US20050048607A1 - Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use - Google Patents

Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use Download PDF

Info

Publication number
US20050048607A1
US20050048607A1 US10/739,413 US73941303A US2005048607A1 US 20050048607 A1 US20050048607 A1 US 20050048607A1 US 73941303 A US73941303 A US 73941303A US 2005048607 A1 US2005048607 A1 US 2005048607A1
Authority
US
United States
Prior art keywords
alk
tgf
cells
type
receptor
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/739,413
Inventor
Kohei Miyazono
Takeshe Imamura
Peter Dijke
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
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
Priority claimed from GB929224057A external-priority patent/GB9224057D0/en
Priority claimed from GB939304677A external-priority patent/GB9304677D0/en
Priority claimed from GB939304680A external-priority patent/GB9304680D0/en
Priority claimed from GB939311047A external-priority patent/GB9311047D0/en
Priority claimed from GB939313763A external-priority patent/GB9313763D0/en
Priority claimed from GB939316099A external-priority patent/GB9316099D0/en
Priority claimed from GB939321344A external-priority patent/GB9321344D0/en
Priority claimed from US08/436,265 external-priority patent/US6316217B1/en
Priority to US10/739,413 priority Critical patent/US20050048607A1/en
Application filed by Individual filed Critical Individual
Publication of US20050048607A1 publication Critical patent/US20050048607A1/en
Priority to US11/980,023 priority patent/US20080131910A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • G01N2333/495Transforming growth factor [TGF]

Definitions

  • This invention relates to proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use.
  • TGF- ⁇ The transforming growth factor- ⁇ superfamily consists of a family of structurally-related proteins, including three different mammalian isoforms of TGF- ⁇ (TGF- ⁇ 1, ⁇ 2 and ⁇ 3), activins, inhibins, müllerian-inhibiting substance and bone morphogenic proteins (BMPs) (for reviews see Roberts and Sporn, (1990) Peptide Growth Factors and Their Receptors, Pt.1, Sporn and Roberts, eds. (Berlin: Springer-Verlag) pp 419-472; Moses et al (1990) Cell 63, 245-247).
  • the proteins of the TGF- ⁇ superfamily have a wide variety of biological activities. TGF- ⁇ acts as a growth inhibitor for many cell types and appears to play a central role in the regulation of embryonic development, tissue regeneration, immuno-regulation, as well as in fibrosis and carcinogenesis (Roberts and Sporn (199) see above).
  • Activins and inhibins were originally identified as factors which regulate secretion of follicle-stimulating hormone secretion (Vale et al (1990) Peptide Growth Factors and Their Receptors, Pt.2, Sporn and Roberts, eds. (Berlin: Springer-Verlag) pp.211-248). Activins were also shown to induce the differentiation of haematopoietic progenitor cells (Murata et al (1988) Proc. Natl. Acad. Sci. USA 85, 2434-2438; Eto et al (1987) Biochem. Biophys. Res. Commun.
  • BMPs or osteogenic proteins which induce the formation of bone and cartilage when implanted subcutaneously (Wozney et al (1988) Science 242, 1528-1534), facilitate neuronal differentiation (Paralkar et al (1992) J. Cell Biol. 119, 1721-1728) and induce monocyte chemotaxis (Cunningham et al (1992) Proc. Natl. Acad. Sci. USA 89, 11740-11744).
  • Müllerian-inhibiting substance induces regression of the Müllerian duct in the male reproductive system (Cate et al (1986) Cell 45, 685-698), and a glial cell line-derived neurotrophic factor enhances survival of midbrain dopaminergic neurons (Lin et al (1993) Science 260, 1130-1132).
  • the action of these growth factors is mediated through binding to specific cell surface receptors.
  • TGF- ⁇ receptors have been most thoroughly characterized.
  • receptor type I 53 kd
  • type II 75 kd
  • type III betaglycan
  • endoglin a homodimer of two 95 kd subunits
  • type I and type II receptors are directly involved in receptor signal transduction (Segarini et al (1989) Mol. Endo., 3, 261-272; Laiho et al (1991) J. Biol. Chem. 266, 9100-9112) and may form a heteromeric complex; the type II receptor is needed for the binding of TGF- ⁇ to the type I receptor and the type I receptor is needed for the signal transduction induced by the type II receptor (Wrana et al (1992) Cell, 71, 1003-1004).
  • the type III receptor and endoglin may have more indirect roles, possibly by facilitating the binding of ligand to type II receptors (Wang et al (1991) Cell, 67 797-805; López-Casillas et al (1993) Cell, 73 1435-1444).
  • Binding analyses with activin A and BMP4 have led to the identification of two co-existing cross-linked affinity complexes of 50-60 kDa and 70-80 kDa on responsive cells (Hino et al (1989) J. Biol. Chem. 264, 10309-10314; Mathews and Vale (1991), Cell 68, 775-785; Paralker et al (1991) Proc. Natl. Acad. Sci. USA 87, 8913-8917).
  • TGF- ⁇ receptors they are thought to be signalling receptors and have been named type I and type II receptors.
  • the cDNA for the activin type II receptor (Act RII) was the first to be cloned (Mathews and Vale (1991) Cell 65, 973-982).
  • the predicted structure of the receptor was shown to be a transmembrane protein with an intracellular serine/threonine kinase domain.
  • the activin receptor is related to the C. elegans daf-1 gene product, but the ligand is currently unknown (Georgi et al (1990) Cell 61, 635-645).
  • activin type IIB receptor activin type IIB receptor
  • activin type IIB receptor activin type IIB receptor
  • TGF- ⁇ type II receptor T ⁇ RII
  • the present invention involves the discovery of related novel peptides, including peptides having the activity of those defined herein as SEQ ID Nos. 2, 4, 8, 10, 12, 14, 16 and 18.
  • Their discovery is based on the realisation that receptor serine/threonine kinases form a new receptor family, which may include the type II receptors for other proteins in the TGF- ⁇ superfamily.
  • a protocol was designed to clone ActRII/daf I related cDNAs. This approach made use of the polymerase chain reaction (PCR), using degenerate primers based upon the amino-acid sequence similarity between kinase domains of the mouse activin type II receptor and daf-I gene products.
  • Activin receptor like kinases 1-6. These cDNAs showed an overall 33-39% sequence similarity with ActRII and TGF- ⁇ type II receptor and 40-92% sequence similarity towards each other in the kinase domains.
  • Soluble receptors according to the invention comprise at least predominantly the extracellular domain. These can be selected from the information provided herein, prepared in conventional manner, and used in any manner associated with the invention.
  • Antibodies to the peptides described herein may be raised in conventional manner. By selecting unique sequences of the peptides, antibodies having desired specificity can be obtained.
  • the antibodies may be monoclonal, prepared in known manner.
  • monoclonal antibodies to the extracellular domain are of potential value in therapy.
  • Products of the invention are useful in diagnostic methods, e.g. to determine the presence in a sample for an analyte binding therewith, such as in an antagonist assay.
  • diagnostic methods e.g. to determine the presence in a sample for an analyte binding therewith, such as in an antagonist assay.
  • Conventional techniques e.g. an enzyme-linked immunosorbent assay, may be used.
  • Products of the invention having a specific receptor activity can be used in therapy, e.g. to modulate conditions associated with activin or TGF- ⁇ activity.
  • Such conditions include fibrosis, e.g. liver cirrhosis and pulmonary fibrosis, cancer, rheumatoid arthritis and glomeronephritis.
  • FIG. 1 shows the alignment of the serine/threonine (S/T) kinase domains (I-VIII) of related receptors from transmembrane proteins, including embodiments of the present invention.
  • S/T serine/threonine
  • I-VIII serine/threonine kinase domains
  • FIGS. 2A to 2 D shows the sequences and characteristics of the respective primers used in the initial PCR reactions.
  • the nucleic acid sequences are also given as SEQ ID Nos. 19 to 22.
