins. Such GST-fusion proteins can enable easy purification of the HDx polypeptide, as for example by the use of glutathione-derivatized matrices (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. (N.Y.: John Wiley & Sons, 1991)).
In another embodiment, a fusion gene coding for a purification leader sequence, such as a poly-(Ηis)/enterokinase cleavage site sequence at the N-terminus of the desired portion of the recombinant protein, can allow purification of the expressed fusion protein by affinity chromatography using a Ni2+ metal resin. The purification leader sequence can then be subsequently removed by treatment with enterokinase to provide the purified protein (e.g., see Hochuli et al. (1987) J. Chromatography 411:177; and Janknecht et al. PNAS 88:8972). Techniques for making fusion genes are known to those skilled in the art. Essentially, the joining of various DNA fragments coding for different polypeptide sequences is performed in accordance with conventional techniques, employing blunt- ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. John Wiley & Sons: 1992).
HDx polypeptides may also be chemically modified to create HDx derivatives by forming covalent or aggregate conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like. Covalent derivatives of HDx proteins can be prepared by linking the chemical moieties to functional groups on amino acid sidechains of the protein or at the N-teιτninus or at the C-terminus of the polypeptide.
The present invention also makes available isolated HDx polypeptides which are isolated from, or otherwise substantially free of other cellular proteins, especially other signal transduction factors and/or transcription factors which may normally be associated with the HDx polypeptide. The term "substantially free of other cellular proteins" (also referred to herein as "contaminating proteins") or "substantially pure or purified preparations" are defined as encompassing preparations of HDx polypeptides having less than 20% (by dry weight) contaminating protein, and preferably having less tiian 5% contaminating protein. Functional forms of the subject polypeptides can be prepared, for the first time, as purified preparations by using a cloned gene as described herein. By "purified", it is meant, when referring to a peptide or DNA or RNA sequence, that the indicated molecule is present in the substantial absence of other biological macromolecules, such as other proteins. The term "purified" as used herein preferably means at least 80% by dry weight, more preferably in the range of 95-99% by weight, and most preferably at least 99.8% by weight, of biological macromolecules of the same type present (but water, buffers, and other small molecules, especially molecules having a molecular weight of less than 5000, can be present). The term "pure" as used herein preferably has the same numerical limits as "purified" immediately above. "Isolated" and "purified" do not encompass either natural materials in their native state or natural materials that have been separated into components (e.g., in an acrylamide gel) but not obtained either as pure (e.g. lacking contaminating proteins, or chromatography reagents such as denaturing agents and polymers, e.g. acrylamide or agarose) substances or solutions. In preferred embodiments, purified HDx preparations will lack any contaminating proteins from the same animal from that HDx is normally produced, as can be accomplished by recombinant expression of, for example, a human HDx protein in a non-human cell.
As described above for recombinant polypeptides, isolated HDx polypeptides can include all or a portion of an amino acid sequences corresponding to an HDx polypeptide represented in any one of SEQ ID Nos: 5-8 or homologous sequences thereto. In preferred embodiments, the HDx polypeptide includes both die v and χ motifs, and preferably possess a histone deacetylase activity.
Isolated peptidyl portions of HDx proteins can be obtained by screening peptides recombinantly produced from the corresponding fragment of the nucleic acid encoding such peptides. In addition, fragments can be chemically synthesized using techniques known in the art such as conventional Merrifield solid phase f-Moc or t-Boc chemistry. For example, an HDx polypeptide of the present invention may be arbitrarily divided into fragments of desired length with no overlap of the fragments, or preferably divided into overlapping fragments of a desired length. The fragments can be produced (recombinantly or by chemical synthesis) and tested to identify those peptidyl fragments which can function as either agonists or antagonists of a wild-type (e.g., "authentic") HDx protein.
The recombinant HDx polypeptides of the present invention also include homologs of the authentic HDx proteins, such as versions of those protein which are resistant to proteolytic cleavage, as for example, due to mutations which alter ubiquitination or other enzymatic targeting associated with the protein.
Modification of the structure of the subject HDx polypeptides can be for such puφoses as enhancing therapeutic or prophylactic efficacy, stability (e.g., ex vivo shelf life and resistance to proteolytic degradation in vivo), or post-translational modifications (e.g., to alter phosphorylation pattern of protein). Such modified peptides, when designed to retain at least one activity of die naturally-occurring form of the protein, or to produce specific antagonists thereof, are considered functional equivalents of the HDx polypeptides described in more detail herein. Such modified peptides can be produced, for instance, by amino acid substitution, deletion, or addition.
For example, it is reasonable to expect that an isolated replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, a threonine with a serine, or a similar replacement of an amino acid with a structurally related amino acid (i.e. isosteric and/or isoelectric mutations) will not have a major effect on the biological activity of the resulting molecule. Conservative replacements are those that take place within a family of amino acids that are related in their side chains. Genetically encoded amino acids are can be divided into four families: (1) acidic = aspartate, glutamate; (2) basic = lysine, arginine, histidine; (3) nonpolar = alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan; and (4) uncharged polar = glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine. Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids. In similar fashion, the amino acid repertoire can be grouped as (1) acidic = aspartate, glutamate; (2) basic = lysine, arginine histidine, (3) aliphatic = glycine, alanine, valine, leucine, isoleucine, serine, threonine, with serine and threonine optionally be grouped separately as aliphatic- hydroxyl; (4) aromatic = phenylalanine, tyrosine, tryptophan; (5) amide = asparagine, glutamine; and (6) sulfur -containing = cysteine and methionine. (see, for example, Biochemistry, 2nd ed., Ed. by L. Stryer, WH Freeman and Co.: 1981). Whether a change in the amino acid sequence of a peptide results in a functional HDx homolog (e.g. functional in the sense that the resulting polypeptide mimics or antagonizes the wild-type form) can be readily determined by assessing the ability of the variant peptide to produce a response in cells in a fashion similar to the wild-type protein, or competitively inhibit such a response. Polypeptides in which more than one replacement has taken place can readily be tested in the same manner.
This invention further contemplates a method for generating sets of combinatorial mutants of the subject HDx proteins as well as truncation mutants, and is especially useful for identifying potential variant sequences (e.g. homologs) that are functional in modulating histone deacetylation. The puφose of screening such combinatorial libraries is to generate, for example, novel HDx homologs which can act as either agonists or antagonist, or alternatively, possess novel activities all together. To illustrate, HDx homologs can be engineered by the present method to provide selective, constitutive activation of enzymatic activity. Thus, combinatorially-derived homologs can be generated to have an increased potency relative to a naturally occurring form of the protein. Likewise, HDx homologs can be generated by the present combinatorial approach to selectively inhibit (antagonize) histone deacetylation. For instance, mutagenesis can provide HDx homologs which are able to bind other regulatory proteins or cytoskeletal elements (or DNA) yet prevent acetylation of histones, e.g. die homologs can be dominant negative mutants. In a preferred embodiment, a dominant negative mutant of an HDx protein is mutated at one or more residues of its catalytic site and/or specificity subsites.
In one aspect of this method, the amino acid sequences for a population of HDx homologs or other related proteins are aligned, preferably to promote the highest homology possible. Such a population of variants can include, for example, HDx homologs from one or more species. Amino acids which appear at each position of the aligned sequences are selected to create a degenerate set of combinatorial sequences. In a preferred embodiment, the variegated library of HDx variants is generated by combinatorial mutagenesis at die nucleic acid level, and is encoded by a variegated gene library. For instance, a mixture of synthetic oligonucleotides can be enzymatically ligated into gene sequences such that the degenerate set of potential HDx sequences are expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g. for phage display) containing the set of HDx sequences therein.
As illustrated in Figure 5B, to analyze the sequences of a population of variants, the amino acid sequences of interest can be aligned relative, to sequence homology. The presence or absence of amino acids from an aligned sequence of a particular variant is relative to a chosen consensus length of a reference sequence, which can be real or artificial. For instance, Figure 5B includes the alignment of the v and χ-motifs for several of the ΗDx gene products. Analysis of the alignment of these sequences from the ΗDx clones can give rise to the generation of a degenerate library of polypeptides comprising potential ΗDx sequences. In an exemplary embodiment, a library of variants based on the HDl sequence, but degenerate across each of the v and χ-motifs can be provided. On such library can be represented by the general formula A-(v motif)-B-(χ motif)-C, wherein the v motif is an amino acid sequence represented in the general formula
DIAX1NWAGGLΗΗAK X2EASGFCιΥNDj^^3X4II^LLKYΗX5RVXYroroi^ 7TO-RVMTVSF the χ motif is an amino acid sequence represented in the general formula
CVEXg VKX9FNX10PLLX1 iLGGGGYTXi 2RNVARCWTYET A corresponds to Metl-Thrl29 of SEQ ID No. 5, B corresponds to Hisl99-Lys283 of SEQ ED No. 5, and C corresponds to Ala317-Ala482 of SEQ ID No. 5, wherein Xi represents He or Val; X2 represents Phe or Ser; X3 represents Phe or Leu; X4 represents Gly or Ala; X5 represents Pro or Gin; Xg represents Gin or Glu; X7 represents Leu or Thr; X represents Val or Tyr; X9 represents Thr or Ser; X10 represents Leu or He; Xj 1 represents Met or Val; and X12 represents He or Val. To further expand the combinatorial set, other conservative mutations relative to those appearing in the human sequences can be provided. For example, in a more expansive library, Xi represents Gly, Ala, Val, He or Leu; X2 represents Phe, Tyr, Thr or Ser; X3 represents Phe, Tyr, Gly, Ala, Val, He or Leu; X represents Gly, Ala, Val, He or Leu; X5 represents Pro, Asn or Gin; X represents .Asn, Gin, Asp or Glu; X7 represents Gly, Ala, Val, He, Leu, Ser or Thr; X8 represents Gly, Ala, Val, He, Leu, Phe or Tyr; X9 represents Thr, Cys, or Ser; X10 represents Gly, Ala, Val, He or Leu; X11 represents Met, Cys, Gly, Ala, Val, He, Leu, Ser or Thr; and X12 represents Gly, Ala, Val, He or Leu. In still another library, each degenerate position can be any one of the naturally occurring amino acids. Likewise, the v and χ-motifs can correspond to the degenerate sequences designated by SEQ ID Nos. 12 and 14, respectively.
There are many ways by which such libraries of potential HDx homologs can be generated from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be carried out in an automatic DNA synthesizer, and the synthetic genes then ligated into an appropriate expression vector. The puφose of a degenerate set of genes is to provide, in one mixture, all of the sequences encoding the desired set of potential HDx sequences. The synthesis of degenerate oligonucleotides is well known in the art (see for example, Narang, SA (1983) Tetrahedron 39:3; Itakura et al. (1981) Recombinant DNA, Proc 3rd Cleveland Sympos. Macromolecules, ed. AG Walton, Amsterdam: Elsevier pp273-289; Itakura et al. (1984) Annu. Rev. Biochem. 53:323; Itakura et al. (1984) Science 198:1056; Ike et al. (1983) Nucleic Acid Res. 11:477. Such techniques have been employed in the directed evolution of other proteins (see, for example, Scott et al. (1990) Science 249:386-390; Roberts et al. (1992) PNAS 89:2429-2433; Devlin et al. (1990) Science 249: 404-406; Cwirla et al. (1990) PNAS 87: 6378-6382; as well as U.S. Patents Nos. 5,223,409, 5,198,346, and 5,096,815).
Likewise, a library of coding sequence fragments can be provided for an HDx clone in order to generate a variegated population of HDx fragments for screening and subsequent selection of bioactive fragments. A variety of techniques are known in the art for generating such libraries, including chemical synthesis. In one embodiment, a library of coding sequence fragments can be generated by (i) treating a double stranded PCR fragment of an HDx coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule; (ii) denaturing d e double stranded DNA; (iii) renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products; (iv) removing single stranded portions from reformed duplexes by treatment with S 1 nuclease; and (v) ligating the resulting fragment library into an expression vector. By this exemplary method, an expression library can be derived which codes for N-terminal, C-terminal and internal fragments of various sizes.
A wide range of techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a certain property. Such techniques will be generally adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of HDx homologs. The most widely used techniques for screening large gene libraries typically comprises cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates relatively easy isolation of the vector encoding the gene whose product was detected.
In an exemplary embodiment, the library of HDx variants is expressed as a fusion protein on the surface of a viral particle. For instance, in the filamentous phage system, foreign peptide sequences can be expressed on the surface of infectious phage, thereby conferring two significant benefits. First, since these phage can be applied to affinity matrices at very high concentrations, a large number of phage can be screened at one time. Second, since each infectious phage displays the combinatorial gene product on its surface, if a particular phage is recovered from an affinity matrix in low yield, the phage can be amplified by another round of infection. The group of almost identical E. coli filamentous phages M13, fd., and fi are most often used in phage display libraries, as either of the phage gill or gVIII coat proteins can be used to generate fusion proteins widiout disrupting the ultimate packaging of the viral particle (Ladner et al. PCT publication WO 90/02909; Garrard et al., PCT publication WO 92/09690; Marks et al.
(1992) J. Biol. Chem. 267:16007-16010; Griffiths et al. (1993) EMBO J 12:725-734;
Clackson et al. (1991) Nature 352:624-628; and Barbas et al. (1992) PNAS 89:4457-
4461).
For example, the recombinant phage antibody system (RPAS, Pharmacia Catalog number 27-9400-01) can be easily modified for use in expressing and screening HDx combinatorial libraries by panning on glutadiione unmobilized histones/GST fusion proteins or RbAp48/GST fusion protein to enrich for HDx homologs which retain an ability to bind a substrate or regulatory protein. Each of these HDx homologs can subsequently be screened for further biological activities in order to differentiate agonists and antagonists. For example, histone-binding homologs isolated from the combinatorial library can be tested for their enzymatic activity directly, or for their effect on cellular proliferation relative to the wild-type form of the protein.
The invention also provides for reduction of the HDx or RbAp48 or histones proteins to generate mimetics, e.g. peptide or non-peptide agents, which are able to disrupt a biological activity of an HDx polypeptide of the present invention, e.g. as catalytic inhibitor or an inhibitor of protein-protein interactions. Thus, such mutagenic techniques as described above are also useful to map the determinants of the HDx proteins which participate in protein-protein or protein-DNA interactions involved in, for example, interaction of the subject HDx polypeptide with histones, RbAp48 or cytoskeletal elements. To illustrate, the critical residues of a subject HDx polypeptide which are involved in molecular recognition of histones can be determined and used to generate HDx-derived peptidomimetics which competitively inhibit binding of the authentic HDx protein with that moiety. Likewise, residues of a histone or of RbAp48 involved in binding to HDx proteins can be identified, and peptides or peptidomimetics based on such residues can also be used as competitive inhibitors of die interaction of an HDx protein with either of those proteins. By employing, for example, scanning mutagenesis to map die amino acid residues of a protein which is involved in binding other proteins, peptidomimetic compounds can be generated which mimic those residues which facilitate the interaction. Such mimetics may then be used to interfere with the normal function of an HDx protein. For instance, non-hydrolyzable peptide analogs of such residues can be generated using benzodiazepine (e.g., see Freidinger et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), azepine (e.g., see Huffman et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), substituted gamma lactam rings (Garvey et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM Publisher: Leiden, Netiierlands, 1988), keto-methylene pseudopeptides (Ewenson et al. (1986) J Med Chem 29:295; and Ewenson et al. in Peptides: Structure and Function (Proceedings of the 9th American Peptide Symposium) Pierce Chemical Co. Rockland, IL, 1985), β-turn dipeptide cores (Nagai et al. (1985) Tetrahedron Lett 26:647; and Sato et al. (1986) J Chem Soc Perkin Trans 1: 1231), and β-aminoalcohols (Gordon et al. (1985) Biochem Biophys Res Commun 126:419; and Dann et al. (1986) Biochem Biophys Res Commun 134:71).
Another aspect of the invention pertains to an antibody specifically reactive with an HDx protein. For example, by using immunogens derived from an HDx protein, e.g. based on the cDNA sequences, anti-protein/anti-peptide antisera or monoclonal antibodies can be made by standard protocols (See, for example, Antibodies: A Laboratory Manual ed. by Harlow and Lane (Cold Spring Harbor Press: 1988)). A mammal, such as a mouse, a hamster or rabbit can be immunized with an immunogenic form of the peptide (e.g., an HDx polypeptide or an antigenic fragment which is capable of eliciting an antibody response). Techniques for conferring immunogenicity on a protein or peptide include conjugation to carriers or other techniques well known in the art. An immunogenic portion of an HDx protein can be administered in d e presence of adjuvant. The progress of immunization can be monitored by detection of antibody titers in plasma or serum. Standard ELISA or other immunoassays can be used with the immunogen as antigen to assess the levels of antibodies. In a preferred embodiment, the subject antibodies are immunospecific for antigenic determinants of an HDx protein of a organism, such as a mammal, e.g. antigenic determinants of a protein represented by one of SEQ DD Nos: 5-8 or closely related homologs (e.g. at least 85% homologous, preferably at least 90% homologous, and more preferably at least 95% homologous). In yet a further preferred embodiment of the present invention, in order to provide, for example, antibodies which are immuno-selective for discrete HDx homologs, e.g. HDl, the anti-HDx polypeptide antibodies do not substantially cross react (i.e. does not react specifically) with a protein which is, for example, less than 85%, 90% or 95% homologous with the selected HDx. By "not substantially cross react", it is meant that the antibody has a binding affinity for a non-homologous protein which is at least one order of magnitude, more preferably at least 2 orders of magnitude, and even more preferably at least 3 orders of magnitude less than the binding affinity of the antibody for the intended target HDx.
Following immunization of an animal with an antigenic preparation of an HDx polypeptide, anti-HDx antisera can be obtained and, if desired, polyclonal anti-HDx antibodies isolated from the serum. To produce monoclonal antibodies, antibody- producing cells (lymphocytes) can be harvested from an immunized animal and fused by standard somatic cell fusion procedures with immortalizing cells such as myeloma cells to yield hybridoma cells. Such techniques are well known in the art, an include, for example, the hybridoma technique (originally developed by Kohler and Milstein, (1975) Nature, 256: 495-497), the human B cell hybridoma technique (Kozbar et al., (1983) Immunology Today, 4: 72), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., (1985) Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc. pp. 77-96). Hybridoma cells can be screened immunochemically for production of antibodies specifically reactive with an HDx polypeptide of the present invention and monoclonal antibodies isolated from a culture comprising such hybridoma cells.
The term antibody as used herein is intended to include fragments thereof which are also specifically reactive with one of the subject HDx polypeptides. Antibodies can be fragmented using conventional techniques and d e fragments screened for utility in the same manner as described above for whole antibodies. For example, F(ab) fragments can be generated by treating antibody with pepsin. The resulting F(ab)2 fragment can be treated to reduce disulfide bridges to produce Fab fragments. The antibody of the present invention is further intended to include bispecific and chimeric molecules having affinity for an HDx protein conferred by at least one CDR region of the antibody.
Both monoclonal and polyclonal antibodies (Ab) directed against authentic HDx polypeptides, or HDx variants, and antibody fragments such as Fab, F(ab)2, Fv and scFv can be used to block the action of one or more HDx proteins and allow the study of the role of these proteins in, for example, differentiation of tissue. Experiments of this nature can aid in deciphering the role of HDx proteins that may be involved in control of proliferation versus differentiation, e.g., in patterning and tissue formation.
Antibodies which specifically bind HDx epitopes can also be used in immunohistochemical staining of tissue samples in order to evaluate the abundance and pattern of expression of each of the subject HDx polypeptides. Anti-HDx antibodies can be used diagnostically in immuno-precipitation and immuno-blotting to detect and evaluate HDx protein levels in tissue as part of a clinical testing procedure. For instance, such measurements can be useful in predictive valuations of the onset or progression of proliferative or differentiative disorders. Likewise, the ability to monitor HDx protein levels in an individual can allow determination of the efficacy of a given treatment regimen for an individual afflicted with such a disorder. The level of HDx polypeptides may be measured from cells in bodily fluid, such as in samples of cerebral spinal fluid or amniotic fluid, or can be measured in tissue, such as produced by biopsy. Diagnostic assays using anti-HDx antibodies can include, for example, immunoassays designed to aid in early diagnosis of a disorder, particularly ones which are manifest at birth. Diagnostic assays using anti-HDx polypeptide antibodies can also include immunoassays designed to aid in early diagnosis and phenotyping neoplastic or hypeφlastic disorders.
Another application of anti-HDx antibodies of the present invention is in the immunological screening of cDNA libraries constructed in expression vectors such as λ gtl l, λgtl8-23, λZAP, and λORF8. Messenger libraries of this type, having coding sequences inserted in the correct reading frame and orientation, can produce fusion proteins. For instance, λgtl l will produce fusion proteins whose amino termini consist of β-galactosidase amino acid sequences and whose carboxy termini consist of a foreign polypeptide. Antigenic epitopes of an HDx protein, e.g. other orthologs of a particular HDx protein or other paralogs from the same species, can then be detected wid antibodies, as, for example, reacting nitrocellulose filters lifted from infected plates with anti-HDx antibodies. Positive phage detected by this assay can then be isolated from the infected plate. Thus, the presence of HDx homologs can be detected and cloned from other animals, as can alternate isoforms (including splicing variants) from humans. Moreover, the nucleotide sequences determined from the cloning of HDx genes from organisms will further allow for the generation of probes and primers designed for use in identifying and/or cloning HDx homologs in other cell types, e.g. from other tissues, as well as HDx homologs from other organisms. For instance, the present invention also provides a probe/primer comprising a substantially purified oligonucleotide, which oligonucleotide comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least 10 consecutive nucleotides of sense or anti-sense sequence selected from the group consisting of SEQ DD Nos: 1-4 or naturally occurring mutants thereof For instance, primers based on the nucleic acid represented in SEQ ID Nos: 1-4 can be used in PCR reactions to clone HDx homologs. Likewise, probes based on the subject HDx sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins. In preferred embodiments, the probe further comprises a label group attached thereto and able to be detected, e.g. the label group is selected from amongst radioisotopes, fluorescent compounds, enzymes, and enzyme co-factors. Such probes can also be used as a part of a diagnostic test kit for identifying cells or tissue which misexpress an HDx protein, such as by measuring a level of an HDx- encoding nucleic acid in a sample of cells from a patient; e.g. detecting HDx mRNA levels or determining whether a genomic HDx gene has been mutated or deleted. To illustrate, nucleotide probes can be generated from the subject HDx genes which facilitate histological screening of intact tissue and tissue samples for the presence (or absence) of HDx-encoding transcripts. Similar to the diagnostic uses of anti-HDx antibodies, the use of probes directed to HDx messages, or to genomic HDx sequences, can be used for both predictive and therapeutic evaluation of allelic mutations which might be manifest in, for example, neoplastic or hypeφlastic disorders (e.g. unwanted cell growth) or abnormal differentiation of tissue. Used in conjunction with immunoassays as described above, the oligonucleotide probes can help facilitate the determination of the molecular basis for a developmental disorder which may involve some abnormality associated with expression (or lack thereof) of an HDx protein. For instance, variation in polypeptide synthesis can be differentiated from a mutation in a coding sequence.
Accordingly, the present method provides a method for determining if a subject is at risk for a disorder characterized by aberrant cell proliferation and or differentiation. In preferred embodiments, method can be generally characterized as comprising detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion characterized by at least one of (i) an alteration affecting the integrity of a gene encoding an HDx-protein, or (ii) the mis-expression of the HDx gene. To illustrate, such genetic lesions can be detected by ascertaining the existence of at least one of (i) a deletion of one or more nucleotides from an HDx gene, (ii) an addition of one or more nucleotides to an HDx gene, (iii) a substitution of one or more nucleotides of an HDx gene, (iv) a gross chromosomal rearrangement of an HDx gene, (v) a gross alteration in the level of a messenger RNA transcript of an HDx gene, (vii) aberrant modification of an HDx gene, such as of the methylation pattern of the genomic DNA, (vii) the presence of a non-wild type splicing pattern of a messenger RNA transcript of an HDx gene, (viii) a non-wild type level of an HDx-protein, and (ix) inappropriate post-translational modification of an HDx-protein. As set out below, the present invention provides a large number of assay techniques for detecting lesions in an HDx gene, and importantly, provides the ability to discern between different molecular causes underlying HDx-dependent aberrant cell growth, proliferation and/or differentiation. In an exemplary embodiment, tiiere is provided a nucleic acid composition comprising a (purified) oligonucleotide probe including a region of nucleotide sequence which is capable of hybridizing to a sense or antisense sequence of an HDx gene, such as represented by any of SEQ ID Nos: 1-4, or naturally occurring mutants thereof, or 5' or 3' flanking sequences or intronic sequences naturally associated with the subject HDx genes or naturally occurring mutants thereof. The nucleic acid of a cell is rendered accessible for hybridization, the probe is exposed to nucleic acid of the sample, and the hybridization of the probe to the sample nucleic acid is detected. Such techniques can be used to detect lesions at either the genomic or mRNA level, including deletions, substitutions, etc., as well as to determine mRNA transcript levels. In certain embodiments, detection of the lesion comprises utilizing the probe/primer in a polymerase chain reaction (PCR) (see, e.g. U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al. (1988) Science 241:1077-1080; and Nakazawa et al. (1944) PNAS 91:360-364), the later of which can be particularly useful for detecting point mutations in the HDx gene. In a merely illustrative embodiment, the method includes the steps of (i) collecting a sample of cells from a patient, (ii) isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, (iii) contacting the nucleic acid sample with one or more primers which specifically hybridize to an HDx gene under conditions such that hybridization and amplification of the HDx gene (if present) occurs, and (iv) detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample.
In still another embodiment, the level of an HDx-protein can be detected by immunoassay. For instance, the cells of a biopsy sample can be lysed, and the level of an HDx-protein present in the cell can be quantitated by standard immunoassay techniques. In yet another exemplary embodiment, aberrant methylation patterns of an HDx gene can be detected by digesting genomic DNA from a patient sample with one or more restriction endonucleases that are sensitive to methylation and for which recognition sites exist in the HDx gene (including in the flanking and intronic sequences). See, for example, Buiting et al. (1994) Human Mol Genet 3:893-895. Digested DNA is separated by gel electrophoresis, and hybridized with probes derived from, for example, genomic or cDNA sequences. The methylation status of the HDx gene can be determined by comparison of the restriction pattern generated from the sample DNA with that for a standard of known methylation. In yet another aspect of the invention, the subject HDx polypeptides can be used to generate a "two hybrid" assay or an "interaction trap" assay (see, for example, U.S. Patent No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J Biol Chem 268:12046-12054; Bartel et al. (1993) Biotechniques 14:920-924; Iwabuchi et al. (1993) Oncogene 8:1693-1696; and Brent WO94/10300), for isolating coding sequences for other cellular proteins which bind HDxs ("HDx-binding proteins" or "HDx-bp"). Such HDx-binding proteins would likely be involved in the regulation of HDx, e.g., as regulatory subunits or transducers, or be substrates which are regulated by an HDx.
Briefly, the interaction trap relies on reconstituting in vivo a functional transcriptional activator protein from two separate fusion proteins. In particular, the method makes use of chimeric genes which express hybrid proteins. To illustrate, a first hybrid gene comprises die coding sequence for a DNA-binding domain of a transcriptional activator fused in frame to the coding sequence for an HDx polypeptide. The second hybrid protein encodes a transcriptional activation domain fused in frame to a sample gene from a cDNA library. If die bait and sample hybrid proteins are able to interact, e.g., form an HDx-dependent complex, they bring into close proximity the two domains of the transcriptional activator. This proximity is sufficient to cause transcription of a reporter gene which is operably linked to a transcriptional regulatory site responsive to the transcriptional activator, and expression of the reporter gene can be detected and used to score for the interaction of the HDx and sample proteins. Furthermore, by making available purified and recombinant HDx polypeptides, the present invention facilitates the development of assays which can be used to screen for drugs, including HDx homologs, which are either agonists or antagonists of the normal cellular function of the subject HDx polypeptides, or of their role in the pathogenesis of cellular differentiation and/or proliferation and disorders related thereto. Moreover, because we have also identified HDx-related proteins, such as the yeast RPD3 proteins, as histone deacetylases, the present invention further provides drug screening assays for detecting agents which modulate the bioactivity of HDx-related proteins. Such agents, when directed to, for example, fungal HDx-related proteins, can be used in the treatment of various infections. In a general sense, the assay evaluates the ability of a compound to modulate binding between an HDx polypeptide and a molecule, be it protein or DNA, that interacts with the HDx polypeptide. It will be apparent from the following description of exemplary assays that, in place of a human (or other mammalian) HDx protein, the assay can be derived with an HDx-related protein such as RPD3. Likewise, in place of human RbAp48 or Sin3A, other HDx-binding proteins can be used, e.g., other human proteins. Exemplary compounds which can be screened include peptides, nucleic acids, carbohydrates, small organic molecules, and natural product extract libraries, such as isolated from animals, plants, fungus and/or microbes.
It is contemplated that any of the novel interactions described herein could be exploited in a drug screening assay. For example, in one embodiment, the interaction between an HDx protein and RbAp48 can be detected in the presence and the absence of a test compound. In another embodiment, the ability of a compound to modulate the binding of an HDx protein, or HDx-related protein such as die yeast RPD3, with histones can be assessed. The identification of a test compound which influences, for example, ΗD1 catalyzed deacetylation of histones would be useful in the modulation of ΗD1 activity in mammalian cells, while the identification of a test compound which selectively inhibits the yeast RPD3 deacetylase activity would be useful as an antifungal agent. In other embodiments the effect of a test compound on the binding of an HDx protein to other molecules, such as cytoskeletal components, or other proteins identified by the HDx-dependent ITS set out above, could be tested. A variety of assay formats will suffice and, in light of the present inventions, will be comprehended by a skilled artisan.
In a preferred embodiment, assays which employ the subject mammalian HDx proteins can be used to identify compounds that have therapeutic indexes more favorable than sodium butyrate, trapoxin, trichostatin or the like. For instance, trapoxin-like drugs can be identified by the present invention which have enhanced tissue-type or cell-type specificity relative to trapoxin. To illustrate, the subject assays can be used to generate compounds which preferentially inhibit IL-2 mediated proliferation activation of lymphocytes, or inhibit proliferation of certain tumor cells, without substantially interfering with other tissues, e.g. hepatocytes. Likewise, similar assays can be used to identify drugs which inhibit proliferation of yeast cells or other lower eukaryotes, but which have a substantially reduced effect on mammalian cells, thereby improving therapeutic index of the drug as an anti-mycotic agent.
In one embodiment, the identification of such compounds is made possible by the use of differential screening assays which detect and compare drug-mediated inhibition of deacetylase activity between two or more different HDx-like enzymes, or compare drug- mediated inhibition of formation of complexes involving two or more different types of HDx-like proteins. To illustrate, the assay can be designed for side-by-side comparison of the effect of a test compound on the deacetylase activity or protein interactions of tissue-type specific HDx proteins. Given the apparent diversity of HDx proteins, it is probable that different functional HDx activities, or HDx complexes exist and, in certain mstances, are localized to particular tissue or cell types. Thus, test compounds can be screened for agents able to inhibit die tissue-specific formation of only a subset of the possible repertoire of HDx/regulatory protein complexes, or which preferentially inhibit certain HDx enzymes. In an exemplary embodiment, an interaction trap assay can be derived using two or more different human HDx "bait" proteins, while the "fish" protein is constant in each, e.g. a human RbAp48 construct. Running the interaction trap side- by-side permits the detection of agents which have a greater effect (e.g. statistically significant) on the formation of one of the HDx/RbAp48 complexes than on the formation of the other HDx complexes.
In similar fashion, differential screening assays can be used to exploit the difference in protein interactions and/or catalytic mechanism of mammalian HDx proteins and yeast RPD3 proteins in order to identify agents which display a statistically significant increase in specificity for inhibiting the yeast enzyme relative to the mammalian enzyme. Thus, lead compounds which act specifically on pathogens, such as fungus involved in mycotic infections, can be developed. By way of illustration, the present assays can be used to screen for agents which may ultimately be useful for inhibiting at least one fungus implicated in such mycosis as candidiasis, aspergillosis, mucormycosis, blastomycosis, geotrichosis, cryptococcosis, chromoblastomycosis, coccidioidomycosis, conidiosporosis, histoplasmosis, maduromycosis, rhinosporidosis, nocaidiosis, para-actinomycosis, penicilliosis, monoliasis, or sporotrichosis. For example, if the mycotic infection to which treatment is desired is candidiasis, the present assay can comprise comparing the relative effectiveness of a test compound on inhibiting the deacetylase activity of a mammalian HDx protein with its effectiveness towards inhibiting the deacetylase activity of an RPD3 homolog cloned from yeast selected from the group consisting of Candida albicans, Candida stellatoidea, Candida tropicalis, Canώda parapsilosis, Candida krusei, Candida pseudotropicalis, Candida quillermondii, or Candida rugosa. Likewise, the present assay can be used to identify anti-fungal agents which may have therapeutic value in the treatment of aspergillosis by selectively targeting RPD3 homologs cloned from yeast such as Aspergillus fumigatus, Aspergillus flavus, Aspergillus niger, Aspergillus nidulans, or Aspergillus terreus. Where the mycotic infection is mucormycosis, the RPD3 deacetylase can be derived from yeast such as Rhizopus arrhizus, Rhizopus oryzae, Absidia corymbifera, Absidia ramosa, or Mucor pusillus. Sources of other RPD3 activities for comparison with a mammalian HDx activity includes the pathogen Pneumocystis carinii.