  • FIG. 3 is a comparison of the amino-acid sequences of human activin type II receptor (Act R-II), mouse activin type IIB receptor (Act R-IIB), human TGF- ⁇ type II receptor (T ⁇ R-II), human TGF- ⁇ type I receptor (ALK-5), human activin receptor type IA (ALK-2), and type IB (ALK-4), ALKs 1 & 3 and mouse ALK-6.
  • FIG. 4 shows, schematically, the structures for Daf-1, Act R-II, Act R-IIB, T ⁇ R-II, T ⁇ R-I/ALK-5, ALK's ⁇ 1, ⁇ 2 (Act RIA), ⁇ 3, ⁇ 4 (Act RIB) & ⁇ 6.
  • FIG. 5 shows the sequence alignment of the cysteine-rich domains of the ALKs, T ⁇ R-II, Act R-II, Act R-IIB and daf-1 receptors.
  • FIG. 6 is a comparison of kinase domains of serine/threonine kinases, showing the percentage amino-acid identity of the kinase domains.
  • FIG. 7 shows the pairwise alignment relationship between the kinase domains of the receptor serine/threonine kinases.
  • the dendrogram was generated using the Jotun-Hein alignment program (Hein (1990) Meth. Enzymol. 183, 626-645).
  • FIG. 8 depicts the phosphorylation of Smad-5 following interaction with ALK-1 but not following interaction with ALK-5.
  • Sequences 1 and 2 are the nucleotide and deduced amino-acid sequences of cDNA for hALK-1 (clone HP57).
  • Sequences 3 and 4 are the nucleotide and deduced amino-acid sequences of cDNA for hALK-2 (clone HP53).
  • Sequences 5 and 6 are the nucleotide and deduced amino-acid sequences of cDNA for hALK-3 (clone ONF5).
  • Sequences 7 and 8 the nucleotide and deduced amino-acid sequences of cDNA for hALK-4 (clone 11H8), complemented with PCR product encoding extracellular domain.
  • Sequences 9 and 10 are the nucleotide and deduced amino-acid sequences of cDNA for hALK-5 (clone EMBLA).
  • Sequences 11 and 12 are the nucleotide and deduced amino-acid sequences of cDNA for mALK-1 (clone AM6).
  • Sequences 13 and 14 are the nucleotide and deduced amino-acid sequences of cDNA for mALK-3 (clones ME-7 and ME-D).
  • Sequences 15 and 16 are the nucleotide and deduced amino-acid sequences of cDNA for mALK-4 (clone 8al).
  • Sequences 17 and 18 are the nucleotide and deduced amino-acid sequences of cDNA for mALK-6 (clone ME-6).
  • Sequence 19 (B1-S) is a sense primer, extracellular domain, cysteine-rich region, BamHI site at 5′ end, 28-mer, 64-fold degeneracy.
  • Sequence 20 (B3-S) is a sense primer, kinase domain II, BamHI site at 5′ end, 25-mer, 162-fold degeneracy.
  • Sequence 21 (B7-S) is a sense primer, kinase domain VIB, S/T kinase specific residues, BamHI site at 5′ end, 24-mer, 288-fold degeneracy.
  • Sequence 22 (E8-AS) is an anti-sense primer, kinase domain, S/T kinase-specific residues EcoRI site at 5′ end, 20-mer, 18-fold degeneracy.
  • Sequence 23 is an oligonucleotide probe.
  • Sequence 24 is a 5′ primer.
  • Sequence 25 is a 3′ primer.
  • Sequence 26 is a consensus sequence in Subdomain I.
  • Sequences 27 and 28 are novel sequence motifs in Subdomain VIB.
  • Sequence 29 is a novel sequence motif in Subdomain VIII.
  • nucleic acid sequences have been isolated, coding for a new sub-family of serine/threonine receptor kinases.
  • the term nucleic acid molecules as used herein refers to any sequence which codes for the murine, human or mammalian form, amino-acid sequences of which are presented herein. It is understood that the well known phenomenon of codon degeneracy provides for a great deal of sequence variation and all such varieties are included within the scope of this invention.
  • the nucleic acid sequences described herein may be used to clone the respective genomic DNA sequences in order to study the genes' structure and regulation.
  • the murine and human cDNA or genomic sequences can also be used to isolate the homologous genes from other mammalian species.
  • the mammalian DNA sequences can be used to study the receptors' functions in various in vitro and in vivo model systems.
  • ALK-5 cDNA As exemplified below for ALK-5 cDNA, it is also recognised that, given the sequence information provided herein, the artisan could easily combine the molecules with a pertinent promoter in a vector, so as to produce a cloning vehicle for expression of the molecule.
  • the promoter and coding molecule must be operably linked via any of the well-recognized and easily-practised methodologies for so doing.
  • the resulting vectors, as well as the isolated nucleic acid molecules themselves, may be used to transform prokaryotic cells (e.g. E. coli ), or transfect eukaryotes such as yeast ( S. cerevisiae ), PAE, COS or CHO cell lines. Other appropriate expression systems will also be apparent to the skilled artisan.
  • cDNA clones encoding the active open reading frames can be subcloned into expression vectors and transfected into eukaryotic cells, for example COS cells.
  • the transfected cells which can express the receptor can be subjected to binding assays for radioactively-labelled members of the TGF- ⁇ superfamily (TGF- ⁇ , activins, inhibins, bone morphogenic proteins and müllerian-inhibiting substances), as it may be expected that the receptors will bind members of the TGF- ⁇ superfamily.
  • biochemical or cell-based assays can be designed to identify the ligands, in tissue extracts or conditioned media, for receptors in which a ligand is not known.
  • Antibodies raised to the receptors may also be used to identify the ligands, using the immunoprecipitation of the cross-linked complexes.
  • purified receptor could be used to isolate the ligands using an affinity-based approach.
  • the determination of the expression patterns of the receptors may also aid in the isolation of the ligand.
  • ALKs activin receptor-like kinases
  • poly (A) ⁇ RNA was isolated from a human erythroleukemia cell line (HEL 92.1.7) obtained from the American Type Culture Collection (ATCC TIB 180). These cells were chosen as they have been shown to respond to both activin and TGF-S. Moreover leukaemic cells have proved to be rich sources for the cloning of novel receptor tyrosine kinases (Partanen et al (1990) Proc. Natl. Acad. Sci. USA 87, 8913-8917 and (1992) Mol. Cell. Biol. 12, 1698-1707).
  • RNA was prepared by the guanidinium isothiocyanate method (Chirgwin et al (1979) Biochemistry 18, 5294-5299). mRNA was selected using the poly-A or poly AT tract mRNA isolation kit (Promega, Madison, Wis., U.S.A.) as described by the manufacturers, or purified through an oligo (dT)-cellulose column as described by Aviv and Leder (1972) Proc. Natl. Acad. Sci. USA 69, 1408-1412.
  • the isolated mRNA was used for the synthesis of random primed (Amersham) cDNA, that was used to make a ⁇ gt10 library with 1 ⁇ 10 5 independent cDNA clones using the Riboclone cDNA synthesis system (Promega) and ⁇ gt10 in vitro packaging kit (Amersham) according to the manufacturers' procedures.
  • An amplified oligo (dT) primed human placenta ⁇ ZAPII cDNA library of 5 ⁇ 10 5 independent clones was used.
  • RNA isolated from AG1518 human foreskin fibroblasts was used to prepare a primary random primed ⁇ ZAPII cDNA library of 1.5 ⁇ 10 6 independent clones using the RiboClone cDNA synthesis system and Gigapack Gold II packaging extract (Stratagene).
  • a primary oligo (dT) primed human foreskin fibroblast ⁇ gt10 cDNA library was prepared.
  • An amplified oligo (dT) primed HEL cell ⁇ gt11 cDNA library of 1.5 ⁇ 10 6 independent clones (Poncz et al (1987) Blood 69 219-223) was used.
  • a twelve-day mouse embryo ⁇ EXIox cDNA library was obtained from Novagen (Madison, Wis., U.S.A.); a mouse placenta ⁇ ZAPII cDNA library was also used.
  • FIG. 1 shows the aligned serine/threonine kinase domains (I-VIII), of four related receptors of the TGF- ⁇ superfamily, i.e. hT ⁇ R-II, mActR-IIB, mActR-II and the daf-1 gene product, using the nomenclature of the subdomains according to Hanks et al (1988) Science 241, 45-52.
  • PCR primers were taken from regions of homology between the activin type II receptor and the daf-1 gene product, with particular emphasis on residues that confer serine/threonine specificity (see Table 2) and on residues that are shared by transmembrane kinase proteins and not by cytoplasmic kinases.
  • the primers were designed so that each primer of a PCR set had an approximately similar GC composition, and so that self complementarity and complementarity between the 3′ ends of the primer sets were avoided. Degeneracy of the primers was kept as low as possible, in particular avoiding serine, leucine and arginine residues (6 possible codons), and human codon preference was applied. Degeneracy was particularly avoided at the 3′ end as, unlike the 5′ end, where mismatches are tolerated, mismatches at the 3′ end dramatically reduce the efficiency of PCR.
  • restriction enzyme sites were included at the 5′ end of the primers, with a GC clamp, which permits efficient restriction enzyme digestion.
  • the primers utilised are shown in FIG. 2 .
  • Oligonucleotides were synthesized using Gene assembler plus (Pharmacia-LKB) according to the manufacturers instructions.
  • the mRNA prepared from HEL cells as described above was reverse-transcribed into cDNA in the presence of 50 mM Tris-HCl, pH 8.3, 8 mM MgCl 2 , 30 mM KCl, 10 mM dithiothreitol, 2 mM nucleotide triphosphates, excess oligo (dT) primers and 34 units of AMV reverse transcriptase at 42° C. for 2 hours in 40 ⁇ l of reaction volume.
  • Amplification by PCR was carried out with a 7.5% aliquot (3 ⁇ l) of the reverse-transcribed mRNA, in the presence of 10 mM Tris-HCl, pH 8.3, 50 mM KCl, 1.5 M MgCl 2 , 0.01% gelatin, 0.2 mM nucleotide triphosphates, 1 ⁇ M of both sense and antisense primers and 2.5 units of Taq polymerase (Perkin Elmer Cetus) in 100 ⁇ l reaction volume. Amplifications were performed on a thermal cycler (Perkin Elmer Cetus) using the following program: first 5 thermal cycles with denaturation for 1 minute at 94° C., annealing for 1 minute at 50° C., a 2 minute ramp to 55° C.
  • PCR products obtained were used to screen various cDNA libraries described supra. Labelling of the inserts of PCR products was performed using random priming method (Feinberg and Vogelstein (1983) Anal. Biochem, 132 6-13) using the Megaprime DNA labelling system (Amersham). The oligonucleotide derived from the sequence of the PCR product 5.2 was labelled by phosphorylation with T4 polynucleotide kinase following standard protocols (Sambrook et al, supra). Hybridization and purification of positive bacteriophages were performed using standard molecular biological techniques.
  • the double-stranded DNA clones were all sequenced using the dideoxynucleotide chain-termination method as described by Sanger et al, supra, using T7 DNA polymerase (Pharmacia-LKB) or Sequenase (U.S. Biochemical Corporation, Cleveland, Ohio, U.S.A.). Compressions of nucleotides were resolved using 7-deaza-GTP (U.S. Biochemical Corp.) DNA sequences were analyzed using the DNA STAR computer program (DNA STAR Ltd. U.K.). Analyses of the sequences obtained revealed the existence of six distinct putative receptor serine/threonine kinases which have been named ALK 1-6.
  • the longest open reading frame encodes a protein of 503 amino-acids, with high sequence similarity to receptor serine/threonine kinases (see below).
  • the first methionine codon, the putative translation start site is at nucleotide 283-285 and is preceded by an in-frame stop codon.
  • This first ATG is in a more favourable context for translation initiation (Kozak (1987) Nucl. Acids Res., 15, 8125-8148) than the second and third in-frame ATG at nucleotides 316-318 and 325-327.
  • the putative initiation codon is preceded by a 5′ untranslated sequence of 282 nucleotides that is GC-rich (80% GC), which is not uncommon for growth factor receptors (Kozak (1991) J. Cell Biol., 115, 887-903).
  • the 3′ untranslated sequence comprises 193 nucleotides and ends with a poly-A tail. No bona fide poly-A addition signal is found, but there is a sequence (AATACA), 17-22 nucleotides upstream of the poly-A tail, which may serve as a poly-A addition signal.
  • ALK-2 cDNA was cloned by screening an amplified oligo (dT) primed human placenta cDNA library with a radiolabelled insert derived from the PCR product 11.2.
  • Sequence analysis of cDNA clone HP53 revealed a sequence of 2719 nucleotides with a poly-A tail.
  • the longest open reading frame encodes a protein of 509 amino-acids.
  • the first ATG at nucleotides 104-106 agrees favourably with Kozak's consensus sequence with an A at position 3. This ATG is preceded in-frame by a stop codon.
  • the 5′ untranslated sequence is 103 nucleotides.
  • the 3′ untranslated sequence of 1089 nucleotides contains a polyadenylation signal located 9-14 nucleotides upstream from the poly-A tail.
  • the cDNA clone HP64 lacks 498 nucleotides from the 5′ end compared to HP53, but the sequence extended at the 3′ end with 190 nucleotides and poly-A tail is absent. This suggests that different polyadenylation sites occur for ALK-2. In Northern blots, however, only one transcript was detected (see below).
  • the cDNA for human ALK-3 was cloned by initially screening an oligo (dT) primed human foreskin fibroblast cDNA library with an oligonucleotide (SEQ ID No. 23) derived from the PCR product 5.2.
  • oligo (dT) primed human foreskin fibroblast cDNA library with an oligonucleotide (SEQ ID No. 23) derived from the PCR product 5.2.
  • SEQ ID No. 23 oligonucleotide
  • ONF5 The most 5′ sequence of ON11, a 540 nucleotide XbaI restriction fragment encoding a truncated kinase domain, was subsequently used to probe a random primed fibroblast cDNA library from which one cDNA clone with an insert size of 3 kb, termed ONF5, was isolated (SEQ ID No. 5). Sequence analysis of ONF5 revealed a sequence of 2932 nucleotides without a poly-A tail, suggesting that this clone was derived by internal priming. The longest open reading frame codes for a protein of 532 amino-acids.
  • the first ATG codon which is compatible with Kozak's consensus sequence (Kozak, supra), is at 310-312 nucleotides and is preceded by an in-frame stop codon.
  • the 5′ and 3′ untranslated sequences are 309 and 1027 nucleotides long, respectively.
  • ALK-4 cDNA was identified by screening a human oligo (dT) primed human erythroleukemia cDNA library with the radiolabelled insert of the PCR product 11.1 as a probe.
  • One cDNA clone, termed 11H8 was identified with an insert size of 2 kb (SEQ ID No. 7).
  • An open reading frame was found encoding a protein sequence of 383 amino-acids encoding a truncated extracellular domain with high similarity to receptor serine/threonine kinases.
  • the 3′ untranslated sequence is 818 nucleotides and does not contain a poly-A tail, suggesting that the cDNA was internally primed.
  • cDNA encoding the complete extracellular domain was obtained from HEL cells by RT-PCR with 5′ primer (SEQ ID No. 24) derived in part from sequence at translation start site of SKR-2 (a cDNA sequence deposited in GenBank data base, accesion number L10125, that is identical in part to ALK-4) and 3′ primer (SEQ ID No. 25) derived from 11H8 cDNA clone.
  • ALK-5 was identified by screening the random primed HEL cell ⁇ gt 10 cDNA library with the PCR product 11.1 as a probe. This yielded one positive clone termed EMBLA (insert size of 5.3 kb with 2 internal EcoRI sites). Nucleotide sequencing revealed an open reading frame of 1509 bp, coding for 503 amino-acids. The open reading frame was flanked by a 5′ untranslated sequence of 76 bp, and a 3′ untranslated sequence of 3.7 kb which was not completely sequenced. The nucleotide and deduced amino-acid sequences of ALK-5 are shown in SEQ ID Nos. 9 and 10.