In addition to such HDx therapeutic uses, anti-fungal agents developed with such differential screening assays can be used, for example, as preservatives in foodstuff, feed supplement for promoting weight gain in livestock, or in disinfectant formulations for treatment of non-living matter, e.g., for decontaminating hospital equipment and rooms.
In similar fashion, side by side comparison of inhibition of a mammalian HDx proteins and an insect HDx-related proteins, will permit selection of HDx inhibitors which discriminate between the human/mammalian and insect enzymes. Accordingly, the present invention expressly contemplates the use and formulations of the subject HDx therapeutics in insecticides, such as for use in management of insects like the fruit fly.
In yet another embodiment, certain of the subject HDx inhibitors can be selected on the basis of inhibitory specificity for plant HDx-related activities relative to the mammalian enzyme. For example, a plant HDx-related protein can be disposed in a differential screen with one or more of the human enzymes to select those compounds of greatest selectivity for inhibiting the plant enzyme. Thus, the present invention specifically contemplates formulations of the subject HDx inhibitors for agricultural applications, such as in the form of a defoliant or the like. In many drug screening programs which test libraries of compounds and natural extracts, high throughput assays are desirable in order to maximize the number of compounds surveyed in a given period of time. Assays which are performed in cell-free systems, such as may be derived with purified or semi-purified proteins, are often preferred as "primary" screens in that they can be generated to permit rapid development and relatively easy detection of an alteration in a molecular target which is mediated by a test compound. Moreover, the effects of cellular toxicity and/or bioavailability of the test compound can be generally ignored in the in vitro system, the assay instead being focused primarily on the effect of the drug on the molecular target as may be manifest in an alteration of binding affinity with upstream or downstream elements. Accordingly, in an exemplary screening assay of the present invention, a reaction mixture is generated to include an HDx polypeptide, compound(s) of interest, and a "target polypeptide", e.g., a protein, which interacts with the HDx polypeptide, whether as a substrate or by some other protein-protein interaction. Exemplary target polypeptides include histones, RbAp48 polypeptides, Sin3 polypeptides, and or combinantions thereof or with other transciptional regulatory proteins (such as myc, max, etc, see Example 3)). Detection and quantification of complexes containing the HDx protein provide a means for deterrnining a compound's efficacy at inhibiting (or potentiating) complex formation between the HDx and the target polypeptide. The efficacy of the compound can be assessed by generating dose response curves from data obtained using various concentrations of the test compound. Moreover, a control assay can also be performed to provide a baseline for comparison. In the control assay, isolated and purified HDx polypeptide is added to a composition containing the target polypeptide and the formation of a complex is quantitated in the absence of the test compound.
Complex formation between the HDx polypeptide and the target polypeptide may be detected by a variety of techniques. Modulation of the formation of complexes can be quantitated using, for example, detectably labeled proteins such as radiolabeled, fluorescently labeled, or enzymatically labeled HDx polypeptides, by immunoassay, by chromatographic detection, or by detecting the intrinsic activity of the acetylase.
Typically, it will be desirable to immobilize either HDx or the target polypeptide to facilitate separation of complexes from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Binding of HDx to the target polypeptide, in die presence and absence of a candidate agent, can be accomplished in any vessel suitable for containing the reactants. Examples include microtitre plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided which adds a domain that allows the protein to be bound to a matrix. For example, glutathione-S-transferase/HDx (GST/HDx) fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtitre plates, which are then combined with die cell lysates, e.g. an 35S-labeled, and the test compound, and the mixture incubated under conditions conducive to complex formation, e.g. at physiological conditions for salt and pΗ, though slightly more stringent conditions may be desired. Following incubation, the beads are washed to remove any unbound label, and the matrix immobilized and radiolabel determined directly (e.g. beads placed in scintillant), or in the supernatant after the complexes are subsequently dissociated. Alternatively, the complexes can be dissociated from the matrix, separated by SDS-PAGE, and the level of HDx-binding protein found in the bead fraction quantitated from the gel using standard electrophoretic techniques such as described in the appended examples.
Other techniques for immobilizing proteins on matrices are also available for use in the subject assay. For instance, either HDx or target polypeptide can be immobilized utilizing conjugation of biotin and streptavidin. For instance, biotinylated HDx molecules can be prepared from biotin-NΗS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with HDx, but which do not interfere with the interaction between the HDx and target polypeptide, can be derivatized to the wells of the plate, and HDx trapped in the wells by antibody conjugation. As above, preparations of an target polypeptide and a test compound are incubated in the HDx-presenting wells of the plate, and the amount of complex trapped in the well can be quantitated. Exemplary methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the target polypeptide, or which are reactive with HDx protein and compete with the target polypeptide; as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the target polypeptide, either intrinsic or extrinsic activity. In the instance of the latter, the enzyme can be chemically conjugated or provided as a fusion protein with the target polypeptide. To illustrate, the target polypeptide can be chemically cross-linked or genetically fused with horseradish peroxidase, and the amount of polypeptide trapped in the complex can be assessed with a chromogenic substrate of the enzyme, e.g. 3,3'-diamino-benzadine terahydrochloride or 4-chloro-l-napthol. Likewise, a fusion protein comprising the polypeptide and glutathione-S-transferase can be provided, and complex formation quantitated by detecting the GST activity using 1- chloro-2,4-dinitrobenzene (Ηabig et al (1974) J Biol Chem 249:7130).
For processes which rely on immunodetection for quantitating one of the proteins trapped in the complex, antibodies against the protein, such as anti-HDx antibodies, can be used. Altematively, the protein to be detected in the complex can be "epitope tagged" in the form of a fusion protein which includes, in addition to the HDx sequence, a second polypeptide for which antibodies are readily available (e.g. from commercial sources). For instance, the GST fusion proteins described above can also be used for quantification of binding using antibodies against the GST moiety. Other useful epitope tags include myc-epitopes (e.g., see Ellison et al. (1991) J Biol Chem 266:21150-21157) which includes a 10-residue sequence from c-myc, as well as the pFLAG system (Intemational Biotechnologies, Inc.) or the pEZZ-protein A system (Pharamacia, NJ).
In another embodiment of a drug screening, a two hybrid assay can be generated with an HDx and HDx-binding protein. Drug dependent inhibition or potentiation of the interaction can be scored. Where the HDx proteins themselves, or in complexes with other proteins, are capable of binding DNA and modifying transcription of a gene, a transcriptional based assay using, for example, an transcriptional regulatory sequences responsive to HDx complexes operably linked to a detectable marker gene. For illustration, see Example 3.
To test the effect of a histone deacetylase inhibitor on MadN35GALVP16 and Mad(Pro)N35GALVPI6 mediated repression, we treated a duplicate set of transfections with 10 nM trapoxin for eight hours prior to harvest. In the representative experiment shown, 10 nM trapoxin treatment derepressed the activity of MadN35GALVP16 nine¬ fold while it had little effect on the activity of Mad(Pro)N35GALVP16, suggesting that die histone deacetylation plays a direct role in mSin3A transcriptional repression (Figure 13B). In addition, there was typically less than a two-fold effect of trapoxin on the activity of the reporter cene in cells transfected with the expression vector alone or in cells transfected with GALVP16 (data not shown). Following trapoxin treatment, the repression observed for MadN35GALVP16 was still seven times greater than that of Mad(Pro)N35GALVP16, suggesting that the residual deacetylase activity following trapoxin treatment (Figure 13B) continues to drive mSin3A-mediated repression; however, we can not rule out that mSin3A is capable of repression by mechanisms independent of histone deacetylation.
Furthermore, each of the assay systems set out above can be generated in a
"differential" format as set forth above. That is, the assay format can provide information regarding specificity as well as potency. For instance, side-by-side comparison of a test compound's effect on different HDxs can provide information on selectivity, and permit the identification of compounds which selectively modulate the bioactivity of only a subset of the HDx family.
Furthermore, inhibitors of the enzymatic activity of each of the subject HDx proteins can be identified using assays derived from measuring the ability of an agent to inhibit catalytic conversion of a substrate by the subject proteins. For example, the ability of the subject HDx proteins to deacetylate a histone substrate, such as histone Η4 (see examples), in the presence and absence of a candidate inhibitor, can be determined using standard enzymatic assays.
A number of methods have been employed in the art for assaying histone deacetylase activity, and can be incoφorated in the drug screening assays of the present invention. In preferred embodiments, the assay will employ a labeled acetyl group linked to appropriate histone lysine residues as substrates. In other embodiments, a histone substrate peptide can be labeled with a group whose signal is dependent on the simultaneous presence or absence of an acetyl group, e.g., the label can be a fluorogenic group whose fluorescence is modulated (either quenched or potentiated) by the presence of the acetyl moiety. Using standard enzymatic analysis, the ability of a test agent to cause a statistically significant change in substrate conversion by a histone deacetylase can be measured, and as desirable, inhibition constants, e.g., Kj values, can be calculated. The histone substrate can be provided as a purified or semi-purified polypeptide or as part of a cell lysate. Likewise, the histone deacetylase can be provided to the reaction mixture as a purified or semi-purified polypeptide or as a cell lysate. Accordingly, the reaction mixtures of the subject method can range from reconstituted protein mixtures derived with purified preparations of histones and deacetylases, to rnixtures of cell lysates, e.g., by admixing baculovirus lysates containing recombinant histones and deacetylases. In an exemplary embodiment, the histone substrate for the subject assay is provided by isolation of radiolabeled histones from metabolically labelled cells. To illustrate, as described by Hay et al. (1983) JBiol Chem 258:3726-3734, HeLa cells can be labelled in culture by addition of [3H] acetate (New England Nuclear) to the culture media. The addition of butyrate, trapoxin or the like can be used to increase the abundance of acetylated histones in the cells. Radiolabelled histones can be isolated from the cells by extraction with HsSO4 (Marushige et al. (1966) J Mol Biol 15:160-174). Briefly, cells are homogenized in buffer, centrifuged to isolate a nuclear pellet, the subsequently homogenized nuclear pellet centrifuged through sucrose, and die resulting chromatin pellet extracted by addition of H-SO4 to yield [3H]acetyl-labelled histones. In an alternate embodiment, nucleosome preparations containing [3H]acetyl-labelled histones can be isolated from the labelled cells. As described in the art, nucleosomes can be isolated from cell preparations by sucrose gradient centrifugation (Hay et al. (1983) J Biol Chem 258:3726-3734; and Noll (1967) Nature 215:360-363), and polynucleosomes can be prepared by NaCl precipitation from micrococcal nuclease digested cells (Hay et al., supra). Similar procedures for isolating labelled histones from odier cells types, including yeast, have been described. See, for example, Alonso et al. (1986) Biochem Biophys Acta 866:161-169; and Kreiger et al. (1974) JBiol Chem 249:332-334. In yet odier embodiments, the histone is generated by recombinant gene expression, and includes an exogenous tag (e.g., an HA epitope, a poly(his) sequence or the like) which facilitates in purification from cell extracts. In still other embodiments, whole nuclei can be isolated from metabolically labelled cells by micrococcal nuclease digestion (Hay et al., supra)
In still another embodiment, the deacetylase substrate can be provided as an acetylated peptide including a sequence corresponding to the sequence about the specific lysyl residues acetylated on histone, e.g., a peptidyl portions of the core histones H A, H2B, H3 or H4. Such fragments can be produced by cleavage of acetylated histones derived from metabolically labelled cells, e.g., such as by treatment with proteolytic enzymes or cyanogen bromide (Kreiger et al., supra). In other embodiments, the acetylated peptide can be provided by standard solid phase synthesis using acetylated lysine residues (Kreiger et al., supra). Continuing with the illustrative use of [3H]acetyl-labelled histones, the activity of a histone deacetylase in the subject assays is detected by measuring release of [3H]acetate by standard scintillant techniques. In a merely illustrative example, a reaction mixture is provided which comprises a recombinant HDx protein suspended in buffer, along with a sample of [3H]acetyl-labelled histones and (optionally) a test compound. The reaction mixture is maintained at a desired temperature and pH, such as 22°C at pH7.8, for several hours, and the reaction terminated by boiling or other form of denaturation. Released [3H]acetate is extracted and counted. For example, the quenched reaction mixture can be acidified with concentrated HCl, and used to create a biphasic mixture with ethyl acetate. The resulting 2 phase system is thoroughly mixed, centrifuged, and the ethyl acetate phase collected and counted by standard scintillation methods. Other methods for detecting acetate release will be easily recognized by those skilled in the art.
In yet another embodiment, the drug screening assay is derived to include a whole cell recombinantly expressing one or more of a target protein or HDx protein. The ability of a test agent to alter the activity of the HDx protein can be detected by analysis of the recombinant cell. For example, agonists and antagonists of the HDx biological activity can by detected by scoring for alterations in growth or differentiation (phenotype) of the cell. General techniques for detecting each are well known, and will vary with respect to the source of the particular reagent cell utilized in any given assay. For example, quantification of proliferation of cells in the presence and absence of a candidate agent can be measured with a number of techniques well known in the art, including simple measurement of population growth curves. For instance, where the assay involves proliferation in a liquid medium, turbidimetric techniques (i.e. absorbence/ transmittance of light of a given wavelength through the sample) can be utilized. For example, in the instance where the reagent cell is a yeast cell, measurement of absorbence of light at a wavelength between 540 and 600nm can provide a conveniently fast measure of cell growth. Likewise, ability to form colonies in solid medium (e.g. agar) can be used to readily score for proliferation. In other embodiments, an HDx substrate protein, such as a histone, can be provided as a fusion protein which permits the substrate to be isolated from cell lysates and the degree of acetylation detected. Each of these techniques are suitable for high through-put analysis necessary for rapid screening of large numbers of candidate agents.
In addition, where the ability of an agent to cause or reverse a transformed phenotype, growth in solid media such as agar can further aid in establishing whether a mammalian cell is transformed. Additionally, visual inspection of the moφhology of the reagent cell can be used to determine whether the biological activity of the targeted HDx protein has been affected by the added agent. To illustrate, the ability of an agent to influence an apoptotic phenotype which is mediated in some way by a recombinant HDx protein can be assessed by visual microscopy. Likewise, the formation of certain cellular structures as part of differentiation, such as the formation of neuritic process, can be visualized under a light microscope.
The nature of the effect of test agent on reagent cell can be assessed by measuring levels of expression of specific genes, e.g., by reverse transcription-PCR. Another method of scoring for effect on Hdx activity is by detecting cell-type specific marker expression through immunofluorescent staining. Many such markers are known in the art, and antibodies are readily available. For example, the presence of chondroitin sulphate proteoglycans as well as type-II collagen are correlated with cartilage production in chondrocytes, and each can be detected by immunostaining. Similarly, the human kidney differentiation antigen gpl60, human aminopeptidase A, is a marker of kidney induction, and the cytoskeletal protein troponin I is a marker of heart induction. In yet another embodiment, the alteration of expression of a reporter gene construct provided in the reagent cell provides a means of detecting the effect on HDx activity. For example, reporter gene constructs derived using the transcriptional regulatory sequences, e.g. the promoters, for developmentally regulated genes can be used to drive the expression of a detectable marker, such as a luciferase gene. In an illustrative embodiment, the construct is derived using the promoter sequence from a gene expressed in a particular differentiative phenotype.
It is also deemed to be within die scope of this invention that the recombinant HDx cells of the present assay can be generated so as to comprise heterologous HDx proteins (i.e. cross-species expression). For example, HDx proteins from one species can be expressed in the cells of another under conditions wherein the heterologous protein is able to rescue loss-of-function mutations in the host cell. For example, the reagent cell can be a yeast cell in which a human MDx protein (e.g. exogenously expressed) is the intended target for development of an anti-proliferative agent. To illustrate, the M778 strain, MATa ura3-52 trplΔl his3-200 leu2-l trklΔ rpd3Δ::HIS3, described by Vidal et al. (1991) Mol Cell Biol 6317-6327, which lacks a functional endogenous RPD3 gene can be transfected with an expression plasmid including a mammalian HDx gene in order to complement the RPD3 loss-of-function. For example, the coding sequence for HDl can be cloned into a pRS integrative plasmid containing a selectable marker (Sikorski et al. (1989) Genetics 122:19-27), and resulting construct used to transform the M778 strain. The resulting cells should produce a mammalian HDl protein which may be capable performing at least some of the functions of the yeast RPD3 protein. The HDx transformed yeast cells can be easier to manipulate than mammalian cells, and can provide access to certain assay formats, such as turbidity detection methods, which may not be obtainable with mammalian cells.
Moreover, the combination of the "mammalianized" strain with the strain M537 (MATa ura3-52 trplΔl his3-200 leu2-l trklΔ, Vidal et al., supra) can provide an exquisitely sensitive cell-based assay for detecting agent which specifically inhibit, for example, the yeast RPD3 deacetylase.
In another aspect, the invention provides compounds useful for inhibition of HDxs. In a preferred embodiment, an HDx inhibitor compound of the invention can be represented by the formula A-B-C, in which A is a specificity element for selective binding to an HDx, B is a linker element, and C is an electrophilic moiety capable of reacting with a nucleophilic moiety of an HDx; with the proviso that the compound is not butyrate, trapoxin, or trichostatin.
In another aspect, the invention provides an affinity matrix for binding or purifying an HDx. In a preferred embodiment, the affinity matrix can be represented by the formula S-A-B-C, in which S is a solid or insoluble support, and A, B, and C are as described above. The solid or insoluble support S can be any of a variety of supports, many of which are known in the art, for synthesis of, or immobilization of, compounds, e.g., peptides, benzodiazepines, and the like. For a review of solid-supported synthesis, see, e.g., Ηodge et αl., Polymer-supported Reactions in Organic Synthesis, John Wiley & Sons, New York, 1980. The HDx inhibitor moiety A-B-C can be bonded directly to the support S, or can be bonded to the support S through a linking or spacing moiety, as is known in the art.
In another aspect, the invention provides a method of inhibiting an HDx. The method comprises contacting the HDx with a compound capable of inhibiting HDx activity, under conditions such that HDx activity is inhibited. In preferred embodiments, the compounds can be represented by the formula A-B-C, in which A, B, and C are as described above; with the proviso that the compound is not butyrate, trapoxin, or trichostatin.
In another aspect, the invention provides a method of purifying an HDx. The method includes contacting a reaction mixture comprising an HDx with an affinity matrix capable of selectively binding to an HDx, and separating at least one odier component of the reaction mixture from the HDx. In a preferred embodiment, the affinity matrix can be represented by the formula S-A-B-C, in which S, A, B, and C are as described above.
In general, the elements A, B, and C of the inhibitor compounds are selected to permit selective binding to, and inhibition of, at least one HDx. The elements A, B, and C can be selected to provide specificity for particular HDxs. For example, a series of candidate HDx inhibitor compounds can be synthesized, e.g., according to the combinatorial methods described infra, and the library of candidate compounds screened against one or more HDxs to determine the compound or compounds with optimal activity and specificity for a particular HDx.
Thus, in preferred embodiments, the specificity element A is selected such that the HDx inhibitor compound binds selectively to an HDx. In general, the specificity element A will be selected according to factors such as the binding specificity of the HDx or HDxs to which the inhibitor compound should bind, ease of synthesis, stability in vivo or in vitro, and the like. In certain embodiments, the specificity element A is a cyclotetrapeptidyl moiety. In another embodiment, A is a substituted or unsubstituted aryl moiety. In yet another embodiment, A is a nonaromatic carbocycle. In still another embodiment, A is an amino acyl moiety (e.g., a natural or non-natural amino acyl moiety). In yet another embodiment, A is a heterocyclyl moiety. In preferred embodiments, B is selected from the group consisting of substituted and unsubstituted C -C alkylidene, C -C alkenylidene, C4-C alkynylidene, and D-E-F, in which D and F are independently absent or C -C7 alkylidene, C -C7 alkenylidene, or C2-C7 alkynylidene, and E is O, S, or NR', in which R' is Η, lower alkyl, lower alkenyl, lower alkynyl, aralkyl, aryl, or heterocyclyl. The element B should be selected to permit the specificity element A to interact with an HDx such that specific binding occurs, while poising the electrophilic moiety C for reaction with a nucleophilic moiety of the HDx.
In a preferred embodiment, C is an electrophilic moiety that is approximately isosteric with an N-acetyl group (i.e., C has approximately the same steric bulk as an N- acetyl group)In preferred embodiments, the element C is capable of reacting, covalently or non-covalently, with a nucleophilic moiety of an HDx. In certain preferred embodiments, the element C is capable of binding (e.g., by chelation) to a metal ion, e.g., a divalent metal ion, e.g., zinc or calcium. In preferred embodiments, C is selected from die group consisting of α,β-epoxyketones, α,β-epoxythioketones, α,β-epoxysuIfoxides, hydroxamic acids, α-haloketones, α-halothioketones, α-diazoketones, α- diazothioketones, vinyl epoxides, trifluoromethylketone, trifluoromediylthioketone, enones (e.g., of ketones or thioketones), ynones (e.g., of ketones or thioketones), α,β- aziridinoketones, hydrazones, boronic acids, carboxylates, amides (e.g., -C(O)-amino), sulfones, aldehyde, alkyl halides, epoxides, and the like. In accordance with die foregoing, the moieties A, B, and C can illustratively be represented by the formulas depicted in Figure 6, in which Ri represents one or more substituents selected from the group consisting of amino, halogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, heterocyclyl, azido, carboxyl, alkoxycarbonyl, hydroxyl, alkoxy, cyano, trifluoromethyl, and the like; R" is Cj-C8 alkylidene, C2-C8 alkenylidene, or C2-C8 alkynylidene; R5 is hydrogen, alkyl, alkoxycarbonyl, aryloxycarbonyl, alkylsulfonyl, arylsulfonyl or aryl; R^ is hydrogen, alkyl, aryl, alkoxy, aryloxy, halogen, and the like; R'6 is hydrogen, alkyl, alkenyl, alkynyl, aryl, and the like; R7 is hydrogen, alkyl, aryl, alkoxy, aryloxy, amino, hydroxylamino, alkoxylamino, halogen, and the like; R is hydrogen, alkyl, halogen, and the like; R9 is hydrogen, alkyl, aryl, hydroxyl, alkoxy, aryloxy, amino, and the like; X is a good leaving group, e.g., diazo, halogen, a sulfate or sulfonate ester, e.g., a tosylate or mesylate, and the like; and Y is O or S.
In certain preferred embodiments, an HDx inhibitor compound can be represented by the formula A-B-C, in which A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclotetrapeptides; B is selected from the group consisting of substituted and unsubstituted C -C alkylidene, C -C8 alkenylidene, C4-C8 alkynylidene, C -C8 enyne, and D-E-F, in which D and F are independently absent or a C-C7 alkylidene, an C2-C7 alkenylidene, or an C2-C alkynylidene, and E is O, S, or NR', in which R' represents Η, a lower alkyl, a lower alkenyl, a lower alkynyl, an aralkyl, aryl, or a heterocyclyl; and C is selected from the group consisting of
Figure imgf000062_0001
and B(OΗ)2 (boronic acid); in which Z represents O, S, or NR5, and Y, R5, R'6, and R7 are as defined above.
In preferred embodiments, R'g is hydrogen. In certain preferred embodiments, B is not a
C4-C alkylidene. In preferred embodiments, if B is a C4-C alkylidene, C is not a boronic acid. In other preferred embodiments, the inhibitor compound is not trapoxin.
In certain preferred embodiments, an HDx inhibitor compound can be represented by the formula A-B-C, in which A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclotetrapeptides; B is selected from the group consisting of substituted and unsubstituted C4-C8 alkylidene, C -C8 alkenylidene, C4-C8 alkynylidene, C -C8 enyne, and D-E-F, in which D and F are independently absent or C1-C7 alkylidene, C2-C7 alkenylidene, or C2-C7 alkynylidene, and E is O, S, or NR', in which R' represents H, a lower alkyl, a lower alkenyl, a lower alkynyl, an aralkyl, an aryl, or a heterocyclyl; and C is selected from the group consisting of
Figure imgf000063_0001
embodiments, B is not a C -C8 alkylidene. In preferred embodiments, the inhibitor compound is not trichostatin.
In still another preferred embodiment, an HDx inhibitor compound can be represented by the formula A-B-C, in which A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclotetrapeptides; B is selected from the group consisting of substituted and unsubstituted C4-C alkylidene, C4-C alkenylidene, C4-C8 alkynylidene, C4-C enyne, and D-E-F, in which D and F are independently absent or a C1-C7 alkylidene, a C2-G7 alkenylidene, or a C2-C7 alkynylidene, and E is O, S, or NR', in which R' is H, lower
alkyl, lower alkenyl, lower alkynyl, aralkyl, aryl, or heterocyclyl; and C is
Figure imgf000063_0002
; in which Y is O or S, and R7 is as defined above. Certain HDx inhibitor compounds of the present invention may exist in particular geometric or stereoisomeric forms. For example, amino acids can contain at least one chiral center. The present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
If, for instance, a particular enantiomer of a compound of die present invention is desired, it may be prepared by asymmetric synthesis, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomer. Altematively, where the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts can be formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers.
The term "alkyl" refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. In preferred embodiments, a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C1-C30 for straight chain, C3-C30 for branched chain), and more preferably 20 or fewer. Likewise, preferred cycloalkyls have from 4-10 carbon atoms in their ring structure, and more preferably have 5, 6 or 7 carbons in the ring structure.
Unless the number of carbons is otherwise specified, "lower alkyl" as used herein means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six carton atoms in its backbone structure. Likewise, "lower alkenyl" and "lower alkynyl" have similar chain lengths. Preferred alkyl groups are lower alkyls. In preferred embodiments, a substituent designated herein as alkyl is a lower alkyl. Moreover, the term "alkyl" (or "lower alkyl") as used throughout the specification and claims is intended to include both "unsubstituted alkyls" and "substituted alkyls", the latter of which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents can include, for example, halogen, hydroxyl, carbonyl (such as a carboxylate, alkoxycarbonyl, aryloxycarbonyl, alkylcarbonyl, arylcarbonyl, aldehyde, and the like), thiocarbonyl (such as a thioacid, alkoxycarbonyl, and the like), an alkoxyl, unsubstituted amino, mono- or disubstituted amino, amido, amidine, imine, nitro, azido, sulfhydryl, alkylthio, cyano, trifluoromethyl, sulfonato, sulfamoyl, sulfonamido, heterocyclyl, aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, as described above, if appropriate. Exemplary substituted alkyls are described below. Cycloalkyls can be further substituted with, e.g., alkyls, alkenyls, alkoxys, alkylthios, aminoalkyls, carbonyl-substituted alkyls, - CF3, -CN, and the like. The terms "alkenyl" and "alkynyl" refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively. The term "enyne" refers to an unsaturated aliphatic moiety having at least one double bond and one triple bond. The terms "alkylidene," "alkenylidene," and "alkynylidene" are art-recognized and refer to moieties corresponding to alkyl, alkenyl, and alkynyl moieties as defined above, but having two valences available for bonding.
The term "aryl" as used herein includes 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, phenyl, pyrrolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, thiazolyl, triazolyl, pyrazolyl, pyridyl, pyrazinyl, pyridazinyl and pyrimidyl, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles" or "heteroaromatics". The aromatic ring can be substituted at one or more ring positions with such substituents as described above, as for example, halogen, azido, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, -CF3, -CN, or the like.
The term "aralkyl", as used herein, refers to an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group). The terms "heterocyclyl" or "heterocyclic group" refer to non-aromatic 4- to 10- membered ring structures, more preferably 4- to 7-membered rings, which ring structures include one to four heteroatoms (e.g., O, N, S, P and the like). Heterocyclyl groups include, for example, pyrrolidine, oxolane, thiolane, imidazole, oxazole, piperidine, piperazine, moφholine, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like. The heterocyclic ring can be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, alkoxycarbonyl, aryloxycarbonyl, carboxyl, silyl, ether, alkylthio, alkylsulfonyl, arylsulfonyl, ketone (e.g., -C(O)-alkyl or -C(O)-aryl), aldehyde, heterocyclyl, an aryl or heteroaryl moiety, -CF3, -CN, or the like.
Compounds represented by the formula A-B-C, in which A, B, and C have the values described supra, can be synthesized by standard techniques of organic synthesis. For example, precursor synthons corresponding to each of the moieties A, B, and C, or subunits thereof, can be coupled in linear or convergent syntheses to provide HDx inhibitor compounds, or compounds readily converted thereto. Syntheses of the HDx inhibitor compound trichostatin, and related compounds, have been reported; see, e.g., Massa, S. et al. (1990) J. Med Chem. 33:2845-49; Mori, K., and Kosecki, K. (1988) Tetrahedron 44:6013-20; Koseki, K. and Mori, K. European Patent Application EP 331524 A2; Fleming, I. et al. (1983) Tetrahedron 39:841-46. Analogs of trapoxin have also been synthesized; see, e.g., Yoshida, Η. and Sugita, K. (1992) Jpn. J. Cancer Res. 83:324-28.
Thus, in an illustrative synthesis, a compound represented by the formula A-B-C, in which A is an phenyl group, while B and C can have a variety of values, can be synthesized as shown below:
Figure imgf000066_0001
B
Figure imgf000066_0004
Figure imgf000066_0002
Figure imgf000066_0003
Figure imgf000066_0005
3
Scheme I
According to the Scheme, a functionalized organometallic aryl compound (MX=organometallic moiety; R is any substituent; X is a leaving group, e.g., halogen) (e.g., organotin, boronate, aryllithium, cuprate, Grignard reagent, etc.) is alkylated or acylated to provide functionalized compounds (e.g., the exemplary compounds 1, 2, or 3) which can be further elaborated to provide compounds with a wide variety of substituents and carbon backbones. Other A moieties (e.g., specificity elements) can be obtained by use of appropriate synthons, e.g., by substituting vinylorganometallic compounds for the organometallic aryl compound of the Scheme (followed by further treatment, e.g., reduction, of the vinyl group, if desired, to yield an alkyl A moiety). By way of illustration, as shown for compound I, the carbonyl group can be used for elaboration, e.g., by reduction of the carbonyl group to an alcohol, conversion of the alcohol to a tosylate, and nucleophilic displacement of the tosylate by an acyl compound (e.g., a ketone or ester) to provide a chain-lengthened product (Route A), which can be converted to a C(O)X functionality (e.g., by hydrolysis of an ester and conversion of die resulting carboxylic acid to an acid chloride). Altematively, the carbonyl group of I can be used for olefination (Route B), e.g., Homer-Emmons olefination, to provide an elaborated alkenyl compound. Also, the carbonyl group can be converted to an alkynyl functionality, e.g., via the Corey-Fuchs procedure, to provide an elaborated alkynyl compound. For puφoses of clarity, only certain chain lengths and functional group patterns are shown in the scheme; however, the skilled artisan will appreciate that many other compounds, with a variety of B moieties (i.e., linking moieties), can be synthesized through analogous procedures. The C(O)X functionality (e.g., an acid chloride where X is Cl) can be converted to functional groups such as amide, hydrazido, trifluormethylketone, enone, epoxide, aziridine, and the like, through methods conventional in the art. Thus, the synthetic pathways shown in the Scheme provide access to compounds having a variety of C moieties (e.g., reactive moieties) suitable for substitution in the subject HDx inhibitors.
In vitro chemical synthesis provides a method for generating libraries of compounds that can be screened for ability to bind to or inhibit a target protein, e.g., an
HDx. Although in vitro methods have previously been used in the pharmaceutical industry to identify potential drugs, recently developed methods have focused on rapidly and efficiently generating and screening large numbers of compounds and are amenable to generating HDx inhibitor compound libraries for use in the subject method. The various approaches to simultaneous preparation and analysis of large numbers of compounds
(herein "combinatorial synthesis") each rely on the fundamental concept of synthesis on a solid support introduced for peptides by Merrifield in 1963 (Merrifield, R.B. (1963) J Am
Chem Soc 85:2149-2154). Many types of solid matrices have been successfully used in solid-phase synthesis, and can be selected according to the type of chemistry to be performed on the immobilized moieties, as is discussed in more detail below. Several synthetic schemes have been suggested or employed for the combinatorial synthesis of organic compounds (see, e.g., E.M. Gordon et al, J. Med Chem. 37:1385- 1401 (1994)).
Multipin Synthesis
One method for combinatorial synthesis of compounds is die multipin synthesis method. Briefly, Geysen and co-workers (Geysen et al. (1984) PNAS 81:3998-4002) introduced a method for generating compounds by a parallel synthesis on polyacrylic acid-grated polyethylene pins arrayed in the microtitre plate format. In the original experiments, about 50 nmol of a single compound was covalently linked to the spherical head of each pin, and interactions of each compound with a receptor or antibody could be determined in a direct binding assay. The Geysen technique can be used to synthesize and screen thousands of compounds per week using the multipin method, and the tethered compounds may be reused in many assays. In subsequent work, the level of compound loading on individual pins has been increased to as much as 2 μmol/pin by grafting greater amounts of functionalized acrylate derivatives to detachable pin heads, and the size of the compound library has been increased (Valerio et al. (1993) Int JPept Protein Res 42:1-9). Appropriate linker moieties have also been appended to the pins so that the compounds may be cleaved from the supports after synthesis for assessment of purity and evaluation in competition binding or functional bioassays (Bray et al. (1990) Tetrahedron Lett 31:5811-5814; Valerio et al. (1991) Anal Biochem 197:168-177; Bray et al. (1991) Tetrahedron Lett 32:6163-6166).