  • the predicted amino-acid sequence of mouse ALK-3 is very similar to the human sequence; only 8 amino-acid residues differ (98% identity; see SEQ ID No. 14) and the calculated molecular mass of the primary translated product without the putative signal sequence is 57,447 Da.
  • clones obtained from the initial library screening with PCR product 11.1 four clones hybridized to the probe corresponding to the conserved kinase domain of ALK-4 but not to probes from more divergent parts of ALK-1 to -4. Analysis of these clones revealed that they have an identical sequence which differs from those of ALK-1 to -5 and was termed ALK-6.
  • the longest clone ME6 with a 2.0 kb insert was completely sequenced yielding a 1952 bp fragment consisting of an open reading frame of 1506 bp (502 amino-acids), flanked by a 5′ untranslated sequence of 186 bp, and a 3′ untranslated sequence of 160 bp.
  • the nucleotide and predicted amino-acid sequences of mouse ALK-6 are shown in SEQ ID Nos. 17 and 18.
  • No polyadenylation signal was found in the 3′ untranslated region of ME6, indicating that the cDNA was internally primed in the 3′ end.
  • Only one ATG codon was found in the 5′ part of the open reading frame, which fulfils the rules for translation initiation (Kozak, supra), and was preceded by an in-frame stop codon at nucleotides 163-165.
  • a typical hydrophobic leader sequence was not observed at the N terminus of the translated region. Since there is no ATG codon and putative hydrophobic leader sequence, this ATG codon is likely to be used as a translation initiation site.
  • the calculated molecular mass of the primary translated product with the putative signal sequence is 55,576 Da.
  • Mouse ALK-1 (clone AM6 with 1.9 kb insert) was obtained from the mouse placenta ⁇ ZAPII cDNA library using human ALK-1 cDNA as a probe (see SEQ ID No. 11).
  • Mouse ALK-4 (clone 8al with 2.3 kb insert) was also obtained from this library using human ALK-4 cDNA library as a probe (SEQ ID No. 15).
  • clones HP22, HP57, ONF1, ONF3, ONF4 and HP29 encode the same gene, ALK-1.
  • Clone AM6 encodes mouse ALK-1.
  • HP53, HP64 and HP84 encode the same gene, ALK-2.
  • ONF5, ONF2 and ON11 encode the same gene ALK-3.
  • ME-7 and ME-D encode the mouse counterpart of human ALK-3.
  • 11H8 encodes a different gene ALK-4, whilst 8al encodes the mouse equivalent.
  • EMBLA encodes ALK-5
  • ME-6 encodes ALK-6.
  • FIG. 3 The sequence alignment between the 6 ALK genes and T ⁇ R-II, mActR-II and ActR-IIB is shown in FIG. 3 . These molecules have a similar domain structure; an N-terminal predicted hydrophobic signal sequence (von Heijne (1986) Nucl. Acids Res. 14: 4683-4690) is followed by a relatively small extracellular cysteine-rich ligand binding domain, a single hydrophobic transmembrane region (Kyte & Doolittle (1982) J. Mol. Biol. 157, 105-132) and a C-terminal intracellular portion, which consists almost entirely of a kinase domain ( FIGS. 3 and 4 ).
  • the extracelluar domains of these receptors have cysteine-rich regions, but they show little sequence similarity; for example, less than 20% sequence identity is found between Daf-1, ActR-II, T ⁇ R-II and ALK-5.
  • the ALKs appear to form a subfamily as they show higher sequence similarities (15-47% identity) in their extracellular domains.
  • the extracellular domains of ALK-5 and ALK-4 have about 29% sequence identity.
  • ALK-3 and ALK-6 share a high degree of sequence similarity in their extracellular domains (46% identity).
  • ALKs-1,-2,-3 & -5 The positions of many of the cysteine residues in all receptors can be aligned, suggesting that the extracellular domains may adopt a similar structural configuration. See FIG. 5 for ALKs-1,-2,-3 & -5. Each of the ALKs (except ALK-6) has a potential N-linked glycosylation site, the position of which is conserved between ALK-1 and ALK-2, and between ALK-3, ALK-4 and ALK-5 (see FIG. 4 ).
  • the catalytic domains of kinases can be divided into 12 subdomains with stretches of conserved amino-acid residues.
  • the key motifs are found in serine/threonine kinase receptors suggesting that they are functional kinases.
  • the consensus sequence for the binding of ATP (Gly-X-Gly-X-X-Gly in subdomain I followed by a Lys residue further downstream in subdomain II) is found in all the ALKs.
  • the kinase domains of daf-1, ActR-II, and ALKs show approximately equal sequence similarity with tyrosine and serine/threonine protein kinases.
  • analysis of the amino-acid sequences in subdomains VI and VIII, which are the most useful to distinguish a specificity for phosphorylation of tyrosine residues versus serine/threonine residues indicates that these kinases are serine/threonine kinases; refer to Table 2.
  • these regions are the most divergent between family members (see FIGS. 3 and 4 ).
  • the C termini of the kinase domains of ALKs -1 to -6 are set at Ser-495, Ser-501, Ser-527, Gln-500, Gln-498 and Ser-497, respectively.
  • ALK-1, -2, -3, -4 was determined by Northern blot analysis.
  • a Northern blot filter with mRNAs from different human tissues was obtained from Clontech (Palo Alto, Calif.). The filters were hybridized with 32 P-labelled probes at 42° C. overnight in 50% formaldehyde, 5 ⁇ standard saline citrate (SSC; 1 ⁇ SSC is 50 mM sodium citrate, pH 7.0, 150 mM NaCl), 0.1% SDS, 50 mM sodium phosphate, 5 ⁇ Denhardt's solution and 0.1 mg/ml salmon sperm DNA.
  • SSC standard saline citrate
  • probes were used that did not encode part of the kinase domains, but corresponded to the highly diverged sequences of either 5′ untranslated and ligand-binding regions (probes for ALK-1, -2 and -3) or 3′ untranslated sequences (probe for ALK-4).
  • the probes were labelled by random priming using the Multiprime (or Mega-prime) DNA labelling system and [ ⁇ - 32 P] dCTP (Feinberg & Vogelstein (1983) Anal. Biochem. 132: 6-13). Unincorporated label was removed by Sephadex G-25 chromatography.
  • Filters were washed at 65° C., twice for 30 minutes in 2.5 ⁇ SSC, 0.1% SDS and twice for 30 minutes in 0.3 ⁇ SSC, 0.1% SDS before being exposed to X-ray film. Stripping of blots was performed by incubation at 90-100° C. in water for 20 minutes.
  • ALK-1 is endothelial cell specific.
  • the ALK-5 mRNA size and distribution were determined by Northern blot analysis as above.
  • An EcoR1 fragment of 980 bp of the full length ALK-5 cDNA clone, corresponding to the C-terminal part of the kinase domain and 3′ untranslated region (nucleotides 1259-2232 in SEQ ID No. 9) was used as a probe.
  • the filter was washed twice in 0.5 ⁇ SSC, 0.1% SDS at 55° C. for 15 minutes.
  • ALK-1 Using the probe for ALK-1, two transcripts of 2.2 and 4.9 kb were detected.
  • the ALK-1 expression level varied strongly between different tissues, high in placenta and lung, moderate in heart, muscle and kidney, and low (to not detectable) in brain, liver and pancreas. The relative ratios between the two transcripts were similar in most tissues; in kidney, however, there was relatively more of the 4.9 kb transcript.
  • ALK-2 By reprobing the blot with a probe for ALK-2, one transcript of 4.0 kb was detected with a ubiquitous expression pattern. Expression was detected in every tissue investigated and was highest in placenta and skeletal muscle. Subsequently the blot was reprobed for ALK-3.
  • the distribution of mRNA for mouse ALK-3 and -6 in various mouse tissues was also determined by Northern blot analysis.
  • a multiple mouse tissue blot was obtained from Clontech, Palo Alto, Calif., U.S.A.
  • the filter was hybridized as described above with probes for mouse ALK-3 and ALK-6.
  • the EcoRI-PstI restriction fragment, corresponding to nucleotides 79-1100 of ALK-3, and the SacI-HpaI fragment, corresponding to nucleotides 57-720 of ALK-6, were used as probes.
  • the filter was washed at 65° C. twice for 30 minutes in 2.5 ⁇ SSC, 0.1% SDS and twice for 30 minutes with 0.3 ⁇ SSC, 0.1% SDS and then subjected to autoradiography.
  • a 1.1 kb transcript was found only in spleen.
  • a transcript of 7.2 kb was found in brain and a weak signal was also seen in lung. No other signal was seen in the other tissues tested, i.e. heart, liver, skeletal muscle, kidney and testis.
  • transcripts of ALK-1 and ALK-3 are coded from the same gene.
  • the mechanism for generation of the different transcripts is unknown at present; they may be formed by alternative mRNA splicing, differential polyadenylation, use of different promotors, or by a combination of these events.
  • the cDNA for each ALK was subcloned into a eukaryotic expression vector and transfected into various cell types and then subjected to immunoprecipitation using a rabbit antiserum raised against a synthetic peptide corresponding to part of the intracellular juxtamembrane region. This region is divergent in sequence between the various serine/threonine kinase receptors. The following amino-acid residues were used: ALK-1 145-166 ALK-2 151-172 ALK-3 181-202 ALK-4 153-171 ALK-5 158-179 ALK-6 151-168
  • the rabbit antiserum against ALK-5 was designated VPN.
  • the peptides were synthesized with an Applied Biosystems 430A Peptide Synthesizer using t-butoxycarbonyl chemistry and purified by reversed-phase high performance liquid chromatography.
  • the peptides were coupled to keyhole limpet haemocyanin (Calbiochem-Behring) using glutaraldehyde, as described by Guillick et al (1985) EMBO J. 4, 2869-2877.
  • the coupled peptides were mixed with Freunds adjuvant and used to immunize rabbits.
  • COS-1 cells (American Type Culture Collection) and the R mutant of Mv1Lu cells (for references, see below) were cultured in Dulbecco's modified Eagle's medium containing 10% fetal bovine serum (FBS) and 100 units/ml penicillin and 50 ⁇ g 1 ml streptomycin in 5% CO 2 atmosphere at 37° C.
  • the ALK-5 cDNA (nucleotides (-76)-2232), which includes the complete coding region, was cloned in the pSV7d vector (Truett et al, (1985) DNA 4, 333-349), and used for transfection.
  • Transfection into COS-1 cells was performed by the calcium phosphate precipitation method (Wigler et al (1979) Cell 16, 777-785). Briefly, cells were seeded into 6-well cell culture plates at a density of 5 ⁇ 10 5 cells/well, and transfected the following day with 10 ⁇ g of recombinant plasmid.
  • the cells were washed with 150 mM NaCI, 25 mM Tris-HCl, pH 7.4, and then solubilized with a buffer containing 20 mM Tris-HCl, pH 7.4, 150 mM NaCl, 10 mM EDTA, 1% Triton X-100, 1% deoxycholate, 1.5% Trasylol (Bayer) and 1 mM phenylmethylsulfonylfluoride (PMSF; Sigma). After 15 minutes on ice, the cell lysates were pelleted by centrifugation, and the supernatants were then incubated with 7 ⁇ l of preimmune serum for 1.5 hours at 4° C.
  • Immune complexes were then given 50 ⁇ l of protein A-Sepharose (Pharmacia-LKB) slurry (50% packed beads in 150 mM NaCl, 20 mM Tris-HCl, pH 7.4, 0.2% Triton X-100) and incubated for 45 minutes at 4° C. The beads were spun down and washed four times with a washing buffer (20 mM Tris-HCl, pH 7.4, 500 mM NaCI, 1% Triton X-100, 1% deoxycholate and 0.2% SDS), followed by one wash in distilled water.
  • a washing buffer (20 mM Tris-HCl, pH 7.4, 500 mM NaCI, 1% Triton X-100, 1% deoxycholate and 0.2% SDS
  • the immune complexes were eluted by boiling for 5 minutes in the SDS-sample buffer (100 mM Tris-HCl, pH 8.8, 0.01% bromophenol blue, 36% glycerol, 4% SDS) in the presence of 10 mM DTT, and analyzed by SDS-gel electrophoresis using 7-15% polyacrylamide gels (Blobel and Dobberstein, (1975) J. Cell Biol. 67, 835-851). Gels were fixed, incubated with Amplify (Amersham) for 20 minutes, and subjected to fluorography. A component of 53Da was seen.
  • SDS-sample buffer 100 mM Tris-HCl, pH 8.8, 0.01% bromophenol blue, 36% glycerol, 4% SDS
  • SDS-gel electrophoresis using 7-15% polyacrylamide gels (Blobel and Dobberstein, (1975) J. Cell Biol. 67, 835-851). Gels
  • porcine aortic endothelial (PAE) cells were transfected with an expression vector containing the ALK-5 cDNA, and analyzed for the binding of 125 I-TGF- ⁇ 1.
  • PAE cells were cultured in Ham's F-12 medium supplemented with 10% FBS and antibiotics (Miyazono et al., (1988) J. Biol. Chem. 263, 6407-6415).
  • the ALK-5 cDNA was cloned into the cytomegalovirus (CMV)-based expression vector pcDNA I/NEO (Invitrogen), and transfected into PAE cells by electroporation. After 48 hours, selection was initiated by adding Geneticin (G418 sulphate; Gibco-BRL) to the culture medium at a final concentration of 0.5 mg/ml (Westermark et al., (1990) Proc. Natl. Acad. Sci. USA 87, 128-132).
  • CMV cytomegalovirus
  • pcDNA I/NEO Invitrogen
  • binding buffer phosphate-buffered saline containing 0.9 mM CaCl 2 , 0.49 mM MgCl 2 and 1 mg/ml bovine serum albumin (BSA)
  • BSA bovine serum albumin
  • the cells were harvested by the addition of 1 ml of detachment buffer (10 mM Tris-HC1, pH 7.4, 1 mM EDTA, 10% glycerol, 0.3 mM PMSF). The cells were pelleted by centrifugation, then resuspended in 50 ⁇ l of solubilization buffer (125 mM NaCl, 10 mM Tris-HCl, pH 7.4, 1 mM EDTA, 1% Triton X-100, 0.3 mM PMSF, 1% Trasylol) and incubated for 40 minutes on ice.
  • detachment buffer 10 mM Tris-HC1, pH 7.4, 1 mM EDTA, 10% glycerol, 0.3 mM PMSF.
  • solubilization buffer 125 mM NaCl, 10 mM Tris-HCl, pH 7.4, 1 mM EDTA, 1% Triton X-100, 0.3 mM PMSF, 1% Trasyl
  • 125 I-TGF- ⁇ 1 formed a 70 kDa cross-linked complex in the transfected PAE cells (PAE/T ⁇ R-I cells).
  • the size of this complex was very similar to that of the TGF- ⁇ type I receptor complex observed at lower amounts in the untransfected cells.
  • a concomitant increase of 94 kDa TGF- ⁇ type II receptor complex could also be observed in the PAE/T ⁇ R-I cells.
  • Components of 150-190 kDa which may represent crosslinked complexes between the type I and type II receptors, were also observed in the PAE/T ⁇ R-I cells.
  • the affinity cross-linking was followed by immunoprecipitation using the VPN antiserum. For this, cells in 25 cm 2 flasks were used. The supernatants obtained after cross-linking were incubated with 7 ⁇ l of preimmune serum or VPN antiserum in the presence or absence of 10 ⁇ g of peptide for 1.5 h at 4° C. Immune complexes were then added to 50 ⁇ l of protein A-Sepharose slurry and incubated for 45 minutes at 4° C.