More recent applications of the multipin mediod have taken advantage of the cleavable linker strategy to prepare soluble compound libraries (Maeji et al. (1990) J Immunol Methods 134:23-33; Gammon et al. (1991) J Exp Med 173:609-617; Mutch et al. (1991) PeptRes 4:132-137).
Divide-Couple-Recombine
In another embodiment, a variegated library of HDx inhibitor compounds is provided on a set of beads utilizing the strategy of divide-couple-recombine (see, e.g., Ηoughten (1985) PNAS 82:5131-5135; and U.S. Patents 4,631,211; 5,440,016; 5,480,971). Briefly, as the name implies, at each synthesis step where degeneracy (e.g., a plurality of different moieties) is introduced into the library, the beads are divided into as many separate groups to correspond to the number of different residues (e.g., functional groups or other moieties) to be added at that position, the different residues coupled in separate reactions, and the beads recombined into one pool for the next step.
In one embodiment, the divide-couple-recombine strategy can be carried out using the so-called "tea bag" method first developed by Houghten, where synthesis occurs on resin that is sealed inside porous polypropylene bags (Houghten et al. (1986) PNAS 82:5131-5135). Residues are coupled to the resins by placing the bags in solutions of the appropriate individual activated monomers, while all common steps such as resin washing and deprotection (if appropriate) are performed simultaneously in one reaction vessel. At the end of the synthesis, each bag contains a single compound, and the compounds may be liberated from the resins using a multiple cleavage apparatus (Houghten et al. (1986) Int J Pept Protein Res 27:673-678). This technique offers advantages of considerable synthetic flexibility and has been partially automated (Beck- Sickinger et al. (1991) Pept Res 4:88-94). Moreover, compounds can be produced in sufficient quantities (> 500 μmol) for purification and complete characterization if desired.
Synthesis using the tea-bag approach is useful for the production of a library, albeit of limited size, as is illustrated by its use in a range of molecular recognition problems including antibody epitope analysis (Houghten et al. (1986) PNAS 82:5131- 5135), peptide hormone structure-function studies (Beck-Sickinger et al. (1990) Int J Pept Protein Res 36:522-530; Beck-Sickinger et al. (1990) Eur J Biochem 194:449-456), and protein conformational mapping (Zimmerman et al. (1991) Eur J Biochem 200:519- 528).
Combinatorial Synthesis on Nontraditional Solid Supports
The search for innovative methods of solid-phase synthesis has led to the investigation of alternative polymeric supports to the polystyrene-divinylbenzene matrix originally popularized by Merrifield. Cellulose, either in the form of paper disks (Blankemeyer-Menge et al. (1988) Tetrahedron Lett 29-5871-5874; Frank et al. (1988) Tetrahedron 44:6031-6040; Εichler et al. (1989) Collect Czech Chem Commun 54:1746- 1752; Frank, R. (1993) Bioorg Med Chem Lett 3:425-430) or cotton fragments (Εichler et al. (1991) Pept Res 4:296-307; Schmidt et al. (1993) Bioorg Med Chem Lett 3:441- 446) has been successfully functionalized for peptide synthesis. Typical loadings attained with cellulose paper range from 1 to 3 μmol/cm2, and HPLC analysis of material cleaved from these supports indicates a reasonable quality for the synthesized peptides. Alternatively, peptides may be synthesized on cellulose sheets via non-cleavable linkers and then used in ELISA-based binding studies (Frank, R. (1992) Tetrahedron 48:9217- 9232). The porous, polar nature of this support may help suppress unwanted nonspecific protein binding effects. In one convenient configuration synthesis occurs in an 8 x 12 microtiter plate format. Frank has used this technique to map the dominant epitopes of an antiserum raised against a human cytomegalovirus protein, following the overlapping peptide screening (Pepscan) strategy of Geysen (Frank, R. (1992) Tetrahedron 48:9217- 9232). Other membrane-like supports that may be used for solid-phase synthesis include polystyrene-grafted polyethylene films (Berg et al. (1989) J Am Chem Soc 111:8024- 8026).
Combinatorial Libraries by Light-Directed, Spatially Addressable Parallel Chemical Synthesis
A scheme of combinatorial synthesis in which the identity of a compound is given by its locations on a synthesis substrate is termed a spatially-addressable synthesis. In one embodiment, the combinatorial process is carried out by controlling the addition of a chemical reagent to specific locations on a solid support (Dower et al. (1991) Annu Rep Med Chem 26:271-280; Fodor, S.P.A. (1991) Science 251:767; Pirrung et al. (1992) U.S. Patent No. 5,143,854; Jacobs et al. (1994) Trends Biotechnol 12:19-26). The technique combines two well-developed technologies: solid-phase synthesis chemistry and photolithography. The high coupling yields of solid-phase reactions allows efficient compound synthesis, and the spatial resolution of photolithography affords miniaturization. The merging of these two technologies is done through the use of photolabile protecting groups, e.g., amino protecting groups, in die synthetic procedure. The key points of this technology are illustrated in Gallop et al. (1994) J Med
Chem 37:1233-1251. A synthesis substrate is prepared for compound syntiiesis through die covalent attachment of photolabile nitroveratryloxycarbonyl (NVOC) protected amino linkers. Light is used to selectively activate a specified region of the synthesis support for coupling. Removal of the photolabile protecting groups by lights (deprotection) results in activation of selected areas. After activation, the first of a set of residues, each bearing a photolabile protecting group, is exposed to the entire surface. Coupling only occurs in regions that were addressed by light in the preceding step. The reagent solution is removed, and the substrate is again illuminated through a second mask, activating a different region for reaction with a second protected building block. The pattern of masks and the sequence of reactants define the products and their locations. Since this process utilizes photolithography techniques, the number of compounds that can be synthesized is limited only by the number of synthesis sites that can be addressed with appropriate resolution. The position of each compound is precisely known; hence, its interactions with other molecules can be directly assessed. The target can be labeled with a fluorescent reporter group to facilitate the identification of specific interactions with individual members of the matrix.
In a light-directed chemical synthesis, the products depend on the pattern of illumination and on the order of addition of reactants. By varying the lithographic patterns, many different sets of test compounds can be synthesized in the same number of steps; this leads to the generation of many different masking strategies.
Encoded Combinatorial Libraries
In yet another embodiment, the subject mediod provides an HDx inhibitor compound library provided with an encoded tagging system. A recent improvement in the identification of active compounds from combinatorial libraries employs chemical indexing systems using tags that uniquely encode the reaction steps a given bead has undergone and, by inference, the structure it carries. Conceptually, this approach mimics phage display libraries, where activity derives from expressed peptides, but the structures of the active peptides are deduced from the corresponding genomic DNA sequence. The first encoding of synthetic combinatorial libraries employed DNA as die code. Two forms of encoding have been reported: encoding with sequenceable bio-oligomers (e.g., oligonucleotides and peptides), and binary encoding with non-sequenceable tags.
Tagging with sequenceable bio-oligomers
The principle of using oligonucleotides to encode combinatorial synthetic libraries was described in 1992 (Brenner et al. (1992) PNAS 89:5381-5383), and an example of such a library appeared the following year (Needles et al. (1993) PNAS 90: 10700-10704).
A combinatorial library of nominally 77 (= 823,543) peptides composed of all combinations of Arg, Gin, Phe, Lys, Val, D-Val and Thr (three-letter amino acid code), each of which was encoded by a specific dinucleotide (TA, TC, CT, AT, TT, CA and AC, respectively), was prepared by a series of alternating rounds of peptide and oligonucleotide synthesis on solid support. In this work, the amine linking functionality on die bead was specifically differentiated toward peptide or oligonucleotide synthesis by simultaneously preincubating the beads with reagents that generate protected OΗ groups for oligonucleotide synthesis and protected NΗ2 groups for peptide synthesis (here, in a ratio of 1:20). When complete, the tags each consisted of 69-mers, 14 units of which carried the code. The bead-bound library was incubated with a fluorescently labeled antibody, and beads containing bound antibody that fluoresced strongly were harvested by fluorescence-activated cell sorting (FACS). The DNA tags were amplified by PCR and sequenced, and the predicted peptides were synthesized. Following such techniques, HDx inhibitor compound libraries can be derived and screened using HDxs of the subject invention.
It is noted that an alternative approach useful for generating nucleotide-encoded synthetic peptide libraries employs a branched linker containing selectively protected OΗ and NΗ2 groups (Nielsen et al. (1993) J Am Chem Soc 115:9812-9813; and Nielsen et al. (1994) Methods Compan Methods Enzymol 6:361-371). This approach requires that equimolar quantities of test peptide and tag co-exist, though this may be a potential complication in assessing biological activity, especially with nucleic acid based targets.
The use of oligonucleotide tags permits exquisitely sensitive tag analysis. Even so, the method requires careful choice of orthogonal sets of protecting groups required for alternating co-synthesis of the tag and the library member. Furthermore, the chemical lability of the tag, particularly the phosphate and sugar anomeric linkages, may limit the choice of reagents and conditions that can be employed for the synthesis on non- oligomeric libraries. In preferred embodiments, the libraries employ linkers permitting selective detachment of the test HDx inhibitor compound library member for bioassay, in part (as described infra) because assays employing beads limit the choice of targets, and in part because the tags are potentially susceptible to biodegradation.
Peptides themselves have been employed as tagging molecules for combinatorial libraries. Two exemplary approaches are described in the art, both of which employ branched linkers to solid phase upon which coding and ligand strands are alternately elaborated. In the first approach (Kerr JM et al. (1993) J Am Chem Soc 115:2529-2531), orthogonality in synthesis is achieved by employing acid-labile protection for the coding strand and base-labile protection for the ligand strand.
In an alternative approach (Nikolaiev et al. (1993) Pept Res 6:161-170), branched linkers are employed so that the coding unit and the test peptide are both attached to the same functional group on the resin. In one embodiment, a linker can be placed between the branch point and the bead so that cleavage releases a molecule containing both code and ligand (Ptek et al. (1991) Tetrahedron Lett 32:3891-3894). In another embodiment, the linker can be placed so that the test peptide can be selectively separated from the bead, leaving the code behind. This last construct is particularly valuable because it permits screening of the test peptide without potential interference, or biodegradation, of the coding groups. Examples in the art of independent cleavage and sequencing of peptide library members and their corresponding tags has confirmed that the tags can accurately predict the peptide structure.
It is noted that peptide tags are more resistant to decomposition during ligand synthesis than are oligonucleotide tags, but they must be employed in molar ratios nearly equal to those of the ligand on typical 130 μm beads in order to be successfully sequenced. As with oligonucleotide encoding, the use of peptides as tags requires complex protection/deprotection chemistries. Non-sequenceable tagging: binary encoding
An alternative form of encoding the test peptide library employs a set of non- sequenceable tagging molecules (e.g., molecules having electrophoric moieties) that are used as a binary code (Ohlmeyer et al. (1993) PNAS 90:10922-10926). Exemplary tags are haloaromatic alkyl ethers that are detectable as their trimethylsilyl ethers at less than femtomolar levels by electron capture gas chromatography (ECGC). Variations in the length of the alkyl chain, as -well as the nature and position of the aromatic halide substituents, permit the synthesis of at least 40 such tags, which in principle can encode 240 (e.g., upwards of IO12) different molecules. In the original report (Ohlmeyer et al., supra) the tags were bound to about 1% of the available amine groups of a peptide library via a photocleavable O-nitrobenzyl linker. This approach is convenient when preparing combinatorial libraries of peptides or other amme-containing molecules. A more versatile system has, however, been developed that permits encoding of essentially any combinatorial library. Here, the ligand is attached to the solid support via the photocleavable linker and d e tag is attached through a catechol ether linker via carbene insertion into the bead matrix (Nestler et al. (1994) J Org Chem 59:4723-4724). This orthogonal attachment strategy permits the selective detachment of library members for bioassay in solution and subsequent decoding by ECGC after oxidative detachment of the tag sets.
Binary encoding with tags, e.g., electrophoric tags, has been particularly useful in defining selective interactions of substrates with synthetic receptors (Borchardt et al. (1994) J Am Chem Soc 116:373-374), and model systems for understanding the binding and catalysis of biomolecules. Even using detailed molecular modeling, the identification of the selectivity preferences for synthetic receptors has required the manual synthesis of dozens of potential substrates. The use of encoded libraries makes it possible to rapidly examine all the members of a potential binding set. The use of binary-encoded libraries has made the determination of binding selectivities so facile that structural selectivity has been reported for four novel syntiietic macrobicyclic and tricyclic receptors in a single communication (Wennemers et al. (1995) J Org Chem 60: 1108-1109; and Yoon et al.
(1994) Tetrahedron Lett 35:8557-8560) using the encoded library mentioned above. Similar facility in defining specificity of interaction would be expected for many other biomolecules.
Although the several amide-linked libraries in the art employ binary encoding with the electrophoric tags attached to amine groups, attaching these tags directly to die bead matrix provides far greater versatility in the structures that can be prepared in encoded combinatorial libraries. Attached in this way, the tags and their linker are nearly as unreactive as the bead matrix itself. Two binary-encoded combinatorial libraries have been reported where the tags are attached directly to the solid phase (Ohlmeyer et al.
(1995) PNAS 92:6027-6031) and provide guidance for generating the subject HDx inhibitor compound library. Both libraries were constructed using an orthogonal attachment strategy in which the library member was linked to the solid support by a photolabile linker and d e tags were attached through a linker cleavable only by vigorous oxidation. Because the library members can be repetitively partially photoeluted from the solid support, library members can be utilized in multiple assays. Successive photoelution also permits a very high throughput iterative screening strategy: first, multiple beads are placed in 96-well microtiter plates; second, ligands are partially detached and transferred to assay plates; third, a bioassay identifies the active wells; fourth, the corresponding beads are rearrayed singly into new microtiter plates; fifth, single active compounds are identified; and sixth, the structures are decoded.
The above approach was employed in screening for carbonic anhydrase (CA) binding and identified compounds which exhibited nanomolar affinities for CA. Unlike sequenceable tagging, a large number of structures can be rapidly decoded from binary- encoded libraries (a single ECGC apparatus can decode 50 structures per day). Thus, binary-encoded libraries can be used for the rapid analysis of structure-activity relationships and optimization of both potency and selectivity of an active series. The synthesis and screening of large unbiased binary encoded HDx inhibitor compound libraries for lead identification, followed by preparation and analysis of smaller focused libraries for lead optimization, offers a particularly powerful approach to discovery of HDx inhibitor compounds.
HDx inhibitor compounds can be synthesized on solid support by appropriate functionalization for attachment to a solid matrix, or altematively, by solution-phase synthesis followed by immobilization through an appropriate functional group. Thus, in an illustrative embodiment, an HDx inhibitor compound, which is analogous to trichostatin, can be synthesized on a solid support by attachment through an amino group of the specificity element A, as shown in Figure 7. The solid support is preferably capable of withstanding synthetic conditions required to synthesize the requisite compounds. The compound can preferably be released from the solid support, e.g., by selective cleavage of an amide bond.
The synthetic steps employed to synthesize compounds on solid support are preferably selected to allow a wide variety of residues (e.g., building blocks) to be coupled to the immobilized moieties, preferably under mild conditions. Suitable reaction chemistries include well-known carbon-carbon bond forming reactions such as the Stille and Suzuki couplings, as well as Ηorner-Emmons reactions, Ni/Cr mediated couplings, and the like. Particularly preferred coupling reactions can be performed in the presence of water and do not require harsh conditions or expensive reagents. Thus, in an exemplary synthesis shown in Figure 7, substituted N-methyl-4-
(tributyltin)anilines (in which Rj represents one or more substitutions, e.g., hydrogen, halogen, alkyl, alkoxy, and the like) are coupled in a plurality of reaction vessels to beads of a solid support (e.g., Affigel). The beads are further divided into a plurality of reaction vessels, and suspended in a solvent such as DMF, and one acid chloride building block (corresponding to linking element B) is introduced into each vessel (R2 and R3 represent, e.g., hydrogen, halogen, alkyl, and the like; and the broken line represents an optional double bond). The reactions are stirred under an inert gas (e.g. nitrogen) and a palladium catalyst (e.g., Pd(PPh3)4) is added (0.1-1.0 mol%). The reaction is stirred for 1-24 hours. Upon completion of the reaction, the beads are washed, and placed in a plurality of vessels. The aldehyde moiety is deprotected by mild acid treatment (e.g., PPTS in MeOΗ), and the beads are again washed and placed in a plurality of reaction vessels, and the beads are suspended in dry acetonitrile. One building block (corresponding to the reactive element C) is then added to each reaction vessel. As illustratively shown in Figure 7, a plurality of phosphonates can be employed (R4 represents, e.g., alkyl, alkenyl, alkynyl, alkoxy, and the like). A Ηorner-Emmons reaction is performed by addition of LiCl (1.1 equiv.) and diisopropylethylamine (DD?EA) or DBU (1.2 equiv). Upon completion of the reaction, the beads are washed with water and acetonitrile, and then dried to yield a library of candidate HDx inhibitor compounds on solid support. The compounds can then be released from the solid support into solution; or the compounds can be screened while attached to the solid support. The above combinatorial synthesis can be performed in an encoded mode, e.g., the binary tagging method described supra, by addition of the appropriate tag for each monomer. In this mode, after each reaction has been performed and the corresponding tag attached, the beads from all reactions can be recombined and then divided into aliquots for further derivatization. This method provides the advantage of ease of handling when large libraries are to be synthesized. Regardless of the method of synthesis, the combinatorial library can be screened for activity according to known methods (see, e.g., Gordon et al., supra).
In another aspect, the present invention provides pharmaceutically acceptable compositions which comprise a therapeutically-effective amount of one or more of the compounds described above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. As described in detail below, the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example; drenches (aqueous or non-aqueous solutions or suspensions), tablets, boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (3) topical application, for example, as a cream, ointment or spray applied to the skin; or (4) intravaginally or intrarectally, for example, as a pessary, cream or foam.
The phrase "therapeutically-effective amount" as used herein means that amount of a compound, material, or composition comprising a deacetylase inhibitor of the present invention which is effective for producing some desired therapeutic effect by inhibiting histone deacetylation in at least a sub-population of cells in an animal and thereby blocking the biological consequences of that event in the treated cells, at a reasonable benefit/risk ratio applicable to any medical treatment.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact wid the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The phrase "pharmaceutically-acceptable carrier" as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject deacetylase inhibitor agent from one organ, or portion of d e body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically- acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as co starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil and soybean oil; (10) glycois, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl iaurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations.
As set out above, certain embodiments of the present deacetylase inhibitors may contain a basic functional group, such as amino or alkylamino, and are, thus, capable of forming pharmaceutically-acceptable salts with pharmaceutically-acceptable acids. The term "pharmaceutically-acceptable salts" in this respect, refers to the relatively non-toxic, inorganic and organic acid addition salts of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by separately reacting a purified compound of the invention in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, Iaurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like. (See, for example, Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci. 66: 1-19) In other cases, the deacetylase inhibitory compounds of die present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically-acceptable salts with pharmaceutically-acceptable bases. The term "pharmaceutically-acceptable salts" in these instances refers to the relatively non-toxic, inorganic and organic base addition salts of compounds of the present invention. These salts can likewise be prepared in situ during the final isolation and purification of the compounds, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically- acceptable metal cation, with ammonia, or with a pharmaceutically-acceptable organic primary, secondary or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like. (See, for example, Berge et al., supra)
Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
Examples of pharmaceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. Formulations of the present invention include those suitable for oral, nasal, topical
(including buccal and sublingual), rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the deacetylase inhibitor which produces a tiierapeutic effect. Generally, out of one hundred per cent, tiiis amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention widi the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a deacetylase inhibitor of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. A deacetylase inhibitor of the present invention may also be administered as a bolus, electuary or paste.
In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absoφtion accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycois, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycois and the like.
A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered deacetylase inhibitor moistened with an inert liquid diluent. The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and odier coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incoφorating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
Liquid dosage forms for oral administration of the deacetylase inhibitors of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycois and fatty acid esters of sorbitan, and mixtures thereof.
Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
Suspensions, in addition to the active deacetylase inhibitor, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active deacetylase inhibitor.
Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
Dosage forms for the topical or transdermal administration of a deacetylase inhibitor of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
The ointments, pastes, creams and gels may contain, in addition to an active deacetylase inhibitor of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycois, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing the deacetylase inhibitor in the proper medium. Absoφtion enhancers can also be used to increase the flux of the deacetylase inhibitor across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the deacetylase inhibitor in a polymer matrix or gel.
Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention.
Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more deacetylase inhibitors of the invention in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents. Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absoφtion of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absoφtion such as aluminum monostearate and gelatin. In some cases, in order to prolong the effect of a dmg, it is desirable to slow the absoφtion of the dmg from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amoφhous material having poor water solubility. The rate of absoφtion of die dmg then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Altematively, delayed absoφtion of a parenterally-administered dmg form is accomplished by dissolving or suspending the dmg in an oil vehicle.
Injectable depot forms are made by forming microencapsule matrices of the subject deacetylase inhibitors in biodegradable polymers such as polylactide- polyglycolide. Depending on the ratio of dmg to polymer, and the nature of the particular polymer employed, the rate of dmg release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the dmg in liposomes or microemulsions which are compatible with body tissue. When the compounds of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier. The preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given by forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infiision or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administration is preferred.
These deacetylase inhibitor may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally and topically, as by powders, ointments or drops, including buccally and sublingually. Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.
Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
The selected dosage level will depend upon a variety of factors including the activity of the particular deacetylase inhibitor employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular deacetylase inhibitor employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve d e desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
Another aspect of the present invention relates to a method of inducing and/or maintaining a differentiated state, enhancing survival, and/or inhibiting (or altematively potentiating) proliferation of a cell, by contacting the cells with an agent which modulates HDx-dependent transcription. For instance, it is contemplated by the invention that, in light of the present finding of an apparently broad involvement of HDx proteins in the control of chromatin stmcture and, thus, transcription and replication, the subject method could be used to generate and/or maintain an array of different tissue both in vitro and in vivo. An "HDx therapeutic," whether inhibitory or potentiating with respect to modulating histone deacetylation, can be, as appropriate, any of the preparations described above, including isolated polypeptides, gene therapy constmcts, antisense molecules, peptidomimetics or agents identified in the dmg assays provided herein.
The HDx compounds of the present invention are likely to play an important role in the modulation of cellular proliferation. There are a wide variety of pathological cell proliferative conditions for which HDx therapeutics of the present invention may be used in treatment. For instance, such agents can provide therapeutic benefits where the general strategy being the inhibition of an anomalous cell proliferation. Diseases that might benefit from this methodology include, but are not limited to various cancers and leukemias, psoriasis, bone diseases, fibroproliferative disorders such as involving connective tissues, atherosclerosis and other smooth muscle proliferative disorders, as well as chronic inflammation.
In addition to proliferative disorders, the present invention contemplates the use of HDx therapeutics for the treatment of differentiative disorders which result from, for example, de-differentiation of tissue which may (optionally) be accompanied by abortive reentry into mitosis, e.g. apoptosis. Such degenerative disorders include chronic neurodegenerative diseases of the nervous system, including Alzheimer's disease, Parkinson's disease, Huntingdon's chorea, amylotrophic lateral sclerosis and the like, as well as spinocerebellar degenerations. Other differentiative disorders include, for example, disorders associated with connective tissue, such as may occur due to de- differentiation of chondrocytes or osteocytes, as well as vascular disorders which involve de-differentiation of endothelial tissue and smooth muscle cells, gastric ulcers characterized by degenerative changes in glandular cells, and renal conditions marked by failure to differentiate, e.g. Wilm's tumors. It will also be apparent that, by transient use of modulators of HDx activities, in vivo reformation of tissue can be accomplished, e.g. in the development and maintenance of organs. By controlling the proliferative and differentiative potential for different cells, the subject HDx therapeutics can be used to reform injured tissue, or to improve grafting and moφhology of transplanted tissue. For instance, HDx antagonists and agonists can be employed in a differential manner to regulate different stages of organ repair after physical, chemical or pathological insult. For example, such regimens can be utilized in repair of cartilage, increasing bone density, liver repair subsequent to a partial hepatectomy, or to promote regeneration of lung tissue in the treatment of emphysema. The present method is also applicable to cell culture techniques.
In one embodiment, the HDx therapeutic of the present invention can be used to induce differentiation of uncommitted progenitor cells and thereby give rise to a committed progenitor cell, or to cause further restriction of the developmental fate of a committed progenitor cell towards becoming a terminally-differentiated cell. For example, the present method can be used in vitro or in vivo to induce and/or maintain the differentiation of hematopoietic cells into erythrocytes and other cells of the hematopoietic system. In an illustrative embodiment, the effect of erythropioetin (EPO) on the growth of EPO-responsive erythroid precursor cells is increased to influence their differentiation into red blood cells. For example, as a result of administering an inhibitor of histone deacetylation, the amount of EPO, or other diferentiating agent, required for growth and/or differentiation is reduced (PCT US92/07737). Accordingly, the HDx therapeutics of the present invention, particularly those which antagonize HDx deacetylase activity, can be administered alone or in conjunction with EPO and in a suitable carrier to vertebrates to promote erythropoiesis. Altematively, cells could be treated ex vivo. Such treatment is contemplated in die treatment of a variety of disease states, including in individuals who require bone marrow transplants (e.g. patients with aplastic anemia, acute leukemias, recurrent lymphomas, or solid tumors).
To illustrate, prior to receiving a bone marrow transplant, a recipient is prepared by ablating or removing endogenous hematopoietic stem cells. Such treatment is usually carried out by total body irradiation or delivery of a high dose of an alkylating agent or other chemotherapeutic, cytotoxic agent, Anklesaria, et al. (1987) PNAS 84:7681-7685). Following preparation of the recipient, donor bone marrow cells are injected intravenously. Optionally, the HDx therapeutics of the present invention could be contacted with the cells ex vivo or administered to the subject with the reimplanted cells. It is also contemplated that there may be cell-type specific HDx proteins, and/or that some cell types may be more sensitive to modulation of HDx deacetylase activities. Even within a cell type, the stage of differentiation or position in the cell cycle could influence their response to an HDx therapeutic. Accordingly, the present invention contemplates the use of agents which modulate histone deacetylase activity to specifically inhibit or activate certain cell types. In an illustrative example, T cell proliferation could be preferentially inhibited in order to induce tolerance by using a procedure similar to that for inducing tolerance using sodium butyrate (see, for example, PCT7US93/03045). To illustrate, the HDx therapeutics of the present invention may be used to induce antigen- specific tolerance in any situation in which it is desirable to induce tolerance, such as autoimmune diseases, in allogeneic or xenogeneic transplant recipients, or in graft versus host (GVΗ) reactions. According to the invention, tolerance will typically be induced by presenting the tolerizing compound (e.g., an HDx inhibitor) substantially contemporaneously with the antigen, i.e. reasonably close together in time with the antigen. In preferred embodiments the HDx therapeutic will be administered after presentation of the antigen, so that they will have their effect after the particular repertoire of Th cells begins to undergo clonal expansion.
Yet another aspect of the present invention concerns the application of HDx therapeutics to modulating moφhogenic signals involved in organogenic pathways. Thus, it is contemplated by the invention that compositions comprising HDx therapeutics can also be utilized for both cell culture and therapeutic methods involving generation and maintenance of tissue.
In a further embodiment of the invention, the subject HDx therapeutics will be useful in increasing the amount of protein produced by a cell or recombinant cell. The cell may include any primary cell isolated from any animal, cultured cells, immortalized cells, and established cell lines. The animal cells used in the present invention include cells which intrinsically have an ability to produce a desired protein; cells which are induced to have an ability to produce a desired protein, for example, by stimulation with a cytokine such as an interferon, an interleukin; genetically engineered cells into which a gene for a desired protein is introduced. The protein produced by the process could include any peptides or proteins, including peptide hormone or proteinaceous hormones such as any useful hormone, cytokine, interleukin, or protein which it may be desirable to have in purified form and/or in large quantity.
Another aspect of the invention features transgenic non-human animals which express a heterologous HDx gene of the present invention, or which have had one or more genomic HDx genes dismpted in at least one of the tissue or cell-types of the animal. Accordingly, the invention features an animal model for developmental diseases, which animal has one or more HDx allele which is mis-expressed. For example, a mouse can be bred which has one or more HDx alleles deleted or otherwise rendered inactive. Such a mouse model can then be used to study disorders arising from mis-expressed HDx genes, as well as for evaluating potential therapies for similar disorders.
Another aspect of the present invention concerns transgenic animals which are comprised of cells (of that animal) which contain a transgene of the present invention and which preferably (though optionally) express an exogenous HDx protein in one or more cells in the animal. An HDx transgene can encode the wild-type form of the protein, or can encode homologs thereof, including both agonists and antagonists, as well as antisense constructs. In preferred embodiments, the expression of the transgene is restricted to specific subsets of cells, tissues or developmental stages utilizing, for example, cis-acting sequences that control expression in the desired pattern. In the present invention, such mosaic expression of an HDx protein can be essential for many forms of lineage analysis and can additionally provide a means to assess the effects of, for example, lack of HDx expression which might grossly alter development in small patches of tissue within an otherwise normal embryo. Toward this and, tissue-specific regulatory sequences and conditional regulatory sequences can be used to control expression of the transgene in certain spatial pattems. Moreover, temporal pattems of expression can be provided by, for example, conditional recombination systems or prokaryotic transcriptional regulatory sequences.
Genetic techniques which allow for the expression of transgenes can be regulated via site-specific genetic manipulation in vivo are known to those skilled in the art. For instance, genetic systems are available which allow for the regulated expression of a recombinase that catalyzes the genetic recombination a target sequence. As used herein, the phrase "target sequence" refers to a nucleotide sequence that is genetically recombined by a recombinase. The target sequence is flanked by recombinase recognition sequences and is generally either excised or inverted in cells expressing recombinase activity. Recombinase catalyzed recombination events can be designed such diat recombination of the target sequence results in either the activation or repression of expression of one of the subject HDx proteins. For example, excision of a target sequence which interferes with the expression of a recombinant HDx gene, such as one which encodes an antagonistic homolog or an antisense transcript, can be designed to activate expression of that gene. This interference with expression of the protein can result from a variety of mechanisms, such as spatial separation of die HDx gene from the promoter element or an internal stop codon. Moreover, the transgene can be made wherein d e coding sequence of the gene is flanked by recombinase recognition sequences and is initially transfected into cells in a 3' to 5' orientation with respect to the promoter element. In such an instance, inversion of the target sequence will reorient the subject gene by placing the 5' end of the coding sequence in an orientation with respect to the promoter element which allow for promoter driven transcriptional activation.