  • the protein A-Sepharose beads were washed four times with the washing buffer, once with distilled water, and the samples were analyzed by SDS-gel electrophoresis using 4-15% polyacrylamide gradient gels and autoradiography.
  • a 70 kDa cross-linked complex was precipitated by the VPN antiserum in PAE/T ⁇ R-1 cells, and a weaker band of the same size was also seen in the untransfected cells, indicating that the untransfected PAE cells contained a low amount of endogenous ALK-5.
  • the 70 kDa complex was not observed when preimmune serum was used, or when immune serum was blocked by 10 ⁇ g of peptide.
  • a coprecipitated 94 kDa component could also be observed in the PAE/T ⁇ R-I cells.
  • the latter component is likely to represent a TGF- ⁇ type II receptor complex, since an antiserum, termed DRL, which was raised against a synthetic peptide from the C-terminal part of the TGF- ⁇ type II receptor, precipitated a 94 kDa TGF- ⁇ type II receptor complex, as well as a 70 kDa type I receptor complex from PAE/T ⁇ R-I cells.
  • the carbohydrate contents of ALK-5 and the TGF- ⁇ type II receptor were characterized by deglycosylation using endoglycosidase F as described above and analyzed by SDS-polyacrylamide gel electrophoresis and autoradiography.
  • the ALK-5 cross-linked complex shifted from 70 kDa to 66 kDa, whereas that of the type II receptor shifted from 94 kDa to 82 kDa.
  • the observed larger shift of the type II receptor band compared with that of the ALK-5 band is consistent with the deglycosylation data of the type I and type II receptors on rat liver cells reported previously (Cheifetz et al (1988) J. Biol. Chem.
  • porcine TGF- ⁇ type II receptor has two N-glycosylation sites (Lin et al (1992) Cell 68, 775-785), whereas ALK-5 has only one (see SEQ ID No. 9).
  • TGF- ⁇ 1 Binding of TGF- ⁇ 1 to the type I receptor is known to be abolished by transient treatment of the cells with dithiothreitol (DTT) (Cheifetz and Massague (1991) J. Biol. Chem. 266, 20767-20772; Wrana et al (1992) Cell 71, 1003-1014).
  • DTT dithiothreitol
  • Affinity cross-linking followed by immunoprecipitation by the VPN antiserum showed that neither the ALK-5 nor the type II receptor complexes was precipitated after DTT treatment, indicating that the VPN antiserum reacts only with ALK-5.
  • the data show that the VPN antiserum recognizes a TGF- ⁇ type I receptor, and that the type I and type II receptors form a heteromeric complex.
  • Transient expression plasmids of ALKs-1 to -6 and T ⁇ R-II were generated by subcloning into the pSV7d expression vector or into the pcDNA I expression vector (Invitrogen). Transient transfection of COS-1 cells and iodination of TGF- ⁇ 1 were carried out as described above. Crosslinking and immunoprecipitation were performed as described for PAE cells above.
  • Each one of the ALKs bound 125 I-TGF- ⁇ 1 and was coimmunoprecipitated with the T ⁇ R-II complex using the DRL antiserum. Comparison of the efficiency of the different ALKs to form heteromeric complexes with T ⁇ R-II, revealed that ALK-5 formed such complexes more efficiently than the other ALKs. The size of the crosslinked complex was larger for ALK-3 than for other ALKs, consistent with its slightly larger size.
  • ALK's are physiological type I receptors for TGF- ⁇ .
  • the mink lung epithelial cell line, Mv1Lu is widely used to provide target cells for TGF- ⁇ action and is well characterized regarding TGF- ⁇ receptors (Laiho et al (1990) J. Biol. Chem. 265, 18518-18524; Laiho et al (1991) J. Biol. Chem. 266, 9108-9112). Only the VPN antiserum efficiently precipitated both type I and type II TGF- ⁇ receptors in the wild type Mv1Lu cells.
  • the DRL antiserum also precipitated components with the same size as those precipitated by the VPN antiserum.
  • a mutant cell line (R mutant) which lacks the TGF- ⁇ type I receptor and does not respond to TGF- ⁇ (Laiho et al, supra) was also investigated by cross-linking followed by immunoprecipitation. Consistent with the results obtained by Laiho et al (1990), supra the type III and type II TGF- ⁇ receptor complexes, but not the type I receptor complex, were observed by affinity crosslinking. Crosslinking followed by immunoprecipatition using the DRL antiserum revealed only the type II receptor complex, whereas neither the type I nor type II receptor complexes was seen using the VPN antiserum.
  • the type I and type II TGF- ⁇ receptor complexes could be precipitated by the VPN and DRL antisera in other cell lines, including human foreskin fibroblasts (AG1518), human lung adenocarcinoma cells (A549), and human oral squamous cell carcinoma cells (HSC-2).
  • Affinity cross-linking studies revealed multiple TGF- ⁇ type I receptor-like complexes of 70-77 kDa in these cells. These components were less efficiently competed by excess unlabelled TGF- ⁇ 1 in HSC-2 cells.
  • the type II receptor complex was low or not detectable in A549 and HSC-2 cells.
  • ALKs could restore responsiveness to TGF- ⁇ in the R mutant of Mv1Lu cells, which lack the ligand-binding ability of the TGF- ⁇ type I receptor but have intact type II receptor.
  • Wild-type Mv1Lu cells and mutant cells were transfected with ALK cDNA and were then assayed for the production of plasminogen activator inhibitor-1 (PAI-1) which is produced as a result of TGF- ⁇ receptor activation as described previously by Laiho et al (1991) Mol. Cell Biol. 11, 972-978. Briefly, cells were added with or without 10 ng/ml of TGF- ⁇ 1 for 2 hours in serum-free MCDB 104 without methionine.
  • PAI-1 plasminogen activator inhibitor-1
  • the R mutant cells that were transfected with other ALKs did not produce PAI-1 upon the addition of TGF- ⁇ 1.
  • COS-1 cells were co-transfected as described above. Recombinant human activin A was iodinated using the chloramine T method (Mathews and Vale (1991) Cell 65, 973-982). Transfected COS-1 cells were analysed for binding and crosslinking of 125 I-activin A in the presence or absence of excess unlabelled activin A. The crosslinked complexes were subjected to immunoprecipitation using DRL antisera or specific ALK antisera.
  • ALK-2 and ALK-4 bound 125 I-activin A and were coimmunoprecipitated with ActR-II.
  • Other ALKs also bound 125 I-activin A but with a lower efficiency compared to ALK-2 and ALK-4.
  • activin responsive cells were examined for the expression of endogenous activin type I receptors.
  • Mv1Lu cells, as well as the R mutant, express both type I and type II receptors for activin, and the R mutant cells produce PAI-1 upon the addition of activin A.
  • Mv1Lu cells were labeled with 125 I-activin A, cross-linked and immunoprecipitated by the antisera against ActR-II or ALKs as described above.
  • the type I and type II receptor complexes in Mv1Lu cells were immunoprecipitated only by the antisera against ALK-2, ALK-4 and ActR-II. Similar results were obtained using the R mutant cells.
  • PAE cells do not bind activin because of the lack of type II receptors for activin, and so cells were transfected with a chimeric receptor, to enable them to bind activin, as described herein.
  • activin type I receptor complexes in PAE/Chim A cells were immunoprecipitated by the ALK-2 and ALK-4 antisera. These results show that both ALK-2 and ALK-4 serve as high affinity type I receptors for activin A in these cells.
  • ALK-1, ALK-3 and ALK-6 bind TGF- ⁇ 1 and activin A in the presence of their respective type II receptors, but the functional consequences of the binding of the ligands remains to be elucidated.
  • TGF- ⁇ family members acts as ligands in connection with specific type I and type II receptors, with resulting complexes interacting with members of the Smad family.
  • Smad molecules are homologs of molecules found in Drosophila (“Mad”), and C. elegans (Sma), hence, the acronym “Smad”. These are involved in signal transduction pathways downstream of serine/threonine kinase receptors. See Massagué et al., Trends Cell Biol. 2: 187-192 (1997). The different members of the family have different signaling roles.
  • Smad1 for example, as well as Smad 2 and 3, and perhaps Smad 5, became phosphorylated via specific type 1 serine/threonine kinase receptors, and act in pathway restricted fashion.
  • Xenopus Mad1 induces ventral mesoderm, in the presence of BMP.
  • the human Smad1 has been shown to have ventralizing activity. See Liu et al., Nature 381: 620-623 (1996); Kretzschmer et al., Genes Dev 11: 984-995 (1997). There is also some evidence that TGF- ⁇ phosphorylates Smad1. See Lechleider et al., J. Biol. Chem.
  • COS-7 cells which do not express ALK-1, were transfected with cDNA encoding tagged ALK-1.
  • the tag was hemagluttinin (hereafter “HA”), and a commercially available lipid containing transfecting agent was used.
  • porcine aortic endothelial (PAE) cells were also used, because these cells express TGF ⁇ type II receptors, and ALK-5, but not ALK-1. Hence, PAE cells were either transfected, or not. Transfection protocols are given, supra.
  • the cells were then contacted with 125 I labelled TGF- ⁇ 1, and were then contacted with ALK-1 specific antisera, to ascertain whether cross linking had occurred. See the experiments, supra, as well as ten Dijke et al., Science 264: 101-104 (1994), incorporated by reference. Antisera to ALK-5 were also used.
  • ALK-1 endogenously As well as ALK-5.
  • ALK-5 antiserum and the ALK-1 antiserum both immunoprecipitated type I and type II receptor cross linked complexes.
  • the ALK-1 antiserum immunoprecipitated band migrated slightly more slowly than the band immunprecipitated by the ALK-5 antiserum (see, e.g., FIG. 8 ). This is in agreement with the difference in size of ALK-1 and ALK-5, and it indicates that both ALK-1 and ALK-5 bind TGF- ⁇ in HUVECS.
  • ALK-1 acts as a co-called “type I” TGF- ⁇ receptor in an endogenous, physiological setting.
  • Smad1 and Smad-5 an intracellular signalling molecule which is structurally highly similar to Smad1 were phosphorylated following interaction with activated ALK-1, but not following interaction of TGF- ⁇ and ALK-5. Conversely, the interaction of TGF- ⁇ and ALK-5 led to phosphorylation of Smad 2, but not Smad 1. This supports a conclusion that ALK-1 transduces signal in a manner similar to BMPs.
  • FIG. 8 depicts the phosphorylation of Smad-5 following interaction with ALK-1 but not ALK-5. Phosphorylation of both Smad-5 and Smad1 has been shown for BMP type I receptors suggesting ALK-1 is functionally very similar to ALK3 (BMPR-IA) and (ALK6 BMPR-IB).
  • Affinity cross-linking using 125 I-TGF- ⁇ 1, and immunoprecipitation with Flag antibody was carried out, as discussed supra.
  • the expression of ALK-1 was determined using anti-HA antibody, since the vector used to express ALK-1 effectively tagged it with HA.
  • Examples show that one can identify molecule which inhibit, or enhance expression of a gene whose expression is regulated by phosphorylated Smad1.
  • ALK-1 has been identified as a key constituent of the pathway by which Smad1 is phosphorylated
  • the cells can be those which inherently express both ALK-1 and Smad1, or which have been transformed or transfected with DNA encoding one or both of these.
  • One can determine the phosphorylation via, e.g., the use of anti phosphorylated serine antibodies, as discussed supra.
  • the assay can be carried out using TGF- ⁇ , as a competing agent.
  • the TGF- ⁇ does bind to ALK-1, leading to phosphorylation of Smad1. Hence, by determining a value with TGF- ⁇ alone, one can then compare a value determined with amounts of the substance to be tested, in the presence of TGF- ⁇ . Changes in phosphorylation levels can thus be attributed to the test substance.
  • activators or agonists can also be tested for, or utilized, following the same type of procedures.
  • transcripts which are expressed or not expressed can be identified.
  • systems of expression analysis such as differential display PCR, subtraction hybridization, and other systems which combine driver and testes populations of nucleic acids, whereby transcripts which are expressed or not expressed can be identified.
  • a part of the invention is the regulation of a phosphorylation of Smad-1 or Smad-5, with inhibitors, such as antibodies against the extracellular domain of ALK-1 or TGF- ⁇ , or enhancers, such as TGF- ⁇ itself, or those portions of the TGF- ⁇ molecule which are necessary for binding. Indeed, by appropriate truncation, one can also determine what portions of ALK-1 are required for phosphorylation of Smad1 or Smad-5 to take place.

Abstract

The invention relates to the molecule referred to as ALK-1, and its role as a type I receptor for members of the TGF-β family. The molecule has a role in the phosphorylation of Smad-5 and Smad1, which also act as activators of certain genes. Aspects of the invention relate to this interaction.

Description

    FIELD OF THE INVENTION
  • This invention relates to proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use.
  • BACKGROUND OF THE INVENTION
  • The transforming growth factor-β (TGF-β) superfamily consists of a family of structurally-related proteins, including three different mammalian isoforms of TGF-β (TGF-β1, β2 and β3), activins, inhibins, müllerian-inhibiting substance and bone morphogenic proteins (BMPs) (for reviews see Roberts and Sporn, (1990) Peptide Growth Factors and Their Receptors, Pt.1, Sporn and Roberts, eds. (Berlin: Springer-Verlag) pp 419-472; Moses et al (1990) Cell 63, 245-247). The proteins of the TGF-β superfamily have a wide variety of biological activities. TGF-β acts as a growth inhibitor for many cell types and appears to play a central role in the regulation of embryonic development, tissue regeneration, immuno-regulation, as well as in fibrosis and carcinogenesis (Roberts and Sporn (199) see above).
  • Activins and inhibins were originally identified as factors which regulate secretion of follicle-stimulating hormone secretion (Vale et al (1990) Peptide Growth Factors and Their Receptors, Pt.2, Sporn and Roberts, eds. (Berlin: Springer-Verlag) pp.211-248). Activins were also shown to induce the differentiation of haematopoietic progenitor cells (Murata et al (1988) Proc. Natl. Acad. Sci. USA 85, 2434-2438; Eto et al (1987) Biochem. Biophys. Res. Commun. 142, 1095-1103) and induce mesoderm formation in Xenopus embryos (Smith et al (1990) Nature 345, 729-731; van den Eijnden-Van Raaij et al (1990) Nature 345, 732-734).
  • BMPs or osteogenic proteins which induce the formation of bone and cartilage when implanted subcutaneously (Wozney et al (1988) Science 242, 1528-1534), facilitate neuronal differentiation (Paralkar et al (1992) J. Cell Biol. 119, 1721-1728) and induce monocyte chemotaxis (Cunningham et al (1992) Proc. Natl. Acad. Sci. USA 89, 11740-11744). Müllerian-inhibiting substance induces regression of the Müllerian duct in the male reproductive system (Cate et al (1986) Cell 45, 685-698), and a glial cell line-derived neurotrophic factor enhances survival of midbrain dopaminergic neurons (Lin et al (1993) Science 260, 1130-1132). The action of these growth factors is mediated through binding to specific cell surface receptors.
  • Within this family, TGF-β receptors have been most thoroughly characterized. By covalently cross-linking radio-labelled TGF-β to cell surface molecules followed by polyacrylamide gel electrophoresis of the affinity-labelled complexes, three distinct size classes of cell surface proteins (in most cases) have been identified, denoted receptor type I (53 kd), type II (75 kd), type III or betaglycan (a 300 kd proteoglycan with a 120 kd core protein) (for a review see Massague (1992) Cell 69 1067-1070) and more recently endoglin (a homodimer of two 95 kd subunits) (Cheifetz et al (1992) J. Biol. Chem. 267 19027-19030). Current evidence suggests that type I and type II receptors are directly involved in receptor signal transduction (Segarini et al (1989) Mol. Endo., 3, 261-272; Laiho et al (1991) J. Biol. Chem. 266, 9100-9112) and may form a heteromeric complex; the type II receptor is needed for the binding of TGF-β to the type I receptor and the type I receptor is needed for the signal transduction induced by the type II receptor (Wrana et al (1992) Cell, 71, 1003-1004). The type III receptor and endoglin may have more indirect roles, possibly by facilitating the binding of ligand to type II receptors (Wang et al (1991) Cell, 67 797-805; López-Casillas et al (1993) Cell, 73 1435-1444).