In an illustrative embodiment, either the cre/loxP recombinase system of bacteriophage PI (Lakso et al. (1992) PNAS 89:6232-6236; Orban et al. (1992) PNAS 89:6861-6865) or the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al. (1991) Science 251 :1351-1355; PCT publication WO 92/15694) can be used to generate in vivo site-specific genetic recombination systems. Cre recombinase catalyzes the site-specific recombination of an intervening target sequence located between loxP sequences. loxP sequences are 34 base pair nucleotide repeat sequences to which the Cre recombinase binds and are required for Cre recombinase mediated genetic recombination. The orientation of loxP sequences determines whether the intervening target sequence is excised or inverted when Cre recombinase is present (Abremski et al. (1984) J. Biol. Chem. 259:1509-1514); catalyzing the excision of the target sequence when the loxP sequences are oriented as direct repeats and catalyzes inversion of the target sequence when loxP sequences are oriented as inverted repeats.
Accordingly, genetic recombination of the target sequence is dependent on expression of the Cre recombinase. Expression of the recombinase can be regulated by promoter elements which are subject to regulatory control, e.g., tissue-specific, developmental stage-specific, inducible or repressible by externally added agents. This regulated control will result in genetic recombination of the target sequence only in cells where recombinase expression is mediated by the promoter element. Thus, the activation expression of a recombinant HDx protein can be regulated via control of recombinase expression.
Use of the cre loxP recombinase system to regulate expression of a recombinant HDx protein requires the constmction of a transgenic animal containing transgenes encoding both d e Cre recombinase and the subject protein. Animals containing both the Cre recombinase and a recombinant HDx gene can be provided through the constmction of "double" transgenic animals. A convenient method for providing such animals is to mate two transgenic animals each containing a transgene, e.g., an HDx gene and recombinase gene. One advantage derived from initially constmcting transgenic animals containing an HDx transgene in a recombinase-mediated expressible format derives from the likelihood that the subject protein, whether agonistic or antagonistic, can be deleterious upon expression in the transgenic animal. In such an instance, a founder population, in which the subject transgene is silent in all tissues, can be propagated and maintained. Individuals of this founder population can be crossed with animals expressing the recombinase in, for example, one or more tissues and/or a desired temporal pattern. Thus, the creation of a founder population in which, for example, an antagonistic HDx transgene is silent will allow the study of progeny from that founder in which dismption of HDx mediated induction in a particular tissue or at certain developmental stages would result in, for example, a lethal phenotype.
Similar conditional transgenes can be provided using prokaryotic promoter sequences which require prokaryotic proteins to be simultaneous expressed in order to facilitate expression of the HDx transgene. Exemplary promoters and the corresponding trans-activating prokaryotic proteins are given in U.S. Patent No. 4,833,080.
Moreover, expression of the conditional transgenes can be induced by gene therapy-like methods wherein a gene encoding the trans-activating protein, e.g. a recombinase or a prokaryotic protein, is delivered to the tissue and caused to be expressed, such as in a cell-type specific manner. By this method, an HDx transgene could remain silent into adulthood until "turned on" by the introduction of the trans- activator.
In an exemplary embodiment, the "transgenic non-human animals" of the invention are produced by introducing transgenes into the germline of the non-human animal. Embryonic target cells at various developmental stages can be used to introduce transgenes. Different methods are used depending on the stage of development of the embryonic target cell. The zygote is the best target for micro-injection. In the mouse, the male pronucleus reaches the size of approximately 20 micrometers in diameter which allows reproducible injection of l-2pl of DNA solution. The use of zygotes as a target for gene transfer has a major advantage in that in most cases the injected DNA will be incoφorated into the host gene before the first cleavage (Brinster et al. (1985) PNAS 82:4438-4442). As a consequence, all cells of the transgenic non-human animal will carry the incoφorated transgene. This will in general also be reflected in the efficient transmission of the transgene to offspring of the founder since 50% of the germ cells will harbor the transgene. Microinjection of zygotes is the preferred method for incoφorating transgenes in practicing the invention. Retroviral infection can also be used to introduce HDx transgenes into a non- human animal. The developing non-human embryo can be cultured in vitro to the blastocyst stage. During this time, the blastomeres can be targets for retroviral infection (Jaenich, R. (1976) PNAS 73 : 1260-1264). Efficient infection of the blastomeres is obtained by enzymatic treatment to remove the zona pellucida (Manipulating the Mouse Embryo, Ηogan eds. (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1986). The viral vector system used to introduce the transgene is typically a replication-defective retrovirus carrying the transgene (Jahner et al. (1985) PNAS 82:6927-6931; Van der Putten et al. (1985) PNAS 82:6148-6152). Transfection is easily and efficiently obtained by culturing the blastomeres on a monolayer of vims-producing cells (Van der Putten, supra; Stewart et al. (1987) EMBO J. 6:383-388). Altematively, infection can be performed at a later stage. Vims or vims-producing cells can be injected into the blastocoele (Jahner et al. (1982) Nature 298:623-628). Most of the founders will be mosaic for the transgene since incoφoration occurs only in a subset of the cells which formed the transgenic non-human animal. Further, the founder may contain various retroviral insertions of the transgene at different positions in the genome which generally will segregate in the offspring. In addition, it is also possible to introduce transgenes into the germ line by intrauterine retroviral infection of the midgestation embryo (Jahner et al. (1982) supra). A third type of target cell for transgene introduction is die embryonic stem cell
(ES). ES cells are obtained from pre-implantation embryos cultured in vitro and fused with embryos (Evans et al. (1981) Nature 292:154-156; Bradley et al. (1984) Nature 309:255-258; Gossler et al. (1986) PNAS 83: 9065-9069; and Robertson et al. (1986) Nature 322:445-448). Transgenes can be efficiently introduced into the ES cells by DNA transfection or by retrovims-mediated transduction. Such transformed ES cells can thereafter be combined with blastocysts from a non-human animal. The ES cells thereafter colonize the embryo and contribute to the germ line of the resulting chimeric animal. For review see Jaenisch, R. (1988) Science 240:1468-1474.
Methods of making HDx knock-out or dismption transgenic animals are also generally known. See, for example, Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986). Recombinase dependent knockouts can also be generated, e.g. by homologous recombination to insert recombinase target sequences flanking portions of an endogenous HDx gene, such that tissue specific and/or temporal control of inactivation of an HDx allele can be controlled as above. Exemplification
The invention, now being generally described, will be more readily understood by reference to the following examples, which are included merely for puφoses of illustration of certain aspects and embodiments of the present invention and are not intended to limit the invention.
Example 1
Trapoxin is a microbially derived cyclotetrapeptide that inhibits histone deacetylation in vivo and causes mammalian cells to arrest in the cell cycle. A trapoxin affinity matrix was used to isolate two nuclear proteins that copurified with histone deacetylase activity. Both proteins were identified by peptide microsequencing, and a cDNA encoding die histone deacetylase catalytic subunit {HDl) was cloned from a Jurkat T cell library. As the predicted protein is highly similar to the yeast transcriptional regulator RPD3, this study supports a role for histone deacetylase as a key regulator of eukaryotic transcription.
A requirement for a functional histone deacetylase in cell cycle progression has been implicated by the discovery diat two cytostatic agents, trapoxin and trichostatin (Figure IA), inhibit histone deacetylation in cultured mammalian cells and in fractioned cell extracts (4). In addition to causing G\ and G2 phase cell cycle arrest, these natural products alter gene expression and induce certain mammalian cell lines to differentiate. Whereas sodium butyrate also has these properties, both trapoxin and trichostatin are five orders of magnitude more potent.
Trapoxin is an "irreversible" inhibitor of histone deacetylase activity and its molecular stmcture offers clues as to how it could form a covalent bond with a nucleophilic active site residue. First, trapoxin contains an electrophilic epoxyketone that is essential for biological activity (5). Second, the aliphatic epoxyketone side chain is approximately isosteric with N-acetyl lysine (Figure IA). Trapoxin likely acts as a substrate mimic, with epoxyketone poised to alkylate an active site nucleophile. We therefore regarded trapoxin as a tool that could reveal the molecular identity of histone deacetylase, so that its role in transcriptional regulation and cell cycle progression could be elucidated. Tritium-Iabeled trapoxin was prepared by total synthesis and used to identify trapoxin binding protein in crude extracts from bovine thymus. We used a charcoal precipitation assay to detect a specific trapoxin binding activity primarily in the nuclear fraction of the extracts (6). The binding activity was saturable with nanomolar concentrations of [3H]trapoxin and was completed by the simultaneous addition of unlabled trapoxin. Trichostatin also competed with [3H]trapoxin (f°r synthesis, see Example 2), suggesting that both of these compounds exert their cellular effects by targeting the same molecule.
If trapoxin and trichostatin induce cell cycle arrest by directly inhibiting histone deacetylase, then the binding and enzymatic activities should copurify. To investigate this possibility, we fractioned nuclear thymus proteins by ammonium sulfate precipitation and Mono Q anion exchange chromatography.
Briefly, thymocytes (-12 g) prepared from fresh bovine thymus were homogenized in hypotonic lysis buffer [20 mM tris (pH 7.8), 20 mM NaCl, 1 mM EDTA, 10% glycerol, ImM PMSF, ImM benzamidine, 10 μg/ml each of pepstatin, aprotinin, and leupeptin] by mechanical dismption and the nuclei were isolated by centrifugation at 3000g. Nuclei were resuspended in lysis buffer and the proteins were extracted with 0.4 M ammonium sulfate. The viscous lysate was sonicated and clarified by centrifugation at 100,000g for one hour. Proteins were then precipitated with 90% saturated ammonium sulfate and recovered by centrifugation (100,000g, one hour). After through dialysis against Q buffer (25 mM tris pH 8, 10 mM NH Cl, 0.25 mM EDTA, 10% glycerol), a portion of the nuclear proteins (-12 mg total protein) was loaded onto a HR 10/10 Mono Q column (Pharmacia). The column was washed with 25ml Q buffer and eluted with a 50 ml linear gradient of 10 to 500 mM NH4 Cl. The column was further washed with 25 ml 500 mM Nl and 25 ml 1 M histone deacetylase activities or further purified with the K-trap affinity matrix. All procedures were done at 4°C.
Two peaks of histone deacetylase activity eluted from the Mono Q column between 250 and 350 mM NH4C1 (Figure IB). Trapoxin binding activity, as revealed by the charcoal precipitation assay (40 nM [3H]trapoxin), precisely coeluted with the histone deacetylase peaks. Furthermore, all detectable histone deacetylase activity was abolished by treatment with either trapoxin or trichostatin (20 nM). Similar results were obtained with Mono Q fractioned nuclear extracts prepared form human Jurkat T cells.
To purify the histone deacetylase further, we synthesized an affinity matrix based on the trapoxin stmcture. Because trapoxin itself is not amenable to derivatization and the epoxyketone side chain is indispensable for activity, we chose to replace one of the phenylalanine residues of trapoxin's cyclic core with a lysine that could then be covalently linked to a solid support. This molecule, which we call K-trap, was prepared by a twenty step synthesis starting with commercially available (R)-proline and (S,S)- threiitol acetonide (Figure 2A) (see Example 3). Synthetic K-trap inhibited [3H]thymidine incoφoration in MG-63 human osteosarcoma cells with a potency approximately one tenth that of trapoxin. In vitro histone deacetylase activity was also inhibited potently by this compound (complete inactivation at 20 nM) (8).
K-trap was deprotected with Pd(Ph3P)4 and coupled to an activated agarose matrix (Figure 2 A). Mono Q fractions containing nuclear proteins from bovine thymus were incubated with the K-trap affinity matrix and then tested for both trapoxin binding and histone deacetylase activity. Both activities were depleted (90%) by treatment with the K-trap matrix, yet a control matrix capped with ethanolamine had no effect on either activity (8). Bound polypeptides were eluted by boiling the matrix in 1% SDS buffer and separated b polyacrylamide gel electrophoresis. In vitro binding experiments with soluble [3H]trapoxin indicated that the radiolabel is released into solution following protein denaturation with SDS or gunaidinium hydrochloride. Thus, trapoxin binding proteins were expected to elute from the affinity matrix with SDS.
The silver stained gel of the affinity matrix eluates revealed six major polypeptides with apparent molecular sizes between 45 and 50kD (Figure 2B). The interaction between bovine p46-p50 and the K-trap matrix appeared to be specific, because these proteins were not retained when the incubation was done in the presence of either trapoxin or trichostatin (Figure 2B), nor were they structurally unrelated histone deacetylase inhibitor, trichostatin, to prevent p46-p50 from binding to the K-trap matrix implies that one or more of these polypeptides constitute the biologically relevant protein target of both trapoxin and trichostatin. When the affinity purification was repeated with
Jurkat nuclear extracts, only two major bands, p50 and p55, were observed by silver staining (Figure 2B). Recovery of human p50 and p55 was similarly abolished by trapoxin (Figure 2B) and trichostatin (8). Because the relative intensities of bovine p46- p49 vary with each protein preparation, we suspect that they are proteolytic fragments derived from the bovine equivalent of human p55. One of the bands (p50) is common to both human and bovine sources.
Large scale purification of the bovine proteins led to the resolution of two major bands of -46 and -50 kD in the final preparative electrophoresis step, both of which were submitted for microsequencing. To obtain enough trapoxin binding protein for microsequencing, nuclear ammonium sulfate pellets from 15 bovine thymuses were prepared as described above. Sedimented proteins were resuspended in and dialyzed against buffer A [20 mM bistris (pH 7.2), 20 mM NaCl, 10% glycerol] for 12 hours, and brought to pH 5.8 by dialyzing against bugger A (pH 5.8) for 30 minutes. After centrifugation, the dialysate (-650 mg protein) was loaded onto a Q Sepharose FF column (2.6 x 10 cm; Pharmacia) and the column was washed with 120 ml buffer A (pH 5.8). Proteins wee eluted with a 400 ml linear gradient of 20 to 600 mM NaCl in buffer A. Fractions (10 ml; each fraction contained 1 ml of 1 M tris pH 8 to neutralize the acidic buffer A) were assayed for trapoxin binding activity. Tween-20 was added to active fractions at a final concentration of 0.05%, and these fractions were incubated with K-trap affinity matrix for 16 hours (25 μl per ml Q fraction). After washing the matrix three times with phosphate buffered saline, bound proteins were eluted by boiling in 40 μl of SDS sample buffer per 25 μl of matrix. SDS eluates were combined and the proteins resolved by SDS-PVDF membrane (Biorad). Staining with Ponceau S revealed two major bands (46 and 50 kD). The excised bands were proteolytically digested and the HPLC purified peptide fragments were sequenced at the Harvard Microchemistry Facility.
The bovine protein of larger molecular size (-50 kD) corresponds to a known protein, RbAp48 (11), that consists of seven WD repeat domains (12). Originally identified as a protein that binds to the retinoblastoma gene product (pRb), RbAp48 may constitute an adaptor subunit that targets the histone deacetylase to specific chromatin domains.
The -46 kD bovine protein is highly related to the protein encoded by the yeast RPD3 gene, which has been implicated by several genetic screens as a transcriptional regulator, but whose biochemical function is unknown (13). Partial cDNA sequences for the human gene were identified in the expressed sequence tag database (dbEST) and were used to design polymerase chain reaction (PCR) primers. Briefly, after noting sequence similarity between peptides derived from the purified bovine trapoxin binding protein and yeast RPD3, we checked dbEST to see whether any partial sequences for the human homologue had been reported. Two ESTs (Genbank accession numbers: D31480 and F07807) were identified whose predicted translation products aligned widi high sequence similarity to NH2- and COOH-terminal regions of HDl, respectively, PCR primers were designed based on these tags and a one kilobase PCR product was obtained from a Jurkat cDNA library (Stratagene). A 32P labeled probe prepared by random priming was used to screen the Jurkat library, and ten positive clones were isolated. One of the clones was fully sequenced and found to contain a putative full-length open reading frame (Figure 3A). The peptide sequences obtained from the purified bovine protein align with 100% identity to sequences deduced from this coding region (Figure 3A, boxed residues). We call this human protein HDl (for histone deacetylase), and its predicted size of 55 kD agrees well with the estimated size of p55 isolated from Jurkat nuclear extracts using the K-trap affinity matrix (Figure 2B). A dbEST search indicated the existence of at least two other related human genes.
To determine the relationship between the proteins from bovine thymus (p46- p50) and the proteins isolated from human Jurkat T cells (p50 and p55), an antiserum was generated against a peptide specified by the HDl open reading frame (Figure 3 A, amino acids 319 to 334). Immunoblot analysis of the bovine proteins p46-p49 and the human protein p55 showed that they all react with the antiserum and provides additional evidence that these bands correspond to bovine and human HDl (Figure 3B). A monoclonal antibody that specifically recognizes RbAp48 was used to confirm die identity of bovine and hum p50. Importantly, neither HDl nor RbAp48 was detected when the affinity purification was done in the presence of trapoxin or trichostatin (Figure 3B).
We used affinity purified antibodies directed against a COOH-terminal peptide (amino acids 467 to 482) to immunoprecipitate HDl from cmde nuclear extracts. The immunoprecipitates contained histone deacetylase activity that was inhibited by both trapoxin and trichostatin (Figure 4A). Consistent with the idea that HDl and RbAp48 form a complex in vivo, the two proteins coprecipitated with the anti-DHl antibodies (Figure 4B). Neither HDl, RbAp48, nor the associated histone deacetylase activity were immunoprecipitated in the presence of the HDl COOH-terminal peptide (Figure 4A and 4B) (15). HDl, like RbAp48 (11), is detected predominantly in the nucleus by immunostaining with the aforementioned antibodies (8). Given that HDl and RbAp48 are the major proteins eluted from the K-trap matrix (Figure 2B), it is likely that they interact directly with one another.
We extended the results obtained with the endogenous protein by expressing recombinant FLAG epitope tagged HDl (HDI-F) in Jurkat T cells. Anti-FLAG immunoprecipitates from cells transfected with pBJ5/HD/-F contained histone deacetylase activity that was sensitive to both trapoxin and trichostatin (Figure 4C). Histone deacetylase activity was not precipitated when the antibody was blocked with excess FLAG peptide (15). Interestingly, endogenous RbAp48 did not coprecipitate with overexpressed HDl-F (8), demonstrating that RbAp48 is not required for either histone deacetylase or trapoxin binding activity. The result is consistent with the idea that RbAp48 serves a targeting rather than an enzymatic function. Finally, lysates from cells transfected with pBJ5/HD/-F were incubated with the K-trap affinity matrix in the presence or absence of trapoxin and trichostatin. Protein immunoblot analysis demonstrated an interaction between recombinant HDl-F and the K-trap affinity matrix that was fully competed by nanomolar concentrations of trapoxin or trichostatin (Figure 4D).
HDl is 60% identical to the protein encoded by the yeast RPD3 gene, which was isolated in four independent mutant suppressor screens designed to identify transcriptional repressors (13, 16, 17, 18, 19). No biochemical function for the yeast protein has previously been postulated. A negative regulator of the TRK2 gene, RPD3 is necessary for the transcriptional repression of several genes whose expression is regulated according to specific environmental conditions. Loss of RPD3 also leads to decreased transcriptional activation of certain genes, but this effect may be indirect (13, 17). Although RPD3 had yet to be implicated in silencing at telmomeres or the mating loci, the fact that silencing is eliminated by point mutations in specific lysine residues near the NΗ2-terminus of histones H3 and H4 suggests that lysine deacetylation may contribute to the maintenance of silenced chromatin (20, 21, 22, 23). Indeed, silencing at telomeres and the mating loci has been correlated with the presence of hypoacetylated histones, and sir mutants which are defective in silencing show a corresponding increase in the extent of histone acetylation at these loci (24). The SIR3 and SIR4 proteins have been shown to interact with a bacterially expressed histone H4 NH -terminal domain in vitro (25), and it is possible that deacetylation of one or more lysine residues is required for this interaction in vivo. Our results further support a role for histone deacetylase as a transcriptional regulator and establish a biochemical connection to the genetic studies that originally characterized RPD3.
How does inhibition of histone deacetylase in mammalian cells lead to Gi and G2 phase cell cycle arrest? One possibility is that specific cell cycle regulatory proteins such as the cyclin dependent kinase inhibitors are transcriptionally upregulated in response to histone deacetylase inactivation. Altematively, cell cycle checkpoints may exist that monitor histone acetylation or higher-order chromatin stmcture. It should now be possible to study the regulation of histone deacetylase during the cell cycle, its substrate specificity, and the mechanism by which it is targeted to specific regions of the genome. Example 2
3H-Trapoxin was prepared from (S.S)-threitol acetonide (9) by total synthesis, as outlined in Figures 8A-8C.
As shown in Figure 8A, (S,S)-threitol acetonide (9) was monoprotected by treatment with triisopropylsilylchloride (TLPSC1) and sodium hydride in tetrahydrofuran (THF). The free alcohol was then subjected to Swem oxidation. Wittig reaction of die resulting aldehyde gave compound 10 in good yield for the three steps. Compound 10 was then hydrogenated with deprotection of the primary alcohol, which was then converted to the bromide 11 in excellent yield. Bromide 11 was converted to the organocuprate and reacted with (S)-serine β-lactone to yield the benzyloxycarbonyl- (Cbz) protected amino acid 12.
.As shown in Figure 8B, 12 was coupled to tripeptide methyl ester 14, and the methyl ester was saponified. The amino acid was then cyclized and the silyl protecting group was removed to yield cyclotetrapeptide 18 in 51% yield. Cyclotetrapeptide 18 was tritiated, as shown in Figure 8C, by oxidation of the primary alcohol with the Dess-Martin reagent, and the aldehyde was reduced with tritiated sodium borohydride to provide tritiated 18, which was converted to [3H]Trapoxin B by tosylation of the primary alcohol, deprotection of the diol, epoxide ring closure, and oxidation of the secondary alcohol to yield the desired compound. Non-radiolabelled 18 was converted to [3H]Trapoxin B, via tosylate 19, in 68% overall yield.
Example 2
K-Trap was prepared from (S,S)-threitol acetonide (9) by total synthesis, as outlined in Figures 9A-9C. As shown in Figure 9A, monoprotection and Swem oxidation of 9 yielded the aldehyde as above. Wittig homologation yielded carboxylic acid 20, which was converted to the mixed anhydride and treated with lithiated oxazolidinone 21 to provide 22 in excellent yield. Deprotection of the primary alcohol and conversion to the tosylate were followed by treatment of the potassium enolate with trisylazide according to the method of Evans to effect electrophilic azide transfer in good overall yield and stereoselectivity, providing compound 23. Removal of the chiral auxiliary and catalytic reduction of the azido function, with hydrogenation of the olefin, provided amino acid 24, which was N-protected to give the Fmoc derivative 25 in high overall yield. Referring to Figure 9B, protected amino acid 25 was coupled to tripeptide methyl ester 26. The methyl ester was saponified to yield 27, which was cyclized under high- dilution conditions to provide cyclotetrapeptide 28 in 58% yield..
As shown in Figure 9C, compound 28 was converted to K-trap (29) by deprotection of the diol, base-promoted epoxide closure, and oxidation of the secondary alcohol to provide K-trap (29) in good overall yield. The K-trap affinity matrix 30 was provided by palladium-catalyzed removal of the allyloxycarbonyl (Alloc) group from the lysine residue of 29, and immobilization on Affigel 10.
References and Notes for Examples 1-2.
1. B.M. Turner, Cell 75, 5-8 (1993).
2. D.Y. Lee, J.J. Hayes, D. P ss, A.P. Wolffe, Cell 72, 73-84 (1993)
3. S. Kelff, E. D Andrulis, C.W. Anderson, R. Sternglanz, J. Biol. Chem. 270, 24674- 24677 (1995). 4. M. Yoshida, S. Horinouchi, T. Beppu, Bioessays 17, 423-30 (1995)
5. M. Kijima, M. Yoshida, K. Sugita, S. Horinouchi, T. Beppu, J. Biol. Chem. 268, 22429-35 (1993).
6. J. Tauton, J.L. Collins, S.L. Schreiber manuscript in preparation. 8. J. Taunton, CA. Hassig, S.L. Schreiber, unpublished results. 11. Y.W. Qian, et al., Nature 364, 648-52 (1993).
12. E. J. Neer, CJ. Schmidt, R. Nambudripad, T.F. Smith, Nature 371, 297-300 (1994).
13. M. Vidal, R.F. Gaber, Mol. Cell. Biol. 11, 6317-27 (1991).
15. Control experiments indicated that competitor peptides had no effect on histone deacetylase activity per se.
16. K. Nasmuth, D.J. Stillman, D. Kipling, Cell 48, 579-87 (1987).
17. D.J. Stillman, S. Dorland, Y.Yu, Genetics 136, 781-8 (1994).
18. E.A. McKenzie, et al., Mol. Gen. Genet. 240, 374-86 (1993).
19. K. S. Bowdish, A.P. Mitchell, Mol. Cell. Biol. 13, 2172-81 (1993). 20. L.M. Johnson, P.S. Kayne, E.S. Kahn, M. Gmnstein, Proc. Natl. Acad. Sci. U.S.A. 87, 6286-90 (1990).
21. P.C. Megee, B.A. Morgan, B.A. Mittman, M.M. Smith, Science 247, 841-5 (1990).
22. E.C. Park, J.W. Szostak, Mol. Cell. Biol. 10, 4932-4 (1990).
23. O.M. Aparicio, B.L. Billington, D.E. Gottschling, Cell 66, 1279-87 (1991). 24. M. Braunstein, A.B. Rose, S.G. Holmes, CD. Allis, J.R. Borach, Genes Dev. 7, 592-604 (1993).
25. A. Hecht, T. Laroche, S.Strahl Bolsinger, S.M. Gasser, M. Gmnstein, Cell 80, 583- 92 (1995). 26. N.A. Clipstone, G.R. Crabtree, Nature 357, 695-7 (1992).
Example 3
Histone Deacetylase Activity is Required for Full Transcriptional Repression by mSin3 A
The Mad family of basic region-Helix-Loop-Helix-Leucine Zipper (BHLHZip) proteins play an important role in controlling cell proliferation and differentiation (for reviews see: Amati and Land, 1994; Bernards 1995). Four identified Mad family members: Madl, Mxil, Mad3 and Mad4 (Ayer et al., 1993; Zervos et al., 1993; Hurlin et al., 1995a) form heterodimers with another BHLHZip protein, Max; to repress transcription (Ayer et al., 1993;,Hurlin et al., 1995a) and are thought to play a negative role in the control of cell proliferation.
Two mammalian homologs of the Saccharomyces cerevisiae transcriptional corepressor SIN3, mSin3A and mSin3B, have recently been identified as Mad interacting proteins and are required for Mad-mediated transcriptional repression (Ayer et al., 1995; Schreiber-Agus et al., 1995). The most conserved regions of these proteins correspond to four putative paired amphipathic helix (PAH) motifs, which have been proposed to constitute protein-protein interaction surfaces (Wang et al., 1990). The second PAH motif in mSin3A, mSin3B and Sin3p interacts with the mSin3 interaction domain or SID in the amino terminus of the four Mad family members (Ayer et al., 1995; Schreiber-Agus et al., 1995; Hurlin et al., 1995a; Kasten et al., 1996). Madl, Max and mSin3A form ternary complexes capable of binding DNA (Ayer et al., 1995). Point mutations in the SID domain of Madl dis pt its ability to bind mSin3A, negate its function as a trancriptional repressor (Ayer et al., 1995), and eliminate Madl function in several biological assays (Koskinen et al., 1995; Roussel et al., 1996). These findings suggest diat Mad:Max heterocomplexes repress transcription by tethering either mSin3A or mSin3B to DNA. A chimeric protein fusing the SDD of Madl to the GAL4 DNA-binding domain results in repression of simple and complex promoters in a manner that is dependent on mSin3 binding, suggesting that targeting mSin3 to DNA is necessary for repression (Ayer et al., 1996). Nevertheless, the molecular mechanism(s) for mSin3A— mediated repression remain unknown.
As described in example 1, a mammalian histone deacetylase has been identified, and cDNAs encoding the protein, histone deacetylase 1 (HDl or HDACl for the puφoses of this example), have been cloned (see also Taunton et al., 1996b). IBD AC 1 is approximately 60% identical to the S. cerevisiae RPD3 protein, which is a component of a yeast histone deacetylase complex (Rundlett et al., 1996). Single mutations in either RPD3 or SIN3 give the same phenotypes as RPD3/SIN3 double, mutants suggesting that they function in the same pathway (Stillman et al., 1994). Because Mad family proteins use mSin3A as a corepressor and Madl can repress transcription in wild-type yeast but not yeast having a null mutation in SIN3 (Kasten et al., 1996) or RPD3 (D.J. Stillman, personal comm.), it is likely that the mechanism of transcriptional repression by Mad proteins may be conserved between yeast and higher eukaryotes. Consistent with this hypothesis, the DNA-binding transcription factor YY1 interacts with a mammalian RPD3 homolog to repress transcription of a heterologous reporter gene (Yang et. at, 1996). These results demonstrate that mammalian RPD3-like activity functions in transcriptional regulation.
Several lines of evidence suggest that the acetylation status of conserved lysines in the amino terminal domains of histones H3 and H4 play a role in die regulation of transcription. In general, histone hyperacetylation correlates with transcriptionally active or poised genes; conversely, hypoacetylation correlates with transcriptionally repressed heterochromatin (for reviews see: Turner, 1993; Loidl, 1994; Wolffe, 1996). While little is known about the targeting and regulation of histone acetyltransferases and deacetylases, it has been recently shown that several transcriptional coactivators possess inherent acetyltransferase activity (Brownell et al., 1996; Ogryzko et al., 1996) or associate with acetyltransferases (Yang et al., 1996b). We report that mSin3A and HDACl associate in vivo and that the histone deacetylase inhibitor trapoxin interferes with mSin3 A-mediated transcriptional repression.
Results
(i) mSin3A is present in cells as a large stable multiprotein complex.
To study the in vivo function of mSin3A we generated polyclonal antiserum specific for the PAH2 domain of mSin3A. We tested this antiserum by immunoprecipitation using nuclear lysates made from the myeloid leukemia cell line U937 that had been metabolically labeled with S-methionine. Analysis of immunoprecipitates showed an intensely-labeled doublet with an apparent molecular weight of 150 kiloDaltons that was present in the anti-mSin3A immunoprecipitates (Figure 10A). This doublet comigrated with in vitro translated-mSin3A, shared identical V8 protease digestion peptides with in vitro translated-Sin3A and was absent from immunoprecipitations using preimmune semm or immune semm preincubated with the cognate immunogen (data not shown).
Fractionation of U937 nuclear extracts by size exclusion chromatography indicated that mSin3A is present in large molecular weight complexes) (D.E.A., unpublished). To address this possibility, we performed immunoprecipitations from metabolically-labeled U937 cells under conditions that should preserve protein-protein interactions. In addition to mSin3A, the low-stringency mSin3A immunoprecipitates contained several labeled polypeptides of apparent molecular weight 250 kDa, 180 kDa, 55 kDa, 50 kDa, 42 kDa, 33-36 kDa and 30 kDa (Figure 10A). These proteins were not detected in immunoprecipitates using mSin3A antisemm blocked with the cognate immunogen, suggesting that the proteins detected are specifically associated with mSin3A. Furthermore, none of these proteins were detected using high-stringency immunoprecipitation or by western blotting of whole-cell lysates using anti-Sin3A, suggesting that they do not share epitopes with mSin3A and are not proteolytic breakdown products of mSin3A (data not shown). All of the associated proteins appear to be present in substoichiometric amounts to mSin3A, suggesting that mSin3A complexes are heterogeneous.
To test the stability of the mSin3 A complex, we subjected low-stringency mSin3A immunoprecipitates to different salt concentrations and ionic detergent conditions. The proteins that remained bound to mSin3A in the immunecomplex were analyzed by SDS- PAGE. Under the most stringent conditions we observed only a slight loss of mSin3A- associated proteins in the immune complex (Figure 10B). One exception to this finding was the apparently quantitative loss of p42 under slightly-elevated salt concentrations. These findings demonstrate that the mSin3A complex is stable in vivo and suggests that some or all of the mSin3A-associated proteins may facilitate mSin3A function as a transcriptional co-repressor.
(ii) HDACl and RbAp48 are components of the mSin3A complex. Because SIN3 and RPD3 appear to function in the same pathway in yeast and two components of the mSin3A complex, p50 and p55, are similar in apparent molecular weight to HDACl we hypothesized that HDACl or related proteins might be components of the mSin3A repressor complex. To test this hypothesis, the proteins bound to mSin3A immunecomplexes were eluted with ionic detergents and reprecipitated with affinity purified antibodies specific for an internal peptide of HDACl. Only two proteins eluted from the mSin3A complex were re-precipitated by HDACl antisemm (Figure 11 A). These polypeptides comigrated with p50 and p55 from the low stringency mSin3A immunoprecipitation, suggesting that proteins highly related to HDACl are complexed to mSin3A in vivo. p55 comigrates with in vitro translated HDACl and is recognized by an antibody specific for a carboxy-terminal epitope unique to HDACl. Another CDNA encoding an HDACl homolog, HDAC2, has recently been identified (Yang et al., 1996a). It is likely that p50 represents HDAC2 (data not shown).
In a reciprocal experiment, we performed low stringency immunoprecipitations using antisemm specific for an epitope at the carboxy-terminus of HDACl. HDACl immunoprecip-itates contain several proteins that were specifically competed with the immunizing peptide (Figure 1 IB). A polypeptide doublet that comigrated with mSin3A was detected in the HDACl immunocomplexes (Figure I IB and 11C). To confirm that the doublet coprecipitating with HDACl is mSin3A, the HDACl immunocomplex was eluted and reprecipitated with antisemm specific for mSin3A (Figure 11C). The two proteins in this precipitate comigrated with mSin3A, confirming that mSin3A and HDACl are associated in vivo.
To determine whether HDACl associated with mSin3A in vivo is enzymatically active, we assayed low-stringency immunoprecipitates for histone deacetylase activity. We used a synthetic peptide corresponding to the first twenty four amino acids of histone H4 as a substrate for our deacetylase assay (Taunton et al., 1996b). Low-stringency anti- mSin3A immunoprecipitates contained deacetylase activity; however, only background levels of deacetylase activity were detected in the immunoprecipitates if the mSin3A anti¬ semm was blocked with cognate immunogen (Figure 1 ID). To confirm the authenticity of the mSin3A associated activity we treated the immunoprecipitates with synthetic trapoxin, a specific inhibitor of histone deacetylase activity (Taunton et al., 1996a). Treatment of mSin3A complexes in vitro with 10 nM trapoxin reduced deacetylation by approximately 50% (Figure HD), suggesting that the precipitated deacetylase activity can be attributed to trapoxin-sensitive histone deacetylases bound to mSin3A. We detected an interaction between HDACl and RbAp48 in vivo (Figure IIB, 11C and Example 1). The low-stringency mSin3A immunoprecipitation shown in Figure 1 IC also contained a protein that comigrated with RbAp48 (marked with an asterisk) that was not readily visible on the shorter exposures of low stringency mSin3 immunoprecipitations (Figure 11 A). We have identified RbAp48 in mSin3A immunoprecipitates from cell extracts of nontransfected cells by western blotting, further demonstrating that mSin3A and RbAp48 associate in vivo (Figure 12 A).
To address further the association between HDACl and RbAp48 with mSin3A, we expressed the mammalian proteins in insect cells using recombinant baculoviruses. To this end, we expressed recombinant FLAG-epitope tagged HDACl (HDACl -F) that could be immuno-purified by anti-FLAG antibodies and histidine-tagged mSin3A (mSin3A-H) that could be purified by nickel affinity (data not shown). HDACl -F was immunoprecipitated from infected Sf9 cell extracts by anti-FLAG antibodies in the presence or absence of mSin3A-H. HDAC1-F was also precipitated by Ni2+-NTA agarose in a manner that was dependent on coexpression of mSin3A-H (Figure 12B), demonstrating that a complex between HDAC 1 and mSin3A is formed in insect cells using exogenously expressed human proteins.
Consistent with our finding that RbAp48 is associated with mSin3A and HDACl in vivo, we show that baculovirus expressed Flu-epitope tagged RbAp48 (p48-HA) is specifically precipitated from infected Sf9 cell extracts using anti-FLAG antibody only when HDAC1-F is coexpressed. Furthermore, p48-HA is specifically retained by Ni2+- NTA in the presence of mSin3A-H (Figure 12C). Co-expression of p48-HA did not appear to effect the association between HDAC1-F and mSin3A-H, suggesting that the regions of interaction are distinct and that all three proteins can associate simultaneously. These data suggest a direct interaction between mSin3A, HDACl and RbAp48 in vivo.
(ϋi) Transcription repression by mSin3A requires histone deacetylase activity.
To investigate whether histone deacetylation plays a role in mSin3A-mediated transcriptional repression in vivo, we examined mSin3A-specific repression in the presence and absence of the histone deacetylase inhibitor trapoxin. 293 cells were transfected with a luciferase reporter gene constmct containing a minimal promoter consisting of only a TATA box and initiation site derived from the myelomonocytic growth factor gene (Figure 13 A). This reporter has four consensus binding sites for the DNA binding domain of the S. cerevisiae transcriptional activator GAL4 and therefore is responsive to chimeric proteins containing the GAL4 DNA binding domain (GALDBD) (Stemeck et al., 1992). We have used this reporter construct previously to demonstrate that fusion of the SID repressor region of Madl to the GALDBD is necessary for mSin3A-dependent transcriptional repression. Furthermore, we have shown that fusion of SID to the potent transcriptional activator GALVP16, MadN35GALVP16, can cancel the activation function of VPI6 in an mSin3 A-dependent manner. Consistent with our previous results (Ayer et al., 1996), MadN35GALVP16 activated transcription from the reporter gene approximately 100-fold less well than GALVP16 (data not shown). As a negative control we engineered two proline substitutions into the SID of Madl, Mad(Pro); this protein cannot bind mSin3 A in vitro (Ayer et al., 1995). Consistent with an inability to interact with mSin3A, Mad(Pro)GALVP16 is a much less potent repressor (Figure 13B). In control experiments we have shown that the observed effects require the presence of GAL4 sites in the promoter and that both MadN35GALVP16 and Mad(Pro)N35GALVP16 are expressed to equivalent levels in these cells and bind GAL4 sites with similar affinities (data not shown). To test the role of histone deacetylation on the repression observed in our transfection assays, we first examined the effect of trapoxin on histone deacetylase activity in 293 cells. As expected, in vivo treatment with 10 nM trapoxin for eight hours reduced deacetylase activity of both cmde 293 extracts and anti-HDACl immunopurified complexes by approximately 46% and 58%, respectively (Figure 13 C) .
To test the effect of a histone deacetylase inhibitor on MadN35GALVP16 and Mad(Pro)N35GALVPI6 mediated repression, we treated a duplicate set of transfections with 10 nM trapoxin for eight hours prior to harvest. In the representative experiment shown, 10 nM trapoxin treatment derepressed the activity of MadN35GALVP16 nine- fold while it had little effect on the activity of Mad(Pro)N35GALV 16, suggesting that the histone deacetylation plays a direct role in mSin3 A transcriptional repression (Figure 13B). In addition, there was typically less than a two-fold effect of trapoxin on the activity of the reporter cene in cells transfected with the expression vector alone or in cells transfected with GALVP16 (data not shown). Following trapoxin treatment, the repression observed for MadN35GALVP16 was still seven times greater than that of Mad(Pro)N35GALVP16, suggesting that the residual deacetylase activity following trapoxin treatment (Figure 13B) continues to drive mSin3A-mediated repression; however, we can not rule out that mSin3A is capable of repression by mechanisms independent of histone deacetylation. Discussion
Earlier studies implicated mSin3 as the primary candidate for the negative transcriptional function of the DNA binding transcription factor Mad (Ayer et al., 1995; Schreiber-Agus et al. 1995; Ayer et al., 1996). We present evidence that the mSin3A compressor is part of a high molecular weight, multicomponent complex(es) that contains active histone deacetylase, thereby implicating histone deacetylation as a potential mechanism for mSin3A-mediated repression. Furthermore, we observe a pronounced increase in the transcriptional activity of an mSin3 A- silenced reporter gene upon treatment in vivo with the specific histone deacetylase inhibitor trapoxin, suggesting that full transcriptional repression by mSin3A requires histone deacetylation. These results suggest a mechanism of gene regulation through the targeting of an enzyme that alters chromatin stmcture.
These observations are consistent with genetic experiments in yeast, suggesting that the yeast orthologs of mSin3A and HDACl, SIN3 and RPD3 respectively, are epistatic transcriptional regulators (Stillman et al., 1994). Furthermore, recent biochemical evidence demonstrates that Rpd3p is a component of a large molecular weight histone deacetylase complex in yeast (Rundlett et al., 1996). Together with our results, these findings predict a conservation of the mSin3/HDACl functional association in yeast. We have used chimeric transcriptional regulators to discern the effects of trapoxin on the activity of our reporter genes. The MadN35GALVP16 chimera functioned as a repressor by a mechanism that was dependent on the binding of mSin3 A and that was sensitive to trapoxin. The same mutations that inactivate MadN35GALVP16 as a transcriptional repressor (i.e. Mad(Pro)N35GALVP16), also block interaction between Madl and mSin3A in vitro and Madl function in vivo. Therefore, it is likely that Mad:Max heterocomplexes repress transcription in a manner dependent on an mSin3A- associated histone deacetylase.
By co-immunoprecipitation we have demonstrated that mSin3A and HDACl associate in vivo. Consistent with these data we observed nuclear colocalization of mSin3A and HDACl by immunofluorescence microscopy (data not shown). Finally, overexpression in insect cells facilitates co-purification of mSin3A and HDACl (Figure 12B and C), suggesting that the interaction between mSin3A and HDACl is either direct or requires a conserved cofactor. The finding that mSin3A has different associated histone deacetylases (HDACl and HDAC2) suggests that the mSin3A complex(es) may have multiple substrate or target specificities. The heterogeneous nature of the mSin3 A complex potentially reflects a diverse array of repressors, histone deacetylases and different targeting molecules that facilitate mSin3A-dependent alterations in gene expression. At least five additional polypeptides are stably associated with mSin3A (Figures
10 and 11); whose function is currently unknown but tight association with mSin3A in both U937 cells and Jurkat T cells (data not shown) suggests that they in some way mediate mSin3A function. Furthermore, we have identified an association between mSin3A and RbAp48 in vivo, suggesting that this protein may play a role in regulating mSin3A-targeted deacetylation. RbAp48 was originally identified as a retinoblastoma binding protein that contains WD repeats and shares homology with the -subunit of G- proteins (Qian et al., 1993). Subsequently, it has been shown that RbAp48 or its orthologs are involved in targeting different histone modifying enzymes to chromatin (Parthun et al., 1996; Taunton et al., 1996b; Tyler et al., 1996; Verreault et al., 1996). The mSin3 A/RbAp48 complex isolated from U937 cells (Figure 11) is likely to represent only a small fraction of the mSin3 A complexes, but its detection implies that mSin3A may play a role in the control of different aspects of chromatin physiology as well as transcription repression.
It is unclear how different chromatin states facilitate transcription repression and activation or how their distinct biochemical states arise; however, there is ample cytological, genetic and biochemical evidence supporting the model that hyperacetylated chromatin is transcriptionally more active than hypoacetylated chromatin. Acetylation levels in -heterochromatin of Drosophila melanogaster polytene chromosomes are significantly reduced at lysine positions 5, 8, and 16 of histone H4, while the transcriptionally hyperactive X-chromosome of male flies is uniquely hyperacetylated at position 16 (Turner et al., 1992). In yeast, mutation of acetyl-accepting lysines in histone H4 reduces the activity of the GALI, PHOS and CUPl promoters in vivo (Durrin et al., 1991). The transcriptionally silent regions in yeast, HML and HMR, are hypoacetylated and their activation is correlated with acetylation of histone H4 (Braunstein et al„ 1993). Additionally, biochemical studies showed that certain transcription factors have higher affinity for their binding sites when those sites are embedded in chromatin assembled from hyperacetylated histones (Lee et al., 1993; Vettes-Dadey al., 1996). Finally, evidence suggesting that acetylation is required for activation comes from the recent demonstration diat several coactivators either encode acetyltransferases or are associated widi acetyltransferases. Thus, our data support this general model for the control of gene expression by histone acetylation status and provide a biochemical mechanism for deacetylation-mediated repression.
The acetylalion status of a particular chromatin region represents a balance between competing, acetylation and deacetylation reactions. We propose that MadN35GALVPI6 recmits mSin3A-HDAC complexes to specific sites on DNA and shifts this equilibrium towards deacetylation and subsequent transcription repression by creating a high effective molarity of the histone deacetylase. In yeast, the activation domain of VPI 6 has been shown to use the acetyltransferase Gen5p as a coactivator (Marcus et al., 1994; Brownell et al., 1996), suggesting that in mammalian cells VP16 will also use an acetyltransferase as a cofactor. Thus, trapoxin treatment could shift the equilibrium from deacetylation to acetylation and thereby drive activation.
Whether histone deacetylation will always have a negative effect on gene expression is unclear. Mutants in SIN3 and RPD3 can have both positive and negative effects on gene expression (Vidal and Gaber, 1991; Yoshimoto et al., 1992); however, for SIN3 there is evidence that positive effects may be indirect (Wang et al., 1994). In addition, mutations or deletions in RPD3 have recently been shown to enhance telomeric silencing both in yeast and in fruit fly (Sussel et al., 1995; De Rubertis et al., 1996; Rundlert et al., 1996). In mammalian cells, deacetylase inhibitors can inhibit MyoD- (Johnston et al., 1992) and steroid receptor-activated transcription (McKnight et al., 1990; Bresnick et al., 1990). While it remains to be shown that the effects of RPD3 on silencing are direct, this evidence suggests that histone deacetylation can elicit both positive and negative effects on gene expression. Determining the factors diat govern the functional outcome of histone deacetylation will provide fertile ground for further experimentation.
Experimental Procedures
Antibodies, cell culture, and Immunoprecipitations: To generate antisemm specific for mSin3A a GST fusion protein encoding, amino acids 251 through 405 of mSin3A was used to immunize a New Zealand White rabbit. The cmde semm was passed over a GST column to remove the anti-GST antibodies. U937 cells were grown in RPMI supplemented with 10% calf semm (Hyclone), glutamine and penicillin- streptomycin. Low and high stringency immunoprecipitations were performed essentially as described (Ayer and Eisenman, 1993). To elute proteins from low stringency immunoprecipitates, they were incubated for 60 minutes at room temperature in antibody buffer and reprecipitated under high stringency conditions.
Luciferase assays: 293 cells were seeded in triplicate onto 60 mm dishes at 3x10s cells in 4 ml DME with 10% calf semm (Hyclone). Six hours after seeding, cells were transfected with 50 ng luciferase reporter, 50 ng CMV-β-gal, 50 ng expression constmct, and 2.85 μg carrier DNA using the BBS/CaPO method. 10 nM trapoxin was added to the media 8 hours prior to the luciferase assays. Cell lysates were prepared 20-24 hours following transfections, and luciferase and β-galactosidase activities was assayed according to manufacturer directions (Promega, Tropix). Luciferase values (relative light units) were normalized for transfection efficiency by dividing by β-gal activity.
Histone deacetylase assays: In vitro histone deacetylase activity was assayed essentially as described with either 50 μl of cmde cell extract (approximately 5 X IO6 Cells) or immunopurified cell extracts (approximately 2 X IO7 cells) for 2.5 hrs at 37 C (Taunton et al., 1996b). Pretreatment of cmde or immunopurified extract with synthetic trapoxin was performed for 30 minutes at 4 C prior to addition of peptide substrate. TAg Jurkat and 293 cell extracts for histone deacetylase assays were prepared as in Taunton et al., 1996. Anti-HDACl and anti-mSin3A immunoprecipitations were performed as described above and in Figure 11. The Protein-A conjugated immunoprecipitates were washed three times in J-buffer plus I mM EDTA and resuspended in J-buffer without Triton-X-100, and histone deacetylase activity was measured as described.
Baculovimses: cDNAs encoding Flag-tagged HDACl, HA-tagged RbAp48 and His-tagged mSin3A were cloned into the transfer vector pVL 1392 (specific details on the constmction of these vectors is available upon request). Recombinant vims was generated using Baculogold DNA according to the manufactures instmctions (Pharminigen). Sf9 or High 5 cells were infected at high multiplicity, extracts prepared 48 hours post infection and immunoprecipitations performed as described above. Ni2+- NTA agarose and anti-Flag antibody were purchased from Qiagen and Kodak-IBI, respectively.
References
Ayer, D. E., and Eisenman, R. N. (1993). A switch from Myc:Max to Mad:Max heterocomplexes accompanies monocyte macrophage differentiation. Genes Dev. 1, 2110-9. Ayer, D. E., Kretzner, L., and Eisenman, R.N. (1993). Mad: a heterodimeric partner for Max that antagonizes Myc transcriptional activity. Cell 72, 211-22.
Ayer, D. E., Laherty, C D., Lawrence, Q. A., Armstrong, A., and Eisenman, R. N. (1996). Mad proteins contain a dominant transcription repression domain. Mol. Cell. Biol. 16, 5772-5781.
Ayer, D. E., Lawrence, Q. A, and Eisenman, R. N. (1995). Mad-Max transcriptional repression is mediated by ternary complex formation with mammalian homologs of yeast repressor Sin3. Cell 80, 767-776.
Braunstein, M., Rose, A. B., Holmes, S. G., Allis, C. D., and Broach, J. R. (1993).
Transcriptional silencing in yeast is associated with reduced nucleosome acetylation.
Genes Dev. 7, 592-604.
Bresnick, E. H., John, S., Berard, D. S., LeFebvre, P, and Hager, G. L. (1990).
Glucocorticoid receptor-dependent disruption of a specific nucleosome on the mouse mammary tumor vims promoter is prevented by sodium butyrate. Proc. Natl. Acad Sci.
USA 87, 3977-81.
Brownell, J. E, Zhou, J., Ranalli, T., Kobayashi, R, Edmondson, D. G., Roth, S. Y., and
Allis, C. D. (1996). Tetrahymena histone acetyltransferase A: a homolog to yeast Gcn5p linking histone acetylation to gene activation. Cell 84, 843-851. De Rubertis, F., Kadosh, D., Henchoz, S., Pauli, D., Reuter, G., Struhl, K., and Spierer, P. (1996). The histone deacetylase RPD3 counteracts genomic silencing in Drosophila and yeast. Nature 384, 589-591.
Durrin, L.K., Mann, R.K., Kayne, P.S. and Gmnstein, M. (1991). Yeast histone H4 N- terminal sequence is required for promoter activation in vivo. Cell 65, 1023-31.
Hurlin, P.J., Queva, C, Koskinen, P. J., Steingrimsson, E., Ayer, D. E., Copeland, N. G., Jenkins, N. A., and Eisenman, R. N. (1995a). Mad3 and Mad4: Novel Max-interacting transcriptional repressors that suppress c-Myc-dependent transformation and are expressed during, neural and epidermal differentiation. EMBO J 14, 5646-5659.
Johnston, L. A., Tapscott, S. J., and Eisen, H. (1992). Sodium butyrate inhibits myogenesis by interfering with the transcriptional activation function of MyoD and myogenin. Mol. Cell. Biol. 12, 512330.
Kasten, M. M., Ayer, D. E., and Stillman, D. J. (1996). SIN3 -dependent transcriptional repression by interaction with the Madl DNA binding, protein. Mol. Cell. Biol. 16, 4215-4221. Lee, D. Y., Hayes, J. J., Pmss, D., and Wolffe, A. P. (1993). A positive role for histone acetylation in transcription factor access to nucleosomal DNA. Cell 72, 73-84.
Marcus, G. A., Silverman, N., Bemer, S. L., Horiuchi, J., and Guarente, L. (1994). Functional similarity and physical association between GCN5 and ADA2: putative transcriptional adaptors. EMBO J. 13, 4807-15.
McKnight, G. S., Hager, L., and Palmiter, R. D. (1980). Butyrate and related inhibitors of histone deacetylation block the induction of egg white genes by steroid hormones. Cell 22, 469-77.
Ogryzko, V. V., Schiltz, R. L., Russanova, V., Howard, B. H., and Nakatani, Y. (1996). The transcriptional activators p3OO and CBP are histone acetylases- Cell 87, 953-959, Parthun, M. R., Windom, J., and Gottschling, D. E. (1996). The major cytoplasmic histone acetyltransferase in yeast: links to chromatin replication and histone metabolism Cell 87, 85-94.
Quan, Y. W., Wang, Y. C, HoUingsworth, R. E., Jr., Jones, D., Ling, N., and Lee, E. Y. (1993). A retinoblastoma-binding protein related to a negative regulator of Ras in yeast. Nature 364, 648-52.
Rundlett, S. E., Carmen, A. A., Kobayashi, R., Bavykin, S., Turner, B. M., and Gmnstein, M. (1996). HDA1 and RPD3 are members of distinct yeast histone deacetylase complexes that regulate silencing and transcription. Proc. Natl. Aca Sci. USA 93, 14503-14508.
Schreiber-Agus, N., Chin, L., Chen, K., Torres, R., Rao, G., Guida, P., Skoultchi, A. I., and DePinho, R. A. (1995). An amino-terminal domain of Mxi I mediates anti-Myc oncogenic activity and interacts with a homolog of the yeast transcriptional repressor SIN3- Cell 80, 777-786.
Sterneck, E., Blattner, C, Graf, T., and Leutz, A. (1992). Stmcture of the chicken myelomonocytic growth factor gene and specific activation of its promoter in avian myelomonocytic cells by protein kinases. Mol. Cell. Biol. 12, 1728-35.
Stillman, D. J., Dorland, S., and Yu, Y. (1994). Epistasis analysis of suppressor mutations that allow HO expression in the absence of the yeast SWI5 transcriptional activator. Genetics 136, 781-8.
Sussel, L., Vannier, D., and Shore, D. (1995). Suppressors of defective silencing, in yeast: effects on transcriptional repression at the HMR locus, cell growth and telomere stmcture. Genetics 141, 873-888. Taunton, J., Collins, J. L., and Schreiber, S. L. (1996a). Synthesis of Natural and Modified Trapoxins, Useful Reagents for Exploring Histone Deacetylase Function. J. Am. Chem. Soc. 118, 10412-10422.
Taunton, J., Hassig, C. A., and Schreiber, S. L. (1996b). A mammalian histone deacetylase related to the yeast transcriptional regulator Rpd3p. Science 272, 408-411.
Turner, B. M., Birley, A. J., and Lavender, J. (1992). Histone H4 isoforms acetylated at specific lysine residues define individual chromosomes and chromatin domains in Drosophila polytene nuclei. Cell 69, 375-84.
Tyler, J. K., Bulger, M., Kamakaka, R. T., Koybaysbi, R., and Kadonaga, J. K. (1996). The p55 subunit of Drosophila chromatin assembly factor 1 is homologous to a histone deacetylase-associated protein. Mol. Cell. Biol. 16, 6149-6159,
Verreault, A., Kaufman, P. D., Kobayashi, R., and Stillman, B. (1996). Nucleosome assembly by a complex of CAF-1 and acetylated histones H3/H4. Cell 87,95-104.
Vettese-Dadey, M., Grant, P. A, Hebbes, T. R., Crane-Robinson, C, All's, CD., and Workman, J.L. (1996). Acetylation of histone H4 plays a primary role in enhancing transcription factor binding to nucleosomal DNA in vitro. EMBOJ. 15, 2508-2518.
Vidal, M., and Gaber, R. F. (1991). RPD3 encodes a second factor required to achieve maximum positive and negative transcriptional states in Saccharomyces cerevisiae. Mol. Cell. Biol. 11, 6317-27.
Wang, H., Clark, I., Nicholson, P. R., Herskowitz, I., and Stillman, D. J. (1990). The Saccharomyces cerevisiae SIN3 gene, a negative regulator of HO, contains four paired amphipathic helix motifs. Mol. Cell. Biol. 10, 5927-36.
Wang, H., Reynolds-Hager, L., and Stillman, D. J. (1994). Genetic interactions between SIN3 mutations and the Saccharomyces cerevisiae transcriptional activators encoded by MCMI, STE12, and SWII. Mol. Gen. Genet. 245, 675-685.
Yang, W.-M., Inouye, C, Zeng, Y., Bearss, D., and Seto, E. (1996a). Transcriptional repression by YY1 is mediated by interaction with a mammalian homolog of the yeast global regulator RPD3. Proc. Natl. Acad Sci. USA 93, 12845-12850,
Yang, X. J., Ogryzko, V. V., Nishikawa, J., Howard, B. H., and Nakatani, Y. (1996b)- A p300/CBP-associated factor that competes with the adenoviral oncoprotein EIA. Nature 382, 319-324.
Yoshimoto, H., Ohmae, M., and Yamashita, I. (1992). The Saccharomyces cerevisiae GAM2/SIN3 protein plays a role in both activation and repression of transcription. Mol. Gen. Genet. 233, 327-30. Zervos, A. S., Gyuris, J., and Brent, R. (1993). Mxil, a protein that specifically interacts with Max to bind Myc-Max recognition sites. Cell 72, 223-32-
Example 4
Since the priority date of this application, a number of other mammalian HDx genes have been described in the literature. In particular, a mouse HDl clone is identified in GenBank as accession number U807080. Another HDx member, HD2 (HDAC-2) is also described for both human and mouse; see for example, GenBank entries U31814 and U31758. Without exception, each clone includes a v motiff represented in the general formula of SEQ LD No. 12, and a χ motif represented in die general formula SEQ ID No. 14.
All of the above-cited references and publications are hereby incoφorated by reference.
Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific polypeptides, nucleic acids, methods, assays and reagents described herein. Such equivalents are considered to be within the scope of this invention and are covered by the following claims.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Schreiber, Stuart L. Taunton, Jack Haβsig, Christian A. Jamison, Timothy F.
(ii) TITLE OF INVENTION: Hiβtonβ Deacetylases and Uses Related Thereto
(iii) NUMBER OF SEQUENCES: 15
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: FOLEY, HOAG & ELIOT, LLP
(B) STREET: One Post Office Square
(C) CITY: Boston
(D) STATE: MA
(E) COUNTRY: USA
(F) ZIP: 02109
(v) COMPUTER READABLE FORM: (A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: ASCII(text) (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: US
(B) FILING DATE: 26-MAR-1996
(C) CLASSIFICATION: (viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Vincent, Matthew P.
(B) REGISTRATION NUMBER: 36,709
(C) REFERENCE/DOCKET NUMBER: HUV019.25 (ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (617) 832-1000
(B) TELEFAX: (617) 832-7000
(2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1449 base pairβ
(B) TYPE: nucleic acid
(C) STRANDEDNESS: both
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA ( ix ) FEATURE :
(A) NAME/KEY: CDS
(B) LOCATION: 1..1446
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:
ATG GCG CAG ACG CAG GGC ACC CGG AGG AAA GTC TGT TAC TAC TAC GAC 48 Met Ala Gin Thr Gin Gly Thr Arg Arg Lys Val Cys Tyr Tyr Tyr Aβp 1 5 10 15
GGG GAT GTT GGA AAT TAC TAT TAT GGA CAA GGC CAC CCA ATG AAG CCT 96 Gly Asp Val Gly Asn Tyr Tyr Tyr Gly Gin Gly His Pro Met Lys Pro 20 25 30
CAC CGA ATC CGC ATG ACT CAT AAT TTG CTG CTC AAC TAT GGT CTC TAC 144 His Arg lie Arg Met Thr His Asn Leu Leu Leu Asn Tyr Gly Leu Tyr 35 40 45
CGA AAA ATG GAA ATC TAT CGC CCT CAC AAA GCC AAT GCT GAG GAG ATG 192 Arg Lys Met Glu lie Tyr Arg Pro His Lys Ala Asn Ala Glu Glu Met 50 55 60 ACC AAG TAC CAC AGC GAT GA'C TAC ATT AAA TTC TTG CGC TCC ATC CGT 240 Thr Lys Tyr His Ser Asp Asp Tyr lie Lys Phe Leu Arg Ser lie Arg 65 70 75 80
CCA GAT AAC ATG TCG GAG TAC AGC AAG CAG ATG CAG AGA TTC AAC GTT 288 Pro Asp Asn Met Ser Glu Tyr Ser Lys Gin Met Gin Arg Phe Asn Val
85 90 95
GGT GAG GAC TGT CCA GTA TTC GAT GGC CTG TTT GAG TTC TGT CAG TTG 336 Gly Glu Asp Cys Pro Val Phe Asp Gly Leu Phe Glu Phe Cys Gin Leu 100 105 110
TCT ACT GGT GGT TCT GTG GCA AGT GCT GTG AAA CTT AAT AAG CAG CAG 384 Ser Thr Gly Gly Ser Val Ala Ser Ala Val Lys Leu Asn Lys Gin Gin 115 120 125
ACG GAC ATC GCT GTG AAT TGG GCT GGG GGG CTG CAC CAT GCA AAG AAG 432 Thr Asp lie Ala Val Asn Trp Ala Gly Gly Leu His His Ala Lys Lys 130 135 140 TCC GAG GCA TCT GGC TTC TGT TAC GTC AAT GAT ATC GTC TTG GCC ATC 480 Ser Glu Ala Ser Gly Phe Cys Tyr Val Asn Asp lie Val Leu Ala He 145 150 155 160
CTG GAA CTG CTA AAG TAT CAC CAG AGG GTG CTG TAC ATT GAC ATT GAT 528 Leu Glu Leu Leu Lys Tyr His Gin Arg Val Leu Tyr He Aβp He Asp
165 170 175
ATT CAC CAT GGT GAC GGC GTG GAA GAG GCC TTC TAC ACC ACG GAC CGG 576 He His His Gly Asp Gly Val Glu Glu Ala Phe Tyr Thr Thr Asp Arg 180 185 190 GTC ATG ACT GTG TCC TTT CAT AAG TAT GGA GAG TAC TTC CCA GGA ACT 624
Val Met Thr Val Ser Phe His Lys Tyr Gly Glu Tyr Phe Pro Gly Thr
195 200 205
GGG GAC CTA CGG GAT ATC GGG GCT GGC AAA GGC AAG TAT TAT GCT GTT 672
Gly Asp Leu Arg Asp He Gly Ala Gly Lys Gly Lys Tyr Tyr Ala Val
210 215 220 AAC TAC CCG CTC CGA GAC GGG ATT GAT GAC GAG TCC TAT GAG GCC ATT 720 Aβn Tyr Pro Leu Arg Asp Gly He Asp Asp Glu Ser Tyr Glu Ala He 225 230 235 240
TTC AAG CCG GTC ATG TCC AAA GTA ATG GAG ATG TTC CAG CCT AGT GCG 768 Phe Lys Pro Val Met Ser Lys Val Met Glu Met Phe Gin Pro Ser Ala
245 250 255
GTG GTC TTA CAG TGT GGC TCA GAC TCC CTA TCT GGG GAT CGG TTA GGT 816 Val Val Leu Gin Cys Gly Ser Asp Ser Leu Ser Gly Asp Arg Leu Gly 260 265 270
TGC TTC AAT CTA ACT ATC AAA GGA CAC GCC AAG TGT GTG GAA TTT GTC 864 Cys Phe Asn Leu Thr He Lys Gly His Ala Lys Cys Val Glu Phe Val 275 280 285
AAG AGC TTT AAC CTG CCT ATG CTG ATG CTG GGA GGC GGT GGT TAC ACC 912 Lys Ser Phe Aβn Leu Pro Met Leu Met Leu Gly Gly Gly Gly Tyr Thr 290 295 300 ATT CGT AAC GTT GCC CGG TGC TGG ACA TAT GAG ACA GCT GTG GCC CTG 960 He Arg Aβn Val Ala Arg Cys Trp Thr Tyr Glu Thr Ala Val Ala Leu 305 310 315 320
GAT ACG GAG ATC CCT AAT GAG CTT CCA TAC AAT GAC TAC TTT GAA TAC 1008 Asp Thr Glu He Pro Asn Glu Leu Pro Tyr Aβn Aβp Tyr Phe Glu Tyr
325 330 335
TTT GGA CCA GAT TTC AAG CTC CAC ATC AGT CCT TCC AAT ATG ACT AAC 1056 Phe Gly Pro Asp Phe Lys Leu His He Ser Pro Ser Asn Met Thr Asn 340 345 350
CAG AAC ACG AAT GAG TAC CTG GAG AAG ATC AAA CAG CGA CTG TTT GAG 1104 Gin Asn Thr Asn Glu Tyr Leu Glu Lys He Lys Gin Arg Leu Phe Glu 355 360 365
AAC CTT AGA ATG CTG CCG CAC GCA CCT GGG GTC CAA ATG CAG GCG ATT 1152 Aβn Leu Arg Met Leu Pro His Ala Pro Gly Val Gin Met Gin Ala He 370 375 380 CCT GAG GAC GCC ATC CCT GAG GAG AGT GGC GAT GAG GAC GAA GAC GAC 1200 Pro Glu Asp Ala He Pro Glu Glu Ser Gly Aβp Glu Asp Glu Asp Aβp 385 390 395 400
CCT GAC AAG CGC ATC TCG ATC TGC TCC TCT GAC AAA CGA ATT GCC TGT 1248 Pro Asp Lys Arg He Ser He Cys Ser Ser Asp Lys Arg He Ala Cys 405 410 415
GAG GAA GAG TTC TCC GAT TCT GAA GAG GAG GGA GAG GGG GGC CGC AAG 1296 Glu Glu Glu Phe Ser Asp Ser Glu Glu Glu Gly Glu Gly Gly Arg Lys 420 425 430
AAC TCT TCC AAC TTC AAA AAA GCC AAG AGA GTC AAA ACA GAG GAT GAA 1344
Aβn Ser Ser Aβn Phe Lye Lys Ala Lys Arg Val Lys Thr Glu Aβp Glu 435 440 445
AAA GAG AAA GAC CCA GAG GAG AAG AAA GAA GTC ACC GAA GAG GAG AAA 1392
Lye Glu Lye Asp Pro Glu Glu Lys Lys Glu Val Thr Glu Glu Glu Lys 450 455 460 ACC AAG GAG GAG AAG CCA GAA GCC AAA GGG GTC AAG GAG GAG GTC AAG 1440 Thr Lye Glu Glu Lys Pro Glu Ala Lys Gly Val Lys Glu Glu Val Lys 465 470 475 480
TTG GCC TGA 1449 Leu Ala
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 379 base pairβ
(B) TYPE: nucleic acid
(C) STRANDEDNESS: both (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
ATTGACTTCC TGCAGAGAGT CAGCCCCACC AATATGCAAG GCTTCACCAA GAGTCTTAAT 60 GCCTTCAACG TAGGCGATGA CTGCCCAGTG TTTCCCGGGC TCTTTGAGTT CTGCTCGCGT 120
TACACAGGCG CATCTCTGCA AGGAGCAACC CAGCTGAACA ACAAGATCTG TGATATTGCC 180
ATTAACTGGG CTGGTGGTCT GCACCATGCC TAGAAGTTTG AGGCCTCTGG CTTCTGCTAT 240
GTCAACGACA TTGTGTTTGG CATCCTGGAG CTGCTCAAGT ACCACCCTCG GGTGCTCTAC 300
ATTGACATTG ACATCCACCA TGGTGACGGG GTTCAAGAAG CTTTCTACCT CACTGACCGG 360 GTCATGACGG TGTCCTTTC 379
(2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 375 baβe pairβ (B) TYPE: nucleic acid
(C) STRANDEDNESS: both
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
TACTACTGTC TGAACGTGCC CCTGCGGATG GGCATTGATG ACCAGAGTTA CAAGCACCTT 60
TTCCAGCCGG TTATCAACCA GGTAGTGGAC TTCTACCAAC CCACGTGCAT TGTGCTCCAG 120 TGTGGAGCTG ACTCTCTGGG CTGTGATCGA TTGGGCTGCT TTAACCTCAG CATCCGAGGG 180
CATGGGGAAT GCGTTGAATA TGTCAAGAGC TTCAATATCC CTCTACTCGT GCTGGGTGGT 240
GGTGGTTATA CTGTCCGAAA TGTTGCCCGC TGCTGGACAT ATGAGACATC GCTGCTGGTA 300
GAAGAGGCCA TTAGTGAGGA GCTTCCCTAT AGTGAATACT TCGAGTACTT TGCCCCAGAC 360
TTCACACTTC ATCCA 375 (2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 227 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: both
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
GGTCATGCTA AATGTGTAGA AGTTGTAAAA ACTTTTAACT TACCATTACT GATGCTTGGA 60
GGAGGTGGCT ACACAATCCG TAATGTTGCT CGATGTTGGA CATATGAGAC TGCAGTTGCC 120
CTTGATTGTG AGATTCCCAA TGAGTTGCCA TATAATGATT ACTTTGAGTA TTTTGGACCA 180 GACTTCAAAC TGCATATTAG TCCTTCAAAC ATGACAAACC AGAACAC 227
(2) INFORMATION FOR SEQ ID NO:5: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 482 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:
Met Ala Gin Thr Gin Gly Thr Arg Arg Lys Val Cyβ Tyr Tyr Tyr Aβp 1 5 10 15
Gly Aβp Val Gly Aβn Tyr Tyr Tyr Gly Gin Gly His Pro Met Lys Pro 20 25 30 His Arg He Arg Met Thr His Asn Leu Leu Leu Aβn Tyr Gly Leu Tyr 35 40 45
Arg Lys Met Glu He Tyr Arg Pro His Lys Ala Asn Ala Glu Glu Met 50 55 60
Thr Lys Tyr His Ser Asp Asp Tyr He Lys Phe Leu Arg Ser He Arg 65 70 75 80
Pro Asp Asn Met Ser Glu Tyr Ser Lys Gin Met Gin Arg Phe Asn Val 85 90 95
Gly Glu Asp Cys Pro Val Phe Asp Gly Leu Phe Glu Phe Cys Gin Leu 100 105 110 Ser Thr Gly Gly Ser Val Ala Ser Ala Val Lys Leu Aβn Lye Gin Gin 115 120 125
Thr Asp He Ala Val Asn Trp Ala Gly Gly Leu His His Ala Lys Lys 130 135 140
Ser Glu Ala Ser Gly Phe Cyβ Tyr Val Aβn Aβp He Val Leu Ala He 145 150 155 160
Leu Glu Leu Leu Lys Tyr His Gin Arg Val Leu Tyr He Asp He Asp 165 170 175
He Hie His Gly Aβp Gly Val Glu Glu Ala Phe Tyr Thr Thr Asp Arg 180 185 190 Val Met Thr Val Ser Phe His Lys Tyr Gly Glu Tyr Phe Pro Gly Thr 195 200 205
Gly Asp Leu Arg Aβp He Gly Ala Gly Lya Gly Lye Tyr Tyr Ala Val 210 215 220
Aβn Tyr Pro Leu Arg Aβp Gly He Asp Aep Glu Ser Tyr Glu Ala He 225 230 235 240
Phe Lye Pro Val Met Ser Lye Val Met Glu Met Phe Gin Pro Ser Ala 245 250 255
Val Val Leu Gin Cyβ Gly Ser Aep Ser Leu Ser Gly Asp Arg Leu Gly 260 265 270 Cyβ Phe Aβn Leu Thr He Lys Gly His Ala Lys Cys Val Glu Phe Val 275 280 285
Lys Ser Phe Asn Leu Pro Met Leu Met Leu Gly Gly Gly Gly Tyr Thr 290 295 300
He Arg Asn Val Ala Arg Cys Trp Thr Tyr Glu Thr Ala Val Ala Leu 305 310 315 320
Asp Thr Glu He Pro Asn Glu Leu Pro Tyr Asn Asp Tyr Phe Glu Tyr 325 330 335
Phe Gly Pro Asp Phe Lys Leu His He Ser Pro Ser Asn Met Thr Asn 340 345 350 Gin Aβn Thr Asn Glu Tyr Leu Glu Lys He Lys Gin Arg Leu Phe Glu 355 360 365
Aβn Leu Arg Met Leu Pro His Ala Pro Gly Val Gin Met Gin Ala He 370 375 380
Pro Glu Asp Ala He Pro Glu Glu Ser Gly Asp Glu Asp Glu Asp Aβp 385 390 395 400
Pro Asp Lys Arg He Ser He Cys Ser Ser Asp Lys Arg He Ala Cyβ 405 410 415
Glu Glu Glu Phe Ser Aβp Ser Glu Glu Glu Gly Glu Gly Gly Arg Lye 420 425 430 Asn Ser ser Asn Phe Lys Lys Ala Lys Arg Val Lys Thr Glu Asp Glu 435 440 445
Lys Glu Lys Asp Pro Glu Glu Lys Lys Glu Val Thr Glu Glu Glu Lys 450 455 460
Thr Lye Glu Glu Lye Pro Glu Ala Lys Gly Val Lye Glu Glu Val Lys 465 470 475 480
Leu Ala
(2) INFORMATION FOR SEQ ID NO:6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 133 amino acids (B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:6: He Asp Phe Leu Gin Arg Val Ser Pro Thr Asn Met Gin Gly Phe Thr
1 5 10 15
Lys Ser Leu Asn Ala Phe Asn Val Gly Asp Asp Cyβ Pro Val Phe Pro 20 25 30
Gly Leu Phe Glu Phe Cys Ser Arg Tyr Thr Gly Ala Ser Leu Gin Gly 35 40 45 Ala Thr Gin Leu Asn Aβn Lye He Cyβ Asp He Ala He Asn Trp Ala 50 55 60
Gly Gly Leu His His Ala Lys Lys Phe Glu Ala Ser Gly Phe Cys Tyr 65 70 75 80
Val Asn Asp He Val Phe Gly He Leu Glu Leu Leu Lys Tyr Hie Pro 85 90 95
Arg Val Leu Tyr He Aβp He Aβp He Hie His Gly Aβp Gly Val Gin 100 105 110
Glu Ala Phe Tyr Leu Thr Aβp Arg Val Met Thr Val Ser Phe Pro Gin 115 120 125 He Arg Glu He Tyr 130
(2) INFORMATION FOR SEQ ID NO:7: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 125 amino acidβ
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:
Tyr Tyr Cys Leu Asn Val Pro Leu Arg Met Gly He Asp Asp Gin Ser 1 5 10 15
Tyr Lys His Leu Phe Gin Pro Val He Asn Gin Val Val Asp Phe Tyr 20 25 30
Gin Pro Thr Cys He Val Leu Gin Cys Gly Ala Aβp Ser Leu Gly Cys 35 40 45
Asp Arg Leu Gly Cys Phe Aβn Leu Ser He Arg Gly His Gly Glu Cys
50 55 60
Val Glu Tyr Val Lys Ser Phe Asn He Pro Leu Leu Val Leu Gly Gly 65 70 75 80
Gly Gly Tyr Thr Val Arg Aβn Val Ala Arg Cys Trp Thr Tyr Glu Thr 85 90 95
Ser Leu Leu Val Glu Glu Ala He Ser Glu Glu Leu Pro Tyr Ser Glu 100 105 110
Tyr Phe Glu Tyr Phe Ala Pro Aβp Phe Thr Leu His Pro 115 120 125
(2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 80 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:8:
Asn Leu Leu Val Leu Gly His Ala Lys Cyβ Val Glu Val Val Lye Thr 1 5 10 15 Phe Aβn Leu Pro Leu Leu Met Leu Gly Gly Gly Gly Tyr Thr He Arg
20 25 30
Aβn Val Ala Arg Cyβ Trp Thr Tyr Glu Thr Ala Val Ala Leu Aβp Cyβ 35 40 45
Glu He Pro Aβn Glu Leu Pro Tyr Aβn Aβp Tyr Phe Glu Tyr Phe Gly 50 55 60
Pro Aβp Phe Lye Leu His He Ser Pro Ser Aβn Met Thr Aβn Gin Aβn 65 70 75 80
(2) INFORMATION FOR SEQ ID NO:11: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1275 baβe pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: both
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(ix) FEATURE: (A) NAME/KEY: CDS ( B ) LOCATION : 1. . 1275
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:11:
ATG GCC GAC AAG GAA GCA GCC TTC GAC GAC GCA GTG GAA GAA CGA GTG 48 Met Ala Asp Lys Glu Ala Ala Phe Asp Asp Ala Val Glu Glu Arg Val 1 5 10 15 ATC AAC GAG GAA TAC AAA ATA TGG AAA AAG AAC ACC CCT TTT CTT TAT 96 He Aβn Glu Glu Tyr Lye He Trp Lye Lye Aβn Thr Pro Phe Leu Tyr 20 25 30
GAT TTG GTG ATG ACC CAT GCT CTG GAG TGG CCC AGC CTA ACT GCC CAG 144 Asp Leu Val Met Thr His Ala Leu Glu Trp Pro Ser Leu Thr Ala Gin 35 40 45
TGG CTT CCA GAT GTA ACC AGA CCA GAA GGG AAA GAT TTC AGC ATT CAT 192 Trp Leu Pro Asp Val Thr Arg Pro Glu Gly Lye Aβp Phe Ser He Hie 50 55 60
CGA CTT GTC CTG GGG ACA CAC ACA TCG GAT GAA CAA AAC CAT CTT GTT 240 Arg Leu Val Leu Gly Thr His Thr Ser Asp Glu Gin Aβn Hie Leu Val 65 70 75 80
ATA GCC AGT GTG CAG CTC CCT AAT GAT GAT GCT CAG TTT GAT GCG TCA 288 He Ala Ser Val Gin Leu Pro Aβn Aβp Aβp Ala Gin Phe Aβp Ala Ser 85 90 95 CAC TAC GAC AGT GAG AAA GGA GAA TTT GGA GGT TTT GGT TCA GTT AGT 336 Hie Tyr Aβp Ser Glu Lye Gly Glu Phe Gly Gly Phe Gly Ser Val Ser 100 105 110
GGA AAA ATT GAA ATA GAA ATC AAG ATC AAC CAT GAA GGA GAA GTA AAC 384 Gly Lye He Glu He Glu He Lys He Aβn Hie Glu Gly Glu Val Aβn 115 120 125
AGG GCC CGT TAT ATG CCC CAG AAC CCT TGT ATC ATC GCA ACA AAG ACT 432 Arg Ala Arg Tyr Met Pro Gin Asn Pro Cys He He Ala Thr Lys Thr 130 135 140
CCT TCC AGT GAT GTT CTT GTC TTT GAC TAT ACA AAA CAT CCT TCT AAA 480 Pro Ser Ser Asp Val Leu Val Phe Asp Tyr Thr Lys His Pro Ser Lys 145 150 155 160
CCA GAT CCT TCT GGA GAG TGC AAC CCA GAC TTG CGT CTC CGT GGA CAT 528 Pro Asp Pro Ser Gly Glu Cys Asn Pro Asp Leu Arg Leu Arg Gly His 165 170 175 CAG AAG GAA GGC TAT GGG CTT TCT TGG AAC CCA AAT CTC AGT GGG CAC 576 Gin Lys Glu Gly Tyr Gly Leu Ser Trp Aβn Pro Asn Leu Ser Gly His 180 185 190
TTA CTT AGT GCT TCA GAT GAC CAT ACC ATC TGC CTG TGG GAC ATC AGT 624 Leu Leu Ser Ala Ser Asp Asp His Thr He Cys Leu Trp Asp He Ser 195 200 205
GCC GTT CCA AAG GAG GGA AAA GTG GTA GAT GCG AAG ACC ATC TTT ACA 672 Ala Val Pro Lye Glu Gly Lys Val Val Asp Ala Lys Thr He Phe Thr 210 215 220
GGG CAT ACG GCA GTA GTA GAA GAT GTT TCC TGG CAT CTA CTC CAT GAG 720 Gly His Thr Ala Val Val Glu Asp Val Ser Trp His Leu Leu His Glu 225 230 235 240
TCT CTG TTT GGG TCA GTT GCT GAT GAT CAG AAA CTT ATG ATT TGG GAT 768
Ser Leu Phe Gly Ser Val Ala Aβp Aβp Gin Lys Leu Met He Trp Aβp 245 250 255 ACT CGT TCA AAC AAT ACT TCC AAA CCA AGC CAC TCA GTT GAT GCT CAC 816
Thr Arg Ser Asn Asn Thr Ser Lys Pro Ser His Ser Val Aβp Ala His
260 265 270
ACT GCT GAA GTG AAC TGC CTT TCT TTC AAT CCT TAT AGT GAG TTC ATT 864 Thr Ala Glu Val Asn Cyβ Leu Ser Phe Aβn Pro Tyr Ser Glu Phe He 275 280 285
CTT GCC ACA GGA TCA GCT GAC AAG ACT GTT GCC TTG TGG GAT CTG AGA 912
Leu Ala Thr Gly Ser Ala Asp Lys Thr Val Ala Leu Trp Aβp Leu Arg 290 295 300
AAT CTG AAA CTT AAG TTG CAT TCC TTT GAG TCA CAT AAG GAT GAA ATA 960
Asn Leu Lys Leu Lys Leu His Ser Phe Glu Ser His Lys Aβp Glu He 305 310 315 320
TTC CAG GTT CAG TGG TCA CCT CAC AAT GAG ACT ATT TTA GCT TCC AGT 1008
Phe Gin Val Gin Trp Ser Pro His Asn Glu Thr He Leu Ala Ser Ser 325 330 335 GGT ACT GAT CGC AGA CTG AAT GTC TGG GAT TTA AGT AAA ATT GGA GAG 1056
Gly Thr Asp Arg Arg Leu Aβn Val Trp Asp Leu Ser Lys He Gly Glu
340 345 350
GAA CAA TCC CCA GAA GAT GCA GAA GAC GGG CCA CCA GAG TTG TTG TTT 1104 Glu Gin Ser Pro Glu Asp Ala Glu Asp Gly Pro Pro Glu Leu Leu Phe 355 360 365
ATT CAT GGT GGT CAT ACT GCC AAG ATA TCT GAT TTC TCC TGG AAT CCC 1152
He Hie Gly Gly His Thr Ala Lys He Ser Asp Phe Ser Trp Asn Pro 370 375 380
AAT GAA CCT TGG GTG ATT TGT TCT GTA TCA GAA GAC AAT ATC ATG CAA 1200
Asn Glu Pro Trp Val He Cys Ser Val Ser Glu Aβp Asn He Met Gin 385 390 395 400
GTG TGG CAA ATG GCA GAG AAC ATT TAT AAT GAT GAA GAC CCT GAA GGA 1248
Val Trp Gin Met Ala Glu Asn He Tyr Asn Asp Glu Asp Pro Glu Gly 405 410 415 AGC GTG GAT CCA GAA GGA CAA GGG TCC TAG 1278 Ser Val Asp Pro Glu Gly Gin Gly Ser 420 425
(2) INFORMATION FOR SEQ ID NO:12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 69 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:12:
Asp Xaa Xaa Xaa Asn Xaa Xaa Gly Gly Leu His His Ala Lys Lys Xaa 1 5 10 15
Glu Ala Ser Gly Phe Cyβ Tyr Xaa Aβn Asp He Val Xaa Xaa He Xaa 20 25 30
Glu Leu Leu Xaa Tyr His Xaa Arg Val Xaa Tyr He Aβp Xaa Aβp Xaa 35 40 45
Hie Hie Gly Aβp Gly Xaa Glu Glu Ala Phe Tyr Xaa Thr Aβp Arg Val 50 55 60 Met Thr Xaa Ser Phe 65
(2) INFORMATION FOR SEQ ID NO:13: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 69 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide
(v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:13:
Aep He Ala Xaa Aβn Trp Ala Gly Gly Leu Hie Hie Ala Lye Lys Xaa 1 5 10 15 Glu Ala Ser Gly Phe Cys Tyr Val Asn Aβp He Val Xaa Xaa He Leu
20 25 30
Glu Leu Leu Lys Tyr His Xaa Arg Val Leu Tyr He Asp He Asp He 35 40 45 Hiβ His Gly Asp Gly Xaa Glu Glu Ala Phe Tyr Xaa Thr Asp Arg Val 50 55 60
Met Thr Val Ser Phe 65
(2) INFORMATION FOR SEQ ID NO:14:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 33 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:14:
Cys Val Xaa Xaa Xaa Lys Xaa Phe Xaa Xaa Pro Xaa Xaa Xaa Xaa Gly
1 5 10 15
Gly Gly Gly Tyr Thr Xaa Arg Asn Val Ala Arg Xaa Trp Xaa Xaa Glu 20 25 30
Thr
(2) INFORMATION FOR SEQ ID NO:15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(v) FRAGMENT TYPE: internal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:15:
Cyβ Val Glu Xaa Val Lye Xaa Phe Aβn Xaa Pro Leu Leu Xaa Leu Gly 1 5 10 15
Gly Gly Gly Tyr Thr Xaa Arg Aβn Val Ala Arg Cyβ Trp Thr Tyr Glu 20 25 30 Thr