  • Binding analyses with activin A and BMP4 have led to the identification of two co-existing cross-linked affinity complexes of 50-60 kDa and 70-80 kDa on responsive cells (Hino et al (1989) J. Biol. Chem. 264, 10309-10314; Mathews and Vale (1991), Cell 68, 775-785; Paralker et al (1991) Proc. Natl. Acad. Sci. USA 87, 8913-8917). By analogy with TGF-β receptors they are thought to be signalling receptors and have been named type I and type II receptors.
  • Among the type II receptors for the TGF-β superfamily of proteins, the cDNA for the activin type II receptor (Act RII) was the first to be cloned (Mathews and Vale (1991) Cell 65, 973-982). The predicted structure of the receptor was shown to be a transmembrane protein with an intracellular serine/threonine kinase domain. The activin receptor is related to the C. elegans daf-1 gene product, but the ligand is currently unknown (Georgi et al (1990) Cell 61, 635-645). Thereafter, another form of the activin type II receptor (activin type IIB receptor), of which there are different splicing variants (Mathews et al (1992), Science 225, 1702-1705; Attisano et al (1992) Cell 68, 97-108), and the TGF-β type II receptor (TβRII) (Lin et al (1992) Cell 68, 775-785) were cloned, both of which have putative serine/threonine kinase domains.
  • SUMMARY OF THE INVENTION
  • The present invention involves the discovery of related novel peptides, including peptides having the activity of those defined herein as SEQ ID Nos. 2, 4, 8, 10, 12, 14, 16 and 18. Their discovery is based on the realisation that receptor serine/threonine kinases form a new receptor family, which may include the type II receptors for other proteins in the TGF-β superfamily. To ascertain whether there were other members of this family of receptors, a protocol was designed to clone ActRII/daf I related cDNAs. This approach made use of the polymerase chain reaction (PCR), using degenerate primers based upon the amino-acid sequence similarity between kinase domains of the mouse activin type II receptor and daf-I gene products.
  • This strategy resulted in the isolation of a new family of receptor kinases called Activin receptor like kinases (ALK's) 1-6. These cDNAs showed an overall 33-39% sequence similarity with ActRII and TGF-β type II receptor and 40-92% sequence similarity towards each other in the kinase domains.
  • Soluble receptors according to the invention comprise at least predominantly the extracellular domain. These can be selected from the information provided herein, prepared in conventional manner, and used in any manner associated with the invention.
  • Antibodies to the peptides described herein may be raised in conventional manner. By selecting unique sequences of the peptides, antibodies having desired specificity can be obtained.
  • The antibodies may be monoclonal, prepared in known manner. In particular, monoclonal antibodies to the extracellular domain are of potential value in therapy.
  • Products of the invention are useful in diagnostic methods, e.g. to determine the presence in a sample for an analyte binding therewith, such as in an antagonist assay. Conventional techniques, e.g. an enzyme-linked immunosorbent assay, may be used.
  • Products of the invention having a specific receptor activity can be used in therapy, e.g. to modulate conditions associated with activin or TGF-β activity. Such conditions include fibrosis, e.g. liver cirrhosis and pulmonary fibrosis, cancer, rheumatoid arthritis and glomeronephritis.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the alignment of the serine/threonine (S/T) kinase domains (I-VIII) of related receptors from transmembrane proteins, including embodiments of the present invention. The nomenclature of the subdomains is accordingly to Hanks et al (1988).
  • FIGS. 2A to 2D shows the sequences and characteristics of the respective primers used in the initial PCR reactions. The nucleic acid sequences are also given as SEQ ID Nos. 19 to 22.
  • FIG. 3 is a comparison of the amino-acid sequences of human activin type II receptor (Act R-II), mouse activin type IIB receptor (Act R-IIB), human TGF-β type II receptor (TβR-II), human TGF-β type I receptor (ALK-5), human activin receptor type IA (ALK-2), and type IB (ALK-4), ALKs 1 & 3 and mouse ALK-6.
  • FIG. 4 shows, schematically, the structures for Daf-1, Act R-II, Act R-IIB, TβR-II, TβR-I/ALK-5, ALK's −1, −2 (Act RIA), −3, −4 (Act RIB) & −6.
  • FIG. 5 shows the sequence alignment of the cysteine-rich domains of the ALKs, TβR-II, Act R-II, Act R-IIB and daf-1 receptors.
  • FIG. 6 is a comparison of kinase domains of serine/threonine kinases, showing the percentage amino-acid identity of the kinase domains.
  • FIG. 7 shows the pairwise alignment relationship between the kinase domains of the receptor serine/threonine kinases. The dendrogram was generated using the Jotun-Hein alignment program (Hein (1990) Meth. Enzymol. 183, 626-645).
  • FIG. 8 depicts the phosphorylation of Smad-5 following interaction with ALK-1 but not following interaction with ALK-5.
  • BRIEF DESCRIPTION OF THE SEQUENCE LISTINGS
  • Sequences 1 and 2 are the nucleotide and deduced amino-acid sequences of cDNA for hALK-1 (clone HP57).
  • Sequences 3 and 4 are the nucleotide and deduced amino-acid sequences of cDNA for hALK-2 (clone HP53).
  • Sequences 5 and 6 are the nucleotide and deduced amino-acid sequences of cDNA for hALK-3 (clone ONF5).
  • Sequences 7 and 8 the nucleotide and deduced amino-acid sequences of cDNA for hALK-4 (clone 11H8), complemented with PCR product encoding extracellular domain.
  • Sequences 9 and 10 are the nucleotide and deduced amino-acid sequences of cDNA for hALK-5 (clone EMBLA).
  • Sequences 11 and 12 are the nucleotide and deduced amino-acid sequences of cDNA for mALK-1 (clone AM6).
  • Sequences 13 and 14 are the nucleotide and deduced amino-acid sequences of cDNA for mALK-3 (clones ME-7 and ME-D).
  • Sequences 15 and 16 are the nucleotide and deduced amino-acid sequences of cDNA for mALK-4 (clone 8al).
  • Sequences 17 and 18 are the nucleotide and deduced amino-acid sequences of cDNA for mALK-6 (clone ME-6).
  • Sequence 19 (B1-S) is a sense primer, extracellular domain, cysteine-rich region, BamHI site at 5′ end, 28-mer, 64-fold degeneracy.
  • Sequence 20 (B3-S) is a sense primer, kinase domain II, BamHI site at 5′ end, 25-mer, 162-fold degeneracy.
  • Sequence 21 (B7-S) is a sense primer, kinase domain VIB, S/T kinase specific residues, BamHI site at 5′ end, 24-mer, 288-fold degeneracy.
  • Sequence 22 (E8-AS) is an anti-sense primer, kinase domain, S/T kinase-specific residues EcoRI site at 5′ end, 20-mer, 18-fold degeneracy.
  • Sequence 23 is an oligonucleotide probe.
  • Sequence 24 is a 5′ primer.
  • Sequence 25 is a 3′ primer.
  • Sequence 26 is a consensus sequence in Subdomain I.
  • Sequences 27 and 28 are novel sequence motifs in Subdomain VIB.
  • Sequence 29 is a novel sequence motif in Subdomain VIII.
  • DESCRIPTION OF THE INVENTION
  • As described in more detail below, nucleic acid sequences have been isolated, coding for a new sub-family of serine/threonine receptor kinases. The term nucleic acid molecules as used herein refers to any sequence which codes for the murine, human or mammalian form, amino-acid sequences of which are presented herein. It is understood that the well known phenomenon of codon degeneracy provides for a great deal of sequence variation and all such varieties are included within the scope of this invention