Claims

What is claimed is:
1. An isolated or recombinant HDx polypeptide.
2. The polypeptide of claim 1, of mammalian origin.
3. The polypeptide of claim 3, of human origin. 4. The polypeptide of claim 1, which polypeptide comprises an HDx polypeptide sequence at least 88 percent homologous with SEQ ID No: 2, or fragment thereof.
5. The polypeptide of claim 1, which polypeptide comprises an HDx polypeptide sequence at least 95 percent homologous with SEQ DD No: 2, or fragment thereof.
6. The polypeptide of claim 1, which polypeptide comprises an HDx polypeptide sequence designated in SEQ DD No: 2.
7. The polypeptide of claim 1, which polypeptide is encoded by a nucleic acid having a coding sequence, or portion thereof, which hybridizes under stringent conditions to the nucleic acid designated in SEQ DD No. 1.
9. The polypeptide of claim lr which polypeptide is an acetylase activity. 10. The polypeptide of claim 1, which polypeptide binds to an RbAp48 protein.
11. The polypeptide of claim 1, which polypeptide is a fusion protein.
12. The polypeptide of claim 1, which polypeptide has a molecular weight in the range of 45-70 Kd.
13. An isolated or recombinant polypeptide comprising an HDx polypeptide sequence homologous or identical SEQ ID No. 2, or a fragment thereof which retains one or more of (i) a histone deacetylase activity, (ii) a histone binding activity and (ϋi) an RbAp48 binding activity.
14. The polypeptide of claim 13, which polypeptide comprises an HDx sequence represented in the general formula DXXNXGGLΗΗAKKXEASGFCYXNDIVXXI-
XELLXYΗXRVXYTOXDXΗΗGDGXEAFYXTDRVMTXSF.
15. The polypeptide of claim 13, which polypeptide comprises an HDx sequence represented in the general formula C\ C XKXFXXPXXXXGGGGYTXRNVARX-WXXET. 16. The polypeptide of claim 13, which polypeptide deacetylates acetylated histones. 17. The polypeptide of claim 13, which polypeptide is a dominant negative inhibitor which antagonizes deacetylation of acetylated histones.
18. The polypeptide of claim 13, which polypeptide comprises an HDx sequence at least 88 percent homologous with SEQ ID No: 2, or fragment thereof. 19. The polypeptide of claim 13, which polypeptide comprises an HDx sequence at least 95 percent homologous with SEQ ID No: 2, or fragment thereof.
20. The polypeptide of claim 13, which polypeptide includes at least 25 amino acid residues of an HDx polypeptide sequence.
21. The polypeptide of claim 13, wherein said polypeptide modulates cellular proliferation.
22. An isolated or recombinant polypeptide comprising an HDx polypeptide sequence represented SEQ DD No. 2, or a fragment thereof which retains one or more of (i) a histone deacetylase activity, (ii) a histone binding activity and (iii) an RbAp48 binding activity. 23. The polypeptide of claim 22, wherein said fusion protem includes, as a second polypeptide sequence, a polypeptide which functions as a detectable label for detecting the presence of said fusion protein or as a matrix-binding domain for immobilizing said fusion protein.
24. The polypeptide of claim 13, wherein said polypeptide is a fusion protein further comprising, in addition to said HDx sequence, a second polypeptide sequence havmg an amino acid sequence unrelated to an HDx polypeptide sequence.
25. A purified or recombinant HDx polypeptide encoded by a nucleic acid which hybridizes under stringent conditions to a nucleotide sequence designated in SEQ ID No. 1. 26. A purified or recombinant HDx polypeptide comprising a v motif represented in the general formula
DIAX1NWAGGLΗΗAKKX2EASGFCYVNDIVX3X4D.ELLKYΗ- X5RVLYDDIDIHHGDGX6EAFYX7TDRVMTVSF and a χ motif represented in CVEXiV^FjN^P-X^XsLGGGGYTXgRNVARCWTYET. 27. An isolated nucleic acid which encodes a deacetylase activity and hybridizes under stringent conditions to a nucleotide sequence designated in SEQ ID No. 1. 28. An isolated nucleic acid encoding an HDx polypeptide, which polypeptide specifically modulates histone acetylation.
29. The nucleic acid of claim 28, which HDx polypeptide comprises a v motif represented in the general formula DIAXJNWAGGLΗΗAKKX2EASGFCYVNDIVX3X4ILELL-
KYHX5RVLYIDΠDIHHGDGX6EAFYX7TDRVMTVSF and a χ motif represented in CVEX1VKX2F X3P-X4LX5LGGGGYTX6RJ>ΓV'ARCWTYET.
30. The nucleic acid of claim 28, which HDx polypeptide comprises a polypeptide sequence at least 88 percent homologous with SEQ DD No: 2, or fragment thereof. 31. The nucleic acid of claim 28, which HDx polypeptide comprises a polypeptide sequence at least 95 percent homologous with SEQ UD No: 2, or fragment thereof.
32. The nucleic acid of claim 28, which HDx polypeptide comprises a polypeptide sequence designated in SEQ YD No: 2.
33. The nucleic acid of claim 28, which HDx polypeptide has a molecular weight in the range of 45-70 Kd.
34. The nucleic acid of claim 28, which HDx polypeptide is a fusion protein further comprising, in addition to HDx polypeptide sequences, a second polypeptide sequence having an amino acid sequence unrelated to a nucleic acid sequence.
35. The nucleic acid of claim 34, wherein said fusion protein includes, as a second polypeptide sequence, a polypeptide which functions as a detectable label for detecting the presence of said fusion protein or as a matrix-binding domain for immobilizing said fusion protein.
36. The nucleic acid of claim 28, further comprising a transcriptional regulatory sequence operably linked to said nucleotide sequence so as to render said nucleic acid suitable for use as an expression vector.
37. An expression vector, capable of replicating in at least one of a prokaryotic cell and eukaryotic cell, comprising the nucleic acid of claim 36.
38. A host cell transfected with the expression vector of claim 37 and expressing said recombinant polypeptide. 39. A method of producing a recombinant HDx polypeptide comprising culturing die cell of claim 38 in a cell culture medium to express said recombinant polypeptide and isolating said recombinant polypeptide from said cell culture. 40. A transgenic non-human animal having cells which harbor a heterologous transgene encoding an HDx polypeptide.
41. A transgenic non-human animal having cells in which an HDx gene is dismpted.
42. A recombinant transfection system, comprising (i) a gene constmct including the nucleic acid of claim 28 and operably linked to a transcriptional regulatory sequence for causing expression of said HDx polypeptide in eukaryotic cells, and
(ii) a gene delivery composition for delivering said gene constmct to a cell and causing the cell to be transfected with said gene constmct. 43. The recombinant transfection system of claim 42, wherein the gene delivery composition is selected from a group consisting of a recombinant viral particle, a liposome, and a poly-cationic nucleic acid binding agent.
44. A nucleic acid composition comprising a substantially purified oligonucleotide, said oligonucleotide including a region of nucleotide sequence which hybridizes under stringent conditions to at least 25 consecutive nucleotides of sense or antisense sequence of an HDx gene.
45. The nucleic acid composition of claim 44, which oligonucleotide hybridizes under stringent conditions to at least 50 consecutive nucleotides of sense or antisense sequenc of an HDx gene. 46. The nucleic acid composition of claim 44, wherein said oligonucleotide further comprises a label group attached thereto and able to be detected.
47. The nucleic acid composition of claim 44, wherein said oligonucleotide has at least one non-hydrolyzable bond between two adjacent nucleotide subunits.
48. A test kit for detecting cells which contain an HDx-encoding nucleic acid, comprising the nucleic acid composition of claim 44 for measuring, in a sample of cells, a level of nucleic acid encoding an HDx protein.
49. A method for modulating one or more of growth, differentiation, or survival of a mammalian cell responsive to HDx-mediated histone deacetylation, comprising treating the cell with an effective amount of an agent which modulates the deacetylase activity of an HDx polypeptide thereby altering, relative to the cell in the absence of the agent, at least one of (i) rate of growth, (ii) differentiation, or (iii) survival of the cell. 50. An antibody to an HDx polypeptide.
51. The antibody of claim 50, wherein said antibody is monoclonal.
52. A diagnostic assay for identifying a cell or cells at risk for a disorder characterized by unwanted cell proliferation or differentiation, comprising detecting, in a cell sample, the presence or absence of a genetic lesion characterized by at least one of
(i) aberrant modification or mutation of a gene encoding an HDx protein, and (ii) mis-expression of said gene; wherein a wild-type form of said gene encodes an HDx protein characterized by an ability to modulate the signal transduction activity of a TGFβ receptor. 53. The assay of claim 52, wherein detecting said lesion includes: i. providing a diagonistic probe comprising a nucleic acid including a region of nucleotide sequence which hybridizes to a sense or antisense sequence of said gene, or naturally occuring mutants thereof, or 5* or 3' flanking sequences naturally associated with said gene; ii. combining said probe with nucleic acid of said cell sample; and iii. detecting, by hybridization of said probe to said cellular nucleic acid, the existence of at least one of a deletion of one or more nucleotides from said gene, an addition of one or more nucleotides to said gene, a substitution of one or more nucleotides of said gene, a gross chromosomal rearrangement of all or a portion of said gene, a gross alteration in the level of an mRNA transcript of said gene, or a non-wild type splicing pattern of an mRNA transcript of said gene.
54. The assay of claim 53, wherein hybridization of said probe further comprises subjecting the probe and cellular nucleic acid to a polymerase chain reaction (PCR) and detecting abnormalities in an amplified product.
55. The assay of claim 53, wherein hybridization of said probe further comprises subjecting the probe and cellular nucleic acid to a ligation chain reaction (LCR) and detecting abnormalities in an amplified product.
56. The assay of claim 53, wherein said probe hybridizes under stringent conditions to the nucleic acid designated by SEQ DD No. 1.
57. An assay for screening test compounds to identify agents which inhibit the deacetylation of histones comprising: i. providing a reaction mixture including a histone deacetylase activity of an HDx-like polypeptide, a substrate for a histone deacetylase, and a test compound; and ii. detecting the conversion of the substrate to product, wherein a statistically signficant decrease in the conversion of the substrate m the presence of the test compound is indicative of a potential inhibitor of histone deacetylation.
58. The assay of claim 57, wherein the HDx-like polypeptide is of mammalian origin.
59. The assay of claim 57, wherein the HDx-like polypeptide is an RPD3-like deacetylase of fungal origin.
60. The assay of claim 57, wherein the reaction mixture is a reconstituted protein mixture.
61. The assay of claim 57, wherein said reaction mixture is a cell lysate.
62. The assay of claim 57, wherein the HDx-like polypeptide is a recombinant protein. 63. An assay for screening test compounds to identify agents which inhibit histone deacetylase interaction with cellular proteins, comprising: i. providing a reaction mixture including an HDx-like protein, an HDx- binding protein, and a test compound; and ii. detecting the interaction of the HDx-like protein and the HDx binding protein, wherein a statistically signficant decrease in the interaction of the proteins in the presence of the test compound is indicative of a potential inhibitor of a histone deacetylase.
64. The assay of claim 63, wherein the HDx-like protein is of mammalian origin. 65. The assay of claim 63, wherein the HDx-like polypeptide is an RPD3-ϋke deacetylase of fungal origin.
66. The assay of claim 63, wherein the HDx-like protein is a histone, or a portion thereof which interacts with an HDx-like polypeptide.
67. The assay of claim 63, wherein the HDx-like protein is an PbAp48 protein, or a portion thereof which interacts with an HDx-like polypeptide.
68. The assay of claim 63, wherein the reaction mixture is a reconstituted protein mixture. 69. The assay of claim 63, wherein said reaction mixture is a cell lysate.
70. The assay of claim 63, wherein the HDx-like polypeptide is a recombinant protein.
71. The assay of claim 63, wherein one or both of the HDx-like protein and HDx- binding protein is a fusion protein. 72. The assay of claim 63, wherein at least one of the HDx-like protein and HDx- binding protein comprises an endogenous detectable label for detecting the formation of said complex.
73. The method of claim 63, which reaction mixture is a whole cell, and interaction of the HDx-like protein and HDx-binding protein is detected in a two hybrid assay syste .
74. A composition for inhibiting a histone deacetylase comprismg a compound represented by the general formula A-B-C, wherein
A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclopeptides; B is selected from the group consisting of substituted and unsubstituted
C -C8 alkylidenes, C4-C alkenylidenes, C4-C8 alkynylidenes, and -(D-E-F)-, in which D and F are, independently, absent or represent a C2-C7 alkylidene, a C2- C7 alkenylidene or a C2-C7 alkynylidene, and E represents O, S, or NR', in which R' represents Η, a lower alkyl, a lower alkenyl, a lower alkynyl, an aralkyl, aryl, or a heterocyclyl; and
C is selected from the group consisting of
Figure imgf000132_0001
, and a boronic acid; in which Z represents O, S, or NR5, and Y; R5 represents a hydrogen, an alkyl, an alkoxycarbonyl, an aryloxycarbonyl, an alkylsulfonyl, an arylsulfonyl or an aryl; R'6 represents hydrogen, an alkyl, an alkenyl, an alkynyl or an aryl; and R7 represents a hydrogen, an alkyl, an aryl, an alkoxy, an aryloxy, an ammo, a hydroxylamino, an alkoxylamino or a halogen; with the proviso that the compound is not trapoxin. 75. A pharmaceutical preparation comprising (i) the composition of claim 74 in an amount effective for inhibiting proliferation of a cell, and (ii) a pharmaceutically acceptable diluent.
76. A method for modulating one or more of growth, differentiation, or survival of a mammalian cell responsive to HDx-mediated histone deacetylation, comprismg treating the cell with an effective amount of the compisition of claim 74 so as to modulate the deacetylase activity and alter, relative to the cell in the absence of the agent, at least one of (i) the rate of growth, (ii) the differentiation state, or (iii) the rate of survival of the cell.
77. A composition for inhibiting a histone deacetylase comprising a compound represented by the general formula A-B-C, wherein
A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclopeptides;
B is selected from the group consisting of substituted and unsubstituted C4-C8 alkylidenes, C4-C alkenylidenes, C4-C8 alkynylidenes, and -(D-E-F)-, in which D and F are, independently, absent or represent C2-C alkylidenes, C2-C7 alkenylidenes or C2-C7 alkynylidenes, and E represents O, S, or NR', in which R' represents Η, a lower alkyl, a lower alkenyl, a lower alkynyl, an aralkyl, an aryl, or a heterocyclyl; and
C is selected from the group consisting of
Figure imgf000133_0001
, , in which R9 represents a hydrogen, an alkyl, an aryl, a hydroxyl, an alkoxy, an aryloxy or an amino, with the proviso that the inhibitor compound is not trichostatin.
78. A pharmaceutical preparation comprising (i) the composition of claim 77 in an amount effective for inhibiting proliferation of a cell, and (ii) a pharmaceutically acceptable diluent. 79. A method for modulating one or more of growth, differentiation, or survival of a mammalian cell responsive to HDx-mediated histone deacetylation, comprismg treating the cell with an effective amount of the compisition of claim 77 so as to modulate the deacetylase activity and alter, relative to the cell in the absence of the agent, at least one of (i) the rate of growth, (ii) the differentiation state, or (ϋi) die rate of survival of the cell.
80. A composition for inhibiting a histone deacetylase comprising a compound represented by the general formula A-B-C, wherein A is selected from the group consisting of cycloalkyls, unsubstituted and substituted aryls, heterocyclyls, amino acyls, and cyclopeptides;
B is selected from the group consisting of substituted and unsubstituted
C -C8 alkylidenes, C4-C8 alkenylidenes, C4-C8 alkynylidenes, and -(D-E-F)-, in which D and F are, independently, absent or a C2-C7 alkylidene, a C2-C7 alkenylidene, or a C2-C7 alkynylidene, and E represents O, S, or NR', in which R' is Η, lower alkyl, lower alkenyl, lower alkynyl, aralkyl, aryl, or heterocyclyl; and
C represents
Figure imgf000134_0001
; in which Y is O or S, and R7 represents a hydrogen, an alkyl, an aryl, an alkoxy, an aryloxy, an amino, a hydroxylamino, an alkoxylamino or a halogen.
81. A pharmaceutical preparation comprising (i) the composition of claim 80 in an amount effective for inhibiting proliferation of a cell, and (ii) a pharmaceutically acceptable diluent.
82. A method for modulating one or more of growth, differentiation, or survival of a mammalian cell responsive to HDx-mediated histone deacetylation, comprising treating the cell with an effective amount of the compisition of claim 80 so as to modulate the deacetylase activity and alter, relative to the cell in the absence of the agent, at least one of (i) the rate of growth, (ii) the differentiation state, or (iii) the rate of survival of the cell .
PCT/US1997/005275 1996-03-26 1997-03-26 Histone deacetylases, and uses related thereto WO1997035990A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU29905/97A AU2990597A (en) 1996-03-26 1997-03-26 Histone deacetylases, and uses related thereto

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/624,735 US6777217B1 (en) 1996-03-26 1996-03-26 Histone deacetylases, and uses related thereto
US08/624,735 1996-03-26

Publications (3)

Publication Number Publication Date
WO1997035990A2 true WO1997035990A2 (en) 1997-10-02
WO1997035990A3 WO1997035990A3 (en) 1998-03-26
WO1997035990A9 WO1997035990A9 (en) 1998-07-16

Family

ID=24503127

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/005275 WO1997035990A2 (en) 1996-03-26 1997-03-26 Histone deacetylases, and uses related thereto

Country Status (3)

Country Link
US (8) US6777217B1 (en)
AU (1) AU2990597A (en)
WO (1) WO1997035990A2 (en)

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999023885A1 (en) * 1997-11-10 1999-05-20 The Salk Institute For Biological Studies Methods for the use of inhibitors of co-repressors for the treatment of neoplastic diseases
WO1999060407A1 (en) * 1998-05-15 1999-11-25 Cancer Research Campaign Technogoly Limited Assays, methods and means for modulating e2f activity
WO2000008048A2 (en) * 1998-08-04 2000-02-17 Fujisawa Pharmaceutical Co., Ltd. Inhibitor of histone deacetylase
WO2000021979A2 (en) * 1998-10-13 2000-04-20 Fujisawa Pharmaceutical Co., Ltd. Cyclic tetrapeptide and their use as histone deacetylase inhibitor
EP1010705A1 (en) * 1997-09-02 2000-06-21 Japan Energy Corporation Novel cyclic tetrapeptide derivatives and medicinal use thereof
WO2000071703A2 (en) * 1999-05-03 2000-11-30 Methylgene Inc. Inhibition of histone deacetylase
EP1105143A1 (en) * 1998-08-21 2001-06-13 SmithKline Beecham Corporation Human histone deacetylase gene hd4
WO2001067107A1 (en) * 2000-03-04 2001-09-13 Imperial College Innovations Limited Modulation of histone deacetylase
EP1170008A1 (en) * 2000-07-07 2002-01-09 Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus Valproic acid and derivatives thereof as histone deacetylase inhibitors
WO2002030970A2 (en) * 2000-10-13 2002-04-18 Bayer Aktiengesellschaft Human histone deacetylase gene
US6403555B1 (en) 1999-12-08 2002-06-11 Xcyte Therapies, Inc. Depsipeptide and congeners thereof for use as immunosuppressants
WO2002069947A2 (en) * 2001-01-12 2002-09-12 Methylgene, Inc. Antisense oligonucleotides and trichostatin analogue histone deacetylase-4 inhibitors against cancer
EP1243290A2 (en) * 1998-10-19 2002-09-25 Methylgene, Inc. Modulation of gene expression by combination therapy
WO2003006652A2 (en) * 2000-03-24 2003-01-23 Methylgene, Inc. Inhibition of specific histone deacetylase isoforms
WO2003013493A1 (en) * 2001-08-07 2003-02-20 Italfarmaco S.P.A. Histone deacetylase enzyme-inhibiting derivatives of hydroxamic acid as new cytokine synthesis-inhibiting anti-inflammatory drugs
US6552065B2 (en) 2000-09-01 2003-04-22 Novartis Ag Deacetylase inhibitors
WO2003032921A2 (en) 2001-10-16 2003-04-24 Sloan-Kettering Institute For Cancer Research Treatment of neurodegenerative diseases and cancer of the brain
EP1390491A1 (en) * 2001-05-02 2004-02-25 The Regents of the University of California Method for treating neurodegenerative, psychiatric and other disorders with deacetylase inhibitors
US6706762B1 (en) 1997-05-01 2004-03-16 The Salk Institute For Biological Studies Methods for the use of inhibitors of co-repressors for the treatment of neoplastic diseases
EP1400806A1 (en) * 2002-09-18 2004-03-24 G2M Cancer Drugs AG The use of molecular markers for the preclinical and clinical profiling of inhibitors of enzymes having histone deacetylase activity
US6808926B1 (en) 1999-08-27 2004-10-26 Her Majesty The Queen In Right Of Canada, As Represented By The Minister Of Agriculture And Agri-Food Repressing gene expression in plants
US6828302B1 (en) 1999-12-08 2004-12-07 Xcyte Therapies, Inc. Therapeutic uses of depsipeptides and congeners thereof
US6953783B1 (en) 1998-10-19 2005-10-11 Methylgene, Inc. Modulation of gene expression by combination therapy
US7057057B2 (en) 2002-05-22 2006-06-06 Errant Gene Therapeutics, Llc Histone deacetylase inhibitors based on alpha-ketoepoxide compounds
US7063973B2 (en) 2001-06-14 2006-06-20 Sloan-Kettering Institute For Cancer Research HDAC9 polypeptides and polynucleotides and uses thereof
US7244853B2 (en) 2001-05-09 2007-07-17 President And Fellows Of Harvard College Dioxanes and uses thereof
US7250504B2 (en) 2000-03-03 2007-07-31 President And Fellows Of Harvard College Class II human histone deacetylases, and uses related thereto
US7271198B2 (en) 2000-11-21 2007-09-18 Wake Forest University Method of treating autoimmune diseases
US7553667B2 (en) 2002-06-06 2009-06-30 Her Majesty The Queen In Right Of Canada As Represented By The Minister Of Agriculture And Agri-Food Regulation of gene expression using chromatin remodelling factors
US7566535B2 (en) 2002-03-07 2009-07-28 University Of Delaware Enhanced oligonucleotide-mediated nucleic acid sequence alteration
US7582807B2 (en) 2002-06-06 2009-09-01 Her Majesty The Queen In Right Of Canada, As Represented By The Minister Of Agriculture And Agri-Food Regulation of gene expression using chromatin remodelling factors
US7732475B2 (en) 2005-07-14 2010-06-08 Takeda San Diego, Inc. Histone deacetylase inhibitors
US8222423B2 (en) 2006-02-14 2012-07-17 Dana-Farber Cancer Institute, Inc. Bifunctional histone deacetylase inhibitors
US8304451B2 (en) 2006-05-03 2012-11-06 President And Fellows Of Harvard College Histone deacetylase and tubulin deacetylase inhibitors
US8329946B2 (en) 1996-03-26 2012-12-11 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
CN102858779A (en) * 2010-02-19 2013-01-02 派普塔德姆公司 Cyclic tetrapeptides and therapeutic applications thereof
US8383855B2 (en) 2006-02-14 2013-02-26 President And Fellows Of Harvard College Histone deacetylase inhibitors
US8440716B2 (en) 2008-07-23 2013-05-14 President And Fellows Of Harvard College Deacetylase inhibitors and uses thereof
US8691534B2 (en) 2006-12-29 2014-04-08 Celgene Corporation Preparation of romidepsin
US8716344B2 (en) 2009-08-11 2014-05-06 President And Fellows Of Harvard College Class- and isoform-specific HDAC inhibitors and uses thereof
US8999289B2 (en) 2005-03-22 2015-04-07 President And Fellows Of Harvard College Treatment of protein degradation disorders
US9908908B2 (en) 2012-08-30 2018-03-06 Jiangsu Hansoh Pharmaceutical Co., Ltd. Tenofovir prodrug and pharmaceutical uses thereof
EP3461488A1 (en) 2017-09-27 2019-04-03 Onxeo Combination of a dbait molecule and a hdac inhibitor for treating cancer
EP3461480A1 (en) 2017-09-27 2019-04-03 Onxeo Combination of a dna damage response cell cycle checkpoint inhibitors and belinostat for treating cancer
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
WO2023041805A1 (en) 2021-09-20 2023-03-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for improving the efficacy of hdac inhibitor therapy and predicting the response to treatment with hdac inhibitor
WO2023194441A1 (en) 2022-04-05 2023-10-12 Istituto Nazionale Tumori Irccs - Fondazione G. Pascale Combination of hdac inhibitors and statins for use in the treatment of pancreatic cancer

Families Citing this family (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2465978C (en) 2001-09-14 2015-04-07 Soon Hyung Woo Inhibitors of histone deacetylase
US7868204B2 (en) * 2001-09-14 2011-01-11 Methylgene Inc. Inhibitors of histone deacetylase
US6897220B2 (en) 2001-09-14 2005-05-24 Methylgene, Inc. Inhibitors of histone deacetylase
US7154002B1 (en) 2002-10-08 2006-12-26 Takeda San Diego, Inc. Histone deacetylase inhibitors
EP1608628A2 (en) 2003-03-17 2005-12-28 Takeda San Diego, Inc. Histone deacetylase inhibitors
JP4809228B2 (en) * 2003-09-24 2011-11-09 メチルジーン インコーポレイテッド Inhibitors of histone deacetylase
US7253204B2 (en) 2004-03-26 2007-08-07 Methylgene Inc. Inhibitors of histone deacetylase
US20070021612A1 (en) * 2004-11-04 2007-01-25 University Of Notre Dame Du Lac Processes and compounds for preparing histone deacetylase inhibitors and intermediates thereof
US7235688B1 (en) 2004-11-04 2007-06-26 University Of Notre Dame Du Lac Process for preparing histone deacetylase inhibitors and intermediates thereof
EP1824831A2 (en) 2004-12-16 2007-08-29 Takeda San Diego, Inc. Histone deacetylase inhibitors
JP2008540574A (en) 2005-05-11 2008-11-20 タケダ サン ディエゴ インコーポレイテッド Histone deacetylase inhibitor
CA2620333A1 (en) 2005-08-26 2007-03-01 Braincells, Inc. Neurogenesis by muscarinic receptor modulation
EP2258358A3 (en) 2005-08-26 2011-09-07 Braincells, Inc. Neurogenesis with acetylcholinesterase inhibitor
EP1940389A2 (en) 2005-10-21 2008-07-09 Braincells, Inc. Modulation of neurogenesis by pde inhibition
JP2009513672A (en) 2005-10-31 2009-04-02 ブレインセルス,インコーポレイティド GABA receptor-mediated regulation of neurogenesis
EP1965824A4 (en) * 2005-11-18 2011-10-19 Gloucester Pharmaceuticals Inc Metabolite derivatives of the hdac inhibitor fk228
US20100216734A1 (en) 2006-03-08 2010-08-26 Braincells, Inc. Modulation of neurogenesis by nootropic agents
EP2007720B1 (en) * 2006-04-07 2013-12-25 MethylGene Inc. Benzamide derivatives as inhibitors of histone deacetylase
CA2650520A1 (en) * 2006-04-24 2008-01-31 Gloucester Pharmaceuticals Treatment of ras-expressing tumors
US20090018142A9 (en) * 2006-05-02 2009-01-15 Zhengping Zhuang Use of phosphatases to treat tumors overexpressing N-CoR
JP2009536669A (en) 2006-05-09 2009-10-15 ブレインセルス,インコーポレイティド Neurogenesis by angiotensin regulation
CA2651862A1 (en) 2006-05-09 2007-11-22 Braincells, Inc. 5 ht receptor mediated neurogenesis
US8957027B2 (en) 2006-06-08 2015-02-17 Celgene Corporation Deacetylase inhibitor therapy
WO2008030651A1 (en) 2006-09-08 2008-03-13 Braincells, Inc. Combinations containing a 4-acylaminopyridine derivative
US7547781B2 (en) * 2006-09-11 2009-06-16 Curis, Inc. Quinazoline based EGFR inhibitors containing a zinc binding moiety
US8604044B2 (en) * 2006-09-11 2013-12-10 Curis, Inc. Quinazoline based EGFR inhibitors containing a zinc binding moiety
WO2008033743A1 (en) 2006-09-11 2008-03-20 Curis, Inc. Substituted 2-indolinone as ptk inhibitors containing a zinc binding moiety
WO2008033747A2 (en) * 2006-09-11 2008-03-20 Curis, Inc. Multi-functional small molecules as anti-proliferative agents
US20080161320A1 (en) * 2006-09-11 2008-07-03 Xiong Cai Fused bicyclic pyrimidines as ptk inhibitors containing a zinc binding moiety
CA2662580C (en) * 2006-09-11 2013-05-21 Curis, Inc. Tyrosine kinase inhibitors containing a zinc binding moiety
US20100184806A1 (en) 2006-09-19 2010-07-22 Braincells, Inc. Modulation of neurogenesis by ppar agents
BRPI0720734A2 (en) * 2006-12-29 2014-01-07 Gloucester Pharmaceuticals Inc ROMIDEPSIN PREPARATION
EP2117598A2 (en) * 2007-01-10 2009-11-18 Novartis AG Formulations of deacetylase inhibitors
CN101662939B (en) 2007-02-06 2015-11-25 利克斯特生物技术公司 Oxa-bicyclo heptane and oxabicyclo heptene, their preparation and purposes
JP2010522163A (en) * 2007-03-20 2010-07-01 キュリス,インコーポレイテッド Raf kinase inhibitors containing zinc binding sites
US20090035292A1 (en) * 2007-08-03 2009-02-05 Kovach John S Use of phosphatases to treat neuroblastomas and medulloblastomas
US8119616B2 (en) * 2007-09-10 2012-02-21 Curis, Inc. Formulation of quinazoline based EGFR inhibitors containing a zinc binding moiety
TW200922564A (en) * 2007-09-10 2009-06-01 Curis Inc CDK inhibitors containing a zinc binding moiety
WO2009035718A1 (en) 2007-09-10 2009-03-19 Curis, Inc. Tartrate salts or complexes of quinazoline based egfr inhibitors containing a zinc binding moiety
MX2010003417A (en) * 2007-10-01 2010-09-10 Lixte Biotechnology Inc Hdac inhibitors.
WO2010014220A1 (en) * 2008-08-01 2010-02-04 Lixte Biotechnology, Inc. Neuroprotective agents for the prevention and treatment of neurodegenerative diseases
US8227473B2 (en) 2008-08-01 2012-07-24 Lixte Biotechnology, Inc. Oxabicycloheptanes and oxabicycloheptenes, their preparation and use
EP2309853A4 (en) * 2008-08-01 2012-04-25 Lixte Biotechnology Inc Methods for regulating cell mitosis by inhibiting serine/threonine phosphatase
WO2010147612A1 (en) 2009-06-18 2010-12-23 Lixte Biotechnology, Inc. Methods of modulating cell regulation by inhibiting p53
EP2330894B8 (en) 2008-09-03 2017-04-19 BioMarin Pharmaceutical Inc. Compositions including 6-aminohexanoic acid derivatives as hdac inhibitors
KR101048315B1 (en) 2008-09-29 2011-07-13 연세대학교 산학협력단 Pine Red Nematode Histone Deacetylase
HUE027615T2 (en) 2009-01-08 2016-10-28 Curis Inc Phosphoinositide 3-kinase inhibitors with a zinc binding moiety
WO2010099217A1 (en) 2009-02-25 2010-09-02 Braincells, Inc. Modulation of neurogenesis using d-cycloserine combinations
TW201735926A (en) 2010-01-22 2017-10-16 艾斯特隆製藥公司 Reverse amide compounds as protein deacetylase inhibitors and methods of use thereof
RU2016149485A (en) 2010-07-12 2018-10-31 Селджин Корпорейшн SOLID FORMS OF ROMIDEPSIN AND THEIR APPLICATION
US8859502B2 (en) 2010-09-13 2014-10-14 Celgene Corporation Therapy for MLL-rearranged leukemia
US8614223B2 (en) 2010-11-16 2013-12-24 Acetylon Pharmaceuticals, Inc. Pyrimidine hydroxy amide compounds as protein deacetylase inhibitors and methods of use thereof
US8957066B2 (en) 2011-02-28 2015-02-17 Biomarin Pharmaceutical Inc. Histone deacetylase inhibitors
US10059723B2 (en) 2011-02-28 2018-08-28 Biomarin Pharmaceutical Inc. Histone deacetylase inhibitors
ES2712803T3 (en) 2011-02-28 2019-05-14 Biomarin Pharm Inc Histone deacetylase inhibitors
ME03523B (en) 2011-04-01 2020-04-20 Curis Inc Phosphoinositide 3-kinase inhibitor with a zinc binding moiety
AU2013202506B2 (en) 2012-09-07 2015-06-18 Celgene Corporation Resistance biomarkers for hdac inhibitors
AU2013202507B9 (en) 2012-11-14 2015-08-13 Celgene Corporation Inhibition of drug resistant cancer cells
CN105121415B (en) 2013-03-15 2018-10-12 生物马林药物股份有限公司 Hdac inhibitor
KR20160004299A (en) 2013-04-09 2016-01-12 릭스트 바이오테크놀로지, 인코포레이티드 Formulations of oxabicycloheptanes and oxabicycloheptenes
ES2929576T3 (en) 2013-10-08 2022-11-30 Acetylon Pharmaceuticals Inc Combinations of histone deacetylase 6 inhibitors and the Her2 inhibitor lapatinib for use in the treatment of breast cancer
EP3055299B1 (en) 2013-10-10 2021-01-06 Acetylon Pharmaceuticals, Inc. Pyrimidine hydroxy amide compounds as histone deacetylase inhibitors
JP2016534069A (en) 2013-10-24 2016-11-04 メイヨ・ファウンデーション・フォー・メディカル・エデュケーション・アンド・リサーチ Treatment of polycystic disease with HDAC6 inhibitors
PE20161030A1 (en) 2013-12-03 2016-11-06 Acetylon Pharmaceuticals Inc COMBINATIONS OF ACETYLASE HISTONE INHIBITORS AND IMMUNOMODULATING DRUGS
NZ630311A (en) 2013-12-27 2016-03-31 Celgene Corp Romidepsin formulations and uses thereof
US9636298B2 (en) 2014-01-17 2017-05-02 Methylgene Inc. Prodrugs of compounds that enhance antifungal activity and compositions of said prodrugs
US9464073B2 (en) 2014-02-26 2016-10-11 Acetylon Pharmaceuticals, Inc. Pyrimidine hydroxy amide compounds as HDAC6 selective inhibitors
EP3166603B1 (en) 2014-07-07 2020-02-12 Acetylon Pharmaceuticals, Inc. Treatment of leukemia with histone deacetylase inhibitors
CA2969790A1 (en) 2014-12-05 2016-06-09 University of Modena and Reggio Emilia Combinations of histone deacetylase inhibitors and bendamustine
US10272084B2 (en) 2015-06-01 2019-04-30 Regenacy Pharmaceuticals, Llc Histone deacetylase 6 selective inhibitors for the treatment of cisplatin-induced peripheral neuropathy
ES2769255T3 (en) 2015-06-08 2020-06-25 Acetylon Pharmaceuticals Inc Methods for making protein deacetylase inhibitors
US10464906B2 (en) 2015-06-08 2019-11-05 Acetylon Pharmaceuticals, Inc. Crystalline forms of a histone deacetylase inhibitor
JP2019515909A (en) 2016-04-19 2019-06-13 アセチロン ファーマシューティカルズ インコーポレイテッドAcetylon Pharmaceuticals,Inc. HDAC inhibitor alone or in combination with a BTK inhibitor for the treatment of chronic lymphocytic leukemia
WO2018031472A1 (en) 2016-08-08 2018-02-15 Acetylon Pharmaceuticals Inc. Pharmaceutical combinations of histone deacetylase 6 inhibitors and cd20 inhibitory antibodies and uses thereof
US11234986B2 (en) 2018-09-11 2022-02-01 Curis, Inc. Combination therapy with a phosphoinositide 3-kinase inhibitor with a zinc binding moiety

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0708112A1 (en) * 1994-09-22 1996-04-24 Cancer Institute RPDL protein and DNA encoding the same
WO1997011366A1 (en) * 1995-09-20 1997-03-27 Merck & Co., Inc. Histone deacetylase as target for antiprotozoal agents

Family Cites Families (127)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HU165117B (en) * 1971-08-12 1974-06-28
US4160866A (en) * 1976-04-30 1979-07-10 Imperial Chemical Industries Limited Carbamates
DE3242252A1 (en) 1982-11-15 1984-05-17 Bayer Ag, 5090 Leverkusen HETEROCYCLICALLY SUBSTITUTED HYDROXYALKYL-AZOLYL DERIVATIVES
NL194579C (en) 1983-01-21 2002-08-05 Schering Ag Diagnostic.
US4631211A (en) * 1985-03-25 1986-12-23 Scripps Clinic & Research Foundation Means for sequential solid phase organic synthesis and methods using the same
US4683202A (en) * 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4608390A (en) * 1985-04-26 1986-08-26 Abbott Laboratories Lipoxygenase inhibiting compounds
US4833080A (en) * 1985-12-12 1989-05-23 President And Fellows Of Harvard College Regulation of eucaryotic gene expression
CA1319101C (en) 1986-09-03 1993-06-15 Marta Iris Sabara Rotavirus nucleocapsid protein with or without binding peptides as immunologic carriers for macromolecules
US4861798A (en) * 1986-12-29 1989-08-29 Bristol-Myers Company Lipoxygenase inhibitory compounds
US4820828A (en) * 1987-03-04 1989-04-11 Ortho Pharmaceutical Corporation Cinnamohydroxamic acids
US5080891A (en) * 1987-08-03 1992-01-14 Ddi Pharmaceuticals, Inc. Conjugates of superoxide dismutase coupled to high molecular weight polyalkylene glycols
ES2008962A6 (en) 1987-12-17 1989-08-16 Marga Investigacion New-quanidino-thiazol compounds, their preparation, and use as intermediates of famotidine process.
EP0323590A3 (en) 1987-12-24 1990-05-02 Ono Pharmaceutical Co., Ltd. Carbazoyl derivatives
JPH01224381A (en) 1988-03-04 1989-09-07 Japan Tobacco Inc Trichostatic acid, novel synthetic intermediate for trichostatin a and production of trichostatic acid and trichostatin a
DE58900825D1 (en) * 1988-06-13 1992-03-26 Ciba Geigy Ag UNSATURATED BETA KETOESTER ACETALS AND THEIR APPLICATIONS.
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5175191A (en) * 1988-11-14 1992-12-29 Sloan-Kettering Institute For Cancer Research Potent inducers of terminal differentiation and methods of use thereof
WO1991000257A1 (en) 1989-06-30 1991-01-10 The Trustees Of Columbia University In The City Of New York Novel potent inducers of terminal differentiation and methods of use thereof
US5176996A (en) * 1988-12-20 1993-01-05 Baylor College Of Medicine Method for making synthetic oligonucleotides which bind specifically to target sites on duplex DNA molecules, by forming a colinear triplex, the synthetic oligonucleotides and methods of use
US5096815A (en) * 1989-01-06 1992-03-17 Protein Engineering Corporation Generation and selection of novel dna-binding proteins and polypeptides
US5198346A (en) * 1989-01-06 1993-03-30 Protein Engineering Corp. Generation and selection of novel DNA-binding proteins and polypeptides
US5328470A (en) * 1989-03-31 1994-07-12 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
US5256775A (en) * 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US5143854A (en) * 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5264564A (en) * 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
AU6886791A (en) 1989-11-13 1991-06-13 Affymax Technologies N.V. Spatially-addressable immobilization of anti-ligands on surfaces
US5304121A (en) * 1990-12-28 1994-04-19 Boston Scientific Corporation Drug delivery system making use of a hydrogel polymer coating
DE69127130T2 (en) 1990-05-09 1997-12-11 Fuji Photo Film Co Ltd Photographic processing composition and processing method using the same
US5045538A (en) * 1990-06-28 1991-09-03 The Research Foundation Of State University Of New York Inhibition of wasting and protein degradation of mammalian muscle by tetracyclines
GB9026114D0 (en) * 1990-11-30 1991-01-16 Norsk Hydro As New compounds
EP0564531B1 (en) 1990-12-03 1998-03-25 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
JPH06504997A (en) 1990-12-06 1994-06-09 アフィメトリックス, インコーポレイテッド Synthesis of immobilized polymers on a very large scale
EP0501919A1 (en) * 1991-03-01 1992-09-02 Ciba-Geigy Ag Radiation-sensitive compositions based on polyphenols and acetals
WO1992015694A1 (en) 1991-03-08 1992-09-17 The Salk Institute For Biological Studies Flp-mediated gene modification in mammalian cells, and compositions and cells useful therefor
US5225173A (en) * 1991-06-12 1993-07-06 Idaho Research Foundation, Inc. Methods and devices for the separation of radioactive rare earth metal isotopes from their alkaline earth metal precursors
WO1993005807A2 (en) 1991-09-13 1993-04-01 New England Deaconess Hospital Erythropoietin potentiating agents and methods for their use
US5700811A (en) * 1991-10-04 1997-12-23 Sloan-Kettering Institute For Cancer Research Potent inducers of terminal differentiation and method of use thereof
US5369108A (en) 1991-10-04 1994-11-29 Sloan-Kettering Institute For Cancer Research Potent inducers of terminal differentiation and methods of use thereof
US5239113A (en) 1991-10-15 1993-08-24 Monsanto Company Substituted β-amino acid derivatives useful as platelet aggregation inhibitors and intermediates thereof
EP0916396B1 (en) 1991-11-22 2005-04-13 Affymetrix, Inc. (a Delaware Corporation) Combinatorial strategies for polymer synthesis
DK0570594T3 (en) * 1991-12-10 1997-08-25 Shionogi & Co
US5359115A (en) * 1992-03-26 1994-10-25 Affymax Technologies, N.V. Methods for the synthesis of phosphonate esters
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
WO1993019778A1 (en) 1992-04-07 1993-10-14 The Scripps Research Institute Method for inducing tolerance to an antigen using butyrate
US5288514A (en) * 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
PT665897E (en) 1992-10-01 2003-11-28 Trustees Of Columbia U In The COMPLEX COMBINATION CHEMICAL LIBRARIES ENCODED WITH LABELS
ATE316573T1 (en) 1992-10-30 2006-02-15 Gen Hospital Corp INTERACTING TRAP SYSTEM FOR ISOLATION OF PROTEINS
US5480971A (en) * 1993-06-17 1996-01-02 Houghten Pharmaceuticals, Inc. Peralkylated oligopeptide mixtures
US5440016A (en) * 1993-06-18 1995-08-08 Torrey Pines Institute For Molecular Studies Peptides of the formula (KFmoc) ZZZ and their uses
US5716981A (en) * 1993-07-19 1998-02-10 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use
US20030203976A1 (en) * 1993-07-19 2003-10-30 William L. Hunter Anti-angiogenic compositions and methods of use
US5362899A (en) * 1993-09-09 1994-11-08 Affymax Technologies, N.V. Chiral synthesis of alpha-aminophosponic acids
US5440061A (en) * 1994-03-29 1995-08-08 The Procter & Gamble Company Hydrolysis of methyl esters in dimethylsulfoxide for production of fatty acids
US6231600B1 (en) * 1995-02-22 2001-05-15 Scimed Life Systems, Inc. Stents with hybrid coating for medical devices
US6099562A (en) * 1996-06-13 2000-08-08 Schneider (Usa) Inc. Drug coating with topcoat
US5837313A (en) * 1995-04-19 1998-11-17 Schneider (Usa) Inc Drug release stent coating process
JP3330781B2 (en) 1995-05-22 2002-09-30 三菱エンジニアリングプラスチックス株式会社 Polycarbonate resin composition
JPH09124918A (en) 1995-10-30 1997-05-13 Mitsubishi Eng Plast Kk Polycarbonate resin composition
US6030945A (en) * 1996-01-09 2000-02-29 Genentech, Inc. Apo-2 ligand
US6777217B1 (en) * 1996-03-26 2004-08-17 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
US6068987A (en) * 1996-09-20 2000-05-30 Merck & Co., Inc. Histone deacetylase as target for antiprotozoal agents
WO1998016830A2 (en) 1996-10-16 1998-04-23 The President And Fellows Of Harvard College Droplet assay system
ZA9710342B (en) * 1996-11-25 1998-06-10 Alza Corp Directional drug delivery stent and method of use.
US6273913B1 (en) * 1997-04-18 2001-08-14 Cordis Corporation Modified stent useful for delivery of drugs along stent strut
ATE289295T1 (en) 1997-04-22 2005-03-15 Euro Celtique Sa THE USE OF SEMICARBAZONES AND THIOSEMICARBAZONES SUBSTITUTED BY CARBOCYCLIC AND HETEROCYCLIC MATERIALS AS SODIUM CHANNEL BLOCKERS
AUPO721997A0 (en) 1997-06-06 1997-07-03 Queensland Institute Of Medical Research, The Anticancer compounds
US6190619B1 (en) * 1997-06-11 2001-02-20 Argonaut Technologies, Inc. Systems and methods for parallel synthesis of compounds
US5891507A (en) * 1997-07-28 1999-04-06 Iowa-India Investments Company Limited Process for coating a surface of a metallic stent
US6195612B1 (en) * 1998-01-05 2001-02-27 Tama L. Pack-Harris Pharmacy benefit management system and method of using same
US6503708B1 (en) 1998-01-26 2003-01-07 Incyte Genomics, Inc. Microtubule-associated protein
US6428960B1 (en) * 1998-03-04 2002-08-06 Onyx Pharmaceuticals, Inc. Selection method for producing recombinant baculovirus
US6153252A (en) * 1998-06-30 2000-11-28 Ethicon, Inc. Process for coating stents
US6248127B1 (en) * 1998-08-21 2001-06-19 Medtronic Ave, Inc. Thromboresistant coated medical device
DE19846008A1 (en) 1998-10-06 2000-04-13 Bayer Ag Phenylacetic acid heterocyclylamide
TR200101663T2 (en) 1998-12-10 2001-11-21 F.Hoffmann-La Roche Ag Pro-collagen c-proteinase inhibitors
WO2000036132A1 (en) 1998-12-14 2000-06-22 Merck & Co., Inc. Hiv integrase inhibitors
ES2188280T3 (en) 1998-12-16 2003-06-16 Aventis Pharma Ltd ACETALES HETEROARIL-CICLICOS.
AR035313A1 (en) 1999-01-27 2004-05-12 Wyeth Corp ACETILENIC TACE INHIBITORS OF HYDROXAMIC ACID OF SULFONAMIDE BASED ON ALFA-AMINO ACIDS, PHARMACEUTICAL COMPOSITIONS AND THE USE OF THE SAME FOR THE MANUFACTURE OF MEDICINES.
US6258121B1 (en) * 1999-07-02 2001-07-10 Scimed Life Systems, Inc. Stent coating
US6203551B1 (en) * 1999-10-04 2001-03-20 Advanced Cardiovascular Systems, Inc. Chamber for applying therapeutic substances to an implant device
KR20100035666A (en) * 1999-11-23 2010-04-05 메틸진 인크. Inhibitors of histone deacetylase
US6251136B1 (en) * 1999-12-08 2001-06-26 Advanced Cardiovascular Systems, Inc. Method of layering a three-coated stent using pharmacological and polymeric agents
US20030129724A1 (en) * 2000-03-03 2003-07-10 Grozinger Christina M. Class II human histone deacetylases, and uses related thereto
PE20020354A1 (en) 2000-09-01 2002-06-12 Novartis Ag HYDROXAMATE COMPOUNDS AS HISTONE-DESACETILASE (HDA) INHIBITORS
CA2423868C (en) * 2000-09-29 2011-06-07 Prolifix Limited Carbamic acid compounds comprising an amide linkage as hdac inhibitors
GB0023983D0 (en) * 2000-09-29 2000-11-15 Prolifix Ltd Therapeutic compounds
US6495719B2 (en) * 2001-03-27 2002-12-17 Circagen Pharmaceutical Histone deacetylase inhibitors
AR035455A1 (en) * 2001-04-23 2004-05-26 Hoffmann La Roche TRICYCLE DERIVATIVES OF ALQUILHIDROXAMATO, PROCESSES FOR THEIR DEVELOPMENT, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM, AND THE USE OF SUCH COMPOUNDS IN THE PREPARATION OF MEDICINES
US7244853B2 (en) * 2001-05-09 2007-07-17 President And Fellows Of Harvard College Dioxanes and uses thereof
AU2002305523A1 (en) 2001-05-09 2002-11-18 President And Fellows Of Harvard College Dioxanes and uses thereof
US6897220B2 (en) * 2001-09-14 2005-05-24 Methylgene, Inc. Inhibitors of histone deacetylase
US6517889B1 (en) * 2001-11-26 2003-02-11 Swaminathan Jayaraman Process for coating a surface of a stent
AU2002362030A1 (en) 2001-11-27 2003-06-10 Fred Hutchinson Cancer Research Center Methods for inhibiting deacetylase activity
US7148257B2 (en) * 2002-03-04 2006-12-12 Merck Hdac Research, Llc Methods of treating mesothelioma with suberoylanilide hydroxamic acid
EP2266552A3 (en) * 2002-03-04 2011-03-02 Merck HDAC Research, LLC Methods of inducing terminal differentiation
JP2004043446A (en) 2002-05-15 2004-02-12 Schering Ag Histone deacetylase inhibitor and use thereof
NO20032183L (en) * 2002-05-15 2003-11-17 Schering Ag Histone deacetylase inhibitor and its use
WO2003101481A1 (en) * 2002-06-03 2003-12-11 Als Therapy Development Foundation Treatment of neurodegenerative diseases using proteasome modulators
TW200401638A (en) 2002-06-20 2004-02-01 Bristol Myers Squibb Co Heterocyclic inhibitors of kinases
TW200420573A (en) 2002-09-26 2004-10-16 Rib X Pharmaceuticals Inc Bifunctional heterocyclic compounds and methods of making and using same
JP2006503082A (en) * 2002-10-17 2006-01-26 メシルジーン、インコーポレイテッド Inhibitors of histone deacetylase
EP1565193B1 (en) * 2002-11-06 2013-04-24 Dana-Farber Cancer Institute, Inc. Compositions for treating cancer using proteasome inhibitor PS-341
CA2506504A1 (en) 2002-11-20 2004-06-03 Errant Gene Therapeutics, Llc Treatment of lung cells with histone deacetylase inhibitors
CA2851462A1 (en) 2003-01-08 2004-07-29 University Of Washington Antibacterial agents
EP1599449A2 (en) * 2003-02-25 2005-11-30 TopoTarget UK Limited Carbamic acid compounds comprising a bicyclic heteroaryl group as hdac inhibitors
PE20050206A1 (en) 2003-05-26 2005-03-26 Schering Ag PHARMACEUTICAL COMPOSITION CONTAINING AN INHIBITOR OF HISTONE DEACETILASE
CA2531661C (en) 2003-07-07 2013-03-12 Georgetown University Histone deacetylase inhibitors and methods of use thereof
US7842835B2 (en) * 2003-07-07 2010-11-30 Georgetown University Histone deacetylase inhibitors and methods of use thereof
WO2005012247A1 (en) 2003-07-30 2005-02-10 Hôpital Sainte-Justine Compounds and methods for the rapid quantitative analysis of proteins and polypeptides
BRPI0413826A (en) 2003-08-26 2006-10-24 Aton Pharma Inc process of treating cancer with hdac inhibitors
US20050137234A1 (en) 2003-12-19 2005-06-23 Syrrx, Inc. Histone deacetylase inhibitors
JP4989976B2 (en) * 2004-02-13 2012-08-01 プレジデント アンド フェロウズ オブ ハーバード カレッジ 3-3 Disubstituted oxindoles as translation initiation inhibitors
PL1830838T3 (en) 2004-12-03 2013-07-31 Dana Farber Cancer Inst Inc Compositions and methods for treating neoplastic diseases
WO2006060809A2 (en) 2004-12-03 2006-06-08 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof
EP3354265A1 (en) * 2005-03-22 2018-08-01 President and Fellows of Harvard College Treatment of solid tumors
JP5441416B2 (en) * 2006-02-14 2014-03-12 プレジデント アンド フェロウズ オブ ハーバード カレッジ Bifunctional histone deacetylase inhibitor
MX2008010462A (en) 2006-02-14 2009-04-17 Harvard College Histone Deacetylase Inhibitors.
CN101431996A (en) 2006-03-22 2009-05-13 加利福尼亚大学董事会 Inhibitors of protein prenyltransferases
EP2019674B1 (en) * 2006-05-03 2016-11-23 The President and Fellows of Harvard College Histone deacetylase and tubulin deacetylase inhibitors
GB0619753D0 (en) 2006-10-06 2006-11-15 Chroma Therapeutics Ltd Enzyme inhibitors
US8088951B2 (en) * 2006-11-30 2012-01-03 Massachusetts Institute Of Technology Epigenetic mechanisms re-establish access to long-term memory after neuronal loss
EP2060565A1 (en) 2007-11-16 2009-05-20 4Sc Ag Novel bifunctional compounds which inhibit protein kinases and histone deacetylases
US20090205384A1 (en) * 2008-02-18 2009-08-20 Sandisk Il Ltd. Electromechanical locking system
RU2515611C2 (en) * 2008-07-23 2014-05-20 Президент Энд Феллоуз Оф Гарвард Колледж Deacetylase inhibitors and their application
WO2011019393A2 (en) 2009-08-11 2011-02-17 President And Fellows Of Harvard College Class- and isoform-specific hdac inhibitors and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0708112A1 (en) * 1994-09-22 1996-04-24 Cancer Institute RPDL protein and DNA encoding the same
WO1997011366A1 (en) * 1995-09-20 1997-03-27 Merck & Co., Inc. Histone deacetylase as target for antiprotozoal agents

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
BIOCHEMICAL JOURNAL, vol. 303, no. 3, 1 November 1994, pages 723-729, XP002038740 M. MATEO SANCHEZ DEL PINO ET AL.: "Properties of the yeast nuclear histone deacetylase" *
CYTOGENETICS AND CELL GENETICS, vol. 73, no. 1-2, 1996, pages 130-133, XP002038744 Y. FURUKAWA ET AL.: "Isolation and mapping of a human gene (RPD3L1) that is homologous to RPD3, a transcription factor in Saccharomyces cerevisiae" *
EMBL Database Entry HSU31814 ion number U31814; 19 November 1996 YANG W AL.:"Transcriptional repression b is mediated by interaction with a XP002038746 & PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, vol. 93, no. 23, 12 November 1996, WASHINGTON US, pages 12845-12850, WEN-MIG YANG ET AL.: "Transcriptional repression by YY1 is mediated by interaction with a mammalian homolog of the yeast global regulator RPD3" *
JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 265, no. 28, 5 October 1990, MD US, pages 17174-17179, XP000616087 MINORU YOSHIDA ET AL.: "Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro by Trichostatin A" *
JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 268, no. 30, 25 October 1993, MD US, pages 22429-22435, XP000616088 MASAKO KIJIMA ET AL.: "Trapoxin, an antitumor cyclic tetrapeptide, is an irreversible inhibitor of mammalian histone deacetylase" *
JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 270, no. 42, 20 October 1995, MD US, pages 24674-24677, XP002038741 SUSANNE KLEFF ET AL.: "Identification of a gene encoding a yeast histone H4 acetyltransferase" cited in the application *
JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 271, no. 26, 28 June 1996, MD US, pages 15837-15844, XP002038745 ANDREW A. CARMEN ET AL.: "HDA1 and HDA3 are components of a yeast histone deacetylase (HDA) complex" *
MOLECULAR AND CELLULAR BIOLOGY, vol. 11, no. 12, December 1991, pages 6317-6327, XP002038742 MARC VIDAL ET AL.: "RPD3 encodes a second factor required to achieve maximum positive and negative transcriptional states in Saccharomyces cerevisiae" *
SCIENCE, vol. 272, 19 April 1996, LANCASTER, PA US, pages 408-411, XP002038743 JACK TAUNTON ET AL.: "A mammalian histone deacetylase related to the yeast transcriptional regulator Rpd3p" *

Cited By (91)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8362084B2 (en) 1996-03-26 2013-01-29 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
US8399233B2 (en) 1996-03-26 2013-03-19 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
US8426592B2 (en) 1996-03-26 2013-04-23 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
US8329945B2 (en) 1996-03-26 2012-12-11 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
US8329946B2 (en) 1996-03-26 2012-12-11 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
US6706762B1 (en) 1997-05-01 2004-03-16 The Salk Institute For Biological Studies Methods for the use of inhibitors of co-repressors for the treatment of neoplastic diseases
EP1010705A4 (en) * 1997-09-02 2004-09-15 Sumitomo Pharma Novel cyclic tetrapeptide derivatives and medicinal use thereof
EP1010705A1 (en) * 1997-09-02 2000-06-21 Japan Energy Corporation Novel cyclic tetrapeptide derivatives and medicinal use thereof
WO1999023885A1 (en) * 1997-11-10 1999-05-20 The Salk Institute For Biological Studies Methods for the use of inhibitors of co-repressors for the treatment of neoplastic diseases
US6890709B1 (en) 1998-05-15 2005-05-10 Chroma Therapeutics Limited Assays, methods and means for modulating e2f activity
WO1999060407A1 (en) * 1998-05-15 1999-11-25 Cancer Research Campaign Technogoly Limited Assays, methods and means for modulating e2f activity
WO2000008048A2 (en) * 1998-08-04 2000-02-17 Fujisawa Pharmaceutical Co., Ltd. Inhibitor of histone deacetylase
WO2000008048A3 (en) * 1998-08-04 2002-08-15 Fujisawa Pharmaceutical Co Inhibitor of histone deacetylase
EP1105143A4 (en) * 1998-08-21 2002-07-17 Smithkline Beecham Corp Human histone deacetylase gene hd4
EP1105143A1 (en) * 1998-08-21 2001-06-13 SmithKline Beecham Corporation Human histone deacetylase gene hd4
WO2000021979A2 (en) * 1998-10-13 2000-04-20 Fujisawa Pharmaceutical Co., Ltd. Cyclic tetrapeptide and their use as histone deacetylase inhibitor
WO2000021979A3 (en) * 1998-10-13 2000-07-20 Fujisawa Pharmaceutical Co Cyclic tetrapeptide and their use as histone deacetylase inhibitor
US6656905B1 (en) 1998-10-13 2003-12-02 Fujisawa Pharmaceutical Co., Ltd. Cyclic tetrapeptide compound and use thereof
US6953783B1 (en) 1998-10-19 2005-10-11 Methylgene, Inc. Modulation of gene expression by combination therapy
EP1243290A3 (en) * 1998-10-19 2004-03-17 Methylgene, Inc. Modulation of gene expression by combination therapy
EP1243290A2 (en) * 1998-10-19 2002-09-25 Methylgene, Inc. Modulation of gene expression by combination therapy
WO2000071703A2 (en) * 1999-05-03 2000-11-30 Methylgene Inc. Inhibition of histone deacetylase
WO2000071703A3 (en) * 1999-05-03 2001-07-19 Methylgene Inc Inhibition of histone deacetylase
US6808926B1 (en) 1999-08-27 2004-10-26 Her Majesty The Queen In Right Of Canada, As Represented By The Minister Of Agriculture And Agri-Food Repressing gene expression in plants
US6403555B1 (en) 1999-12-08 2002-06-11 Xcyte Therapies, Inc. Depsipeptide and congeners thereof for use as immunosuppressants
US6828302B1 (en) 1999-12-08 2004-12-07 Xcyte Therapies, Inc. Therapeutic uses of depsipeptides and congeners thereof
JP4824890B2 (en) * 1999-12-08 2011-11-30 エクサイト セラピーズ インコーポレーティッド Depsipeptides and their homologs for use as immunosuppressants
US6548479B1 (en) 1999-12-08 2003-04-15 Xcyte Therapies, Inc. Therapeutic uses of depsipeptides and congeners thereof
US7041639B2 (en) 1999-12-08 2006-05-09 Xcyte Therapies, Inc. Depsipeptide and congeners thereof for use as immunosuppressants
US8895284B2 (en) 2000-03-03 2014-11-25 President And Fellows Of Harvard College Class II human histone deacetylases, and uses related thereto
US8435780B2 (en) 2000-03-03 2013-05-07 President And Fellows Of Harvard College Class II human histone deacetylases, and uses related thereto
US8076116B2 (en) 2000-03-03 2011-12-13 President And Fellows Of Harvard College Nucleic acids encoding class II human histone deacetylases, and uses related thereto
US7250504B2 (en) 2000-03-03 2007-07-31 President And Fellows Of Harvard College Class II human histone deacetylases, and uses related thereto
WO2001067107A1 (en) * 2000-03-04 2001-09-13 Imperial College Innovations Limited Modulation of histone deacetylase
WO2003006652A2 (en) * 2000-03-24 2003-01-23 Methylgene, Inc. Inhibition of specific histone deacetylase isoforms
WO2003006652A3 (en) * 2000-03-24 2004-05-13 Methylgene Inc Inhibition of specific histone deacetylase isoforms
WO2002007722A2 (en) * 2000-07-07 2002-01-31 Georg-Speyer-Haus Valproic acid and derivatives thereof as histone deacetylase inhibitors
WO2002007722A3 (en) * 2000-07-07 2002-07-18 Georg Speyer Haus Valproic acid and derivatives thereof as histone deacetylase inhibitors
EP1170008A1 (en) * 2000-07-07 2002-01-09 Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus Valproic acid and derivatives thereof as histone deacetylase inhibitors
US7265154B2 (en) 2000-07-07 2007-09-04 Georg-Speyter-Haus Chemotherapeutisches Forschungsinstitut Valproic acid and derivatives thereof as histone deacetylase inhibitors
US7067551B2 (en) 2000-09-01 2006-06-27 Novartis Ag Deacetylase inhibitors
US6552065B2 (en) 2000-09-01 2003-04-22 Novartis Ag Deacetylase inhibitors
WO2002030970A3 (en) * 2000-10-13 2002-07-11 Bayer Ag Human histone deacetylase gene
WO2002030970A2 (en) * 2000-10-13 2002-04-18 Bayer Aktiengesellschaft Human histone deacetylase gene
US7557141B2 (en) 2000-11-21 2009-07-07 Wake Forest University Health Sciences Method of treating autoimmune diseases
US7271198B2 (en) 2000-11-21 2007-09-18 Wake Forest University Method of treating autoimmune diseases
WO2002069947A2 (en) * 2001-01-12 2002-09-12 Methylgene, Inc. Antisense oligonucleotides and trichostatin analogue histone deacetylase-4 inhibitors against cancer
WO2002069947A3 (en) * 2001-01-12 2003-10-09 Methylgene Inc Antisense oligonucleotides and trichostatin analogue histone deacetylase-4 inhibitors against cancer
EP1390491A4 (en) * 2001-05-02 2005-04-06 Univ California Method for treating neurodegenerative, psychiatric and other disorders with deacetylase inhibitors
EP1390491A1 (en) * 2001-05-02 2004-02-25 The Regents of the University of California Method for treating neurodegenerative, psychiatric and other disorders with deacetylase inhibitors
US7244853B2 (en) 2001-05-09 2007-07-17 President And Fellows Of Harvard College Dioxanes and uses thereof
US7063973B2 (en) 2001-06-14 2006-06-20 Sloan-Kettering Institute For Cancer Research HDAC9 polypeptides and polynucleotides and uses thereof
US7816499B2 (en) 2001-06-14 2010-10-19 Sloan-Kettering Institute For Cancer Research Antibodies that selectively bind HDAC9
US7244604B2 (en) 2001-06-14 2007-07-17 Sloan-Kettering Institute For Cancer Research HDAC9 polypeptides and polynucleotides and uses thereof
WO2003013493A1 (en) * 2001-08-07 2003-02-20 Italfarmaco S.P.A. Histone deacetylase enzyme-inhibiting derivatives of hydroxamic acid as new cytokine synthesis-inhibiting anti-inflammatory drugs
WO2003032921A2 (en) 2001-10-16 2003-04-24 Sloan-Kettering Institute For Cancer Research Treatment of neurodegenerative diseases and cancer of the brain
US7879865B2 (en) 2001-10-16 2011-02-01 Sloan-Kettering Institute For Cancer Research Treatment of cancer of the brain using histone deacetylase inhibitors
US7566535B2 (en) 2002-03-07 2009-07-28 University Of Delaware Enhanced oligonucleotide-mediated nucleic acid sequence alteration
US7057057B2 (en) 2002-05-22 2006-06-06 Errant Gene Therapeutics, Llc Histone deacetylase inhibitors based on alpha-ketoepoxide compounds
US7579372B2 (en) 2002-05-22 2009-08-25 Errant Gene Therapeutics, Llc Histone deacetylase inhibitors based on alpha-ketoepoxide compounds
US7582807B2 (en) 2002-06-06 2009-09-01 Her Majesty The Queen In Right Of Canada, As Represented By The Minister Of Agriculture And Agri-Food Regulation of gene expression using chromatin remodelling factors
US7553667B2 (en) 2002-06-06 2009-06-30 Her Majesty The Queen In Right Of Canada As Represented By The Minister Of Agriculture And Agri-Food Regulation of gene expression using chromatin remodelling factors
WO2004027418A3 (en) * 2002-09-18 2004-05-13 G2M Cancer Drugs Ag The use of molecular markers for the preclinical and clinical profiling of inhibitors of enzymes having histone deacetylase activity
EP1400806A1 (en) * 2002-09-18 2004-03-24 G2M Cancer Drugs AG The use of molecular markers for the preclinical and clinical profiling of inhibitors of enzymes having histone deacetylase activity
WO2004027418A2 (en) * 2002-09-18 2004-04-01 G2M Cancer Drugs Ag The use of molecular markers for the preclinical and clinical profiling of inhibitors of enzymes having histone deacetylase activity
AU2003267386B2 (en) * 2002-09-18 2010-07-01 Karlsruher Institut Fur Technologie The use of molecular markers for the preclinical and clinical profiling of inhibitors of enzymes having histone deacetylase activity
US10172905B1 (en) 2005-03-22 2019-01-08 President And Fellows Of Harvard College Treatment of protein degradation disorders
US9572854B2 (en) 2005-03-22 2017-02-21 President And Fellows Of Harvard College Treatment of protein degradation disorders
US8999289B2 (en) 2005-03-22 2015-04-07 President And Fellows Of Harvard College Treatment of protein degradation disorders
US7732475B2 (en) 2005-07-14 2010-06-08 Takeda San Diego, Inc. Histone deacetylase inhibitors
US7741494B2 (en) 2005-07-14 2010-06-22 Takeda San Diego, Inc. Histone deacetylase inhibitors
US8383855B2 (en) 2006-02-14 2013-02-26 President And Fellows Of Harvard College Histone deacetylase inhibitors
US9724321B2 (en) 2006-02-14 2017-08-08 President & Fellows Of Harvard College Histone deacetylase inhibitors
US8222423B2 (en) 2006-02-14 2012-07-17 Dana-Farber Cancer Institute, Inc. Bifunctional histone deacetylase inhibitors
US10172821B2 (en) 2006-02-14 2019-01-08 President & Fellows of Harvard College Dana-Farber Cancer Institute, Inc. Histone deacetylase inhibitors
US8754237B2 (en) 2006-02-14 2014-06-17 President And Fellows Of Harvard College Bifunctional histone deacetylase inhibitors
US8304451B2 (en) 2006-05-03 2012-11-06 President And Fellows Of Harvard College Histone deacetylase and tubulin deacetylase inhibitors
US8691534B2 (en) 2006-12-29 2014-04-08 Celgene Corporation Preparation of romidepsin
US8440716B2 (en) 2008-07-23 2013-05-14 President And Fellows Of Harvard College Deacetylase inhibitors and uses thereof
US9434686B2 (en) 2008-07-23 2016-09-06 President And Fellows Of Harvard College Deacetylase inhibitors and uses thereof
US9540317B2 (en) 2009-08-11 2017-01-10 President And Fellows Of Harvard College Class- and isoform-specific HDAC inhibitors and uses thereof
US10059657B2 (en) 2009-08-11 2018-08-28 President And Fellows Of Harvard College Class-and isoform-specific HDAC inhibitors and uses thereof
US8716344B2 (en) 2009-08-11 2014-05-06 President And Fellows Of Harvard College Class- and isoform-specific HDAC inhibitors and uses thereof
CN102858779A (en) * 2010-02-19 2013-01-02 派普塔德姆公司 Cyclic tetrapeptides and therapeutic applications thereof
US9908908B2 (en) 2012-08-30 2018-03-06 Jiangsu Hansoh Pharmaceutical Co., Ltd. Tenofovir prodrug and pharmaceutical uses thereof
EP3461488A1 (en) 2017-09-27 2019-04-03 Onxeo Combination of a dbait molecule and a hdac inhibitor for treating cancer
EP3461480A1 (en) 2017-09-27 2019-04-03 Onxeo Combination of a dna damage response cell cycle checkpoint inhibitors and belinostat for treating cancer
WO2019063649A1 (en) 2017-09-27 2019-04-04 Onxeo Combination of a dbait molecule and a hdac inhibitor for treating cancer
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
WO2023041805A1 (en) 2021-09-20 2023-03-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for improving the efficacy of hdac inhibitor therapy and predicting the response to treatment with hdac inhibitor
WO2023194441A1 (en) 2022-04-05 2023-10-12 Istituto Nazionale Tumori Irccs - Fondazione G. Pascale Combination of hdac inhibitors and statins for use in the treatment of pancreatic cancer

Also Published As

Publication number Publication date
AU2990597A (en) 1997-10-17
US20090221474A1 (en) 2009-09-03
WO1997035990A3 (en) 1998-03-26
US8399233B2 (en) 2013-03-19
US8329945B2 (en) 2012-12-11
US20070093413A1 (en) 2007-04-26
US6777217B1 (en) 2004-08-17
US8362084B2 (en) 2013-01-29
US20100137196A1 (en) 2010-06-03
US8329946B2 (en) 2012-12-11
US20110313045A1 (en) 2011-12-22
US7994362B2 (en) 2011-08-09
US20120302510A1 (en) 2012-11-29
US20110218154A1 (en) 2011-09-08
US20110319493A1 (en) 2011-12-29
US8426592B2 (en) 2013-04-23

Similar Documents

Publication Publication Date Title
US6777217B1 (en) Histone deacetylases, and uses related thereto
WO1997035990A9 (en) Histone deacetylases, and uses related thereto
US8895284B2 (en) Class II human histone deacetylases, and uses related thereto
US6147192A (en) Tub interactor (TI) polypeptides and uses therefor
KR100509415B1 (en) Immunosuppressive target proteins
KR100404011B1 (en) Cell-cycle regulating proteins and how to use them
WO1998004590A1 (en) Conservin compositions and therapeutic and diagnostic uses therefor
US20040077046A1 (en) Histone deacetylase-related gene and protein
KR100646074B1 (en) Caspase-8 interacting proteins
US6475778B1 (en) Differentiation enhancing factors and uses therefor
US7507801B2 (en) Adapter gene
US20050112643A1 (en) Atr-2 cell cycle checkpoint
US7396905B1 (en) Calcipressins: endogenous inhibitors of calcineurin, uses and reagents related thereto
US20020037513A1 (en) 33338, a novel human ubiquitin hydrolase-like molecule and uses thereof
US20020146711A1 (en) MMSC2 - an MMAC1 interacting protein
WO2002074976A2 (en) Isolated human ras-like proteins, nucleic acid molecules encoding these human ras-like proteins, and uses thereof
AU1738899A (en) Immunosuppressant target proteins
EP1392726A2 (en) Isolated human ras-like proteins, nucleic acid molecules encoding these human ras-like proteins, and uses thereof
EP1501871A2 (en) Isolated human proteins that show high homology to human disease proteins, nucleic acid molecules encoding these human proteins, and uses thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE HU IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
COP Corrected version of pamphlet

Free format text: PAGES 4/26, 6/26-10/26 AND 23/26-26/26, DRAWINGS, REPLACED BY NEW PAGES 4/26, 6/26-9/26 AND 22/26-26/26; PAGES 11/26-22/26, DRAWINGS, RENUMBERED AS PAGES 10/26-21/26; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 97534675

Format of ref document f/p: F

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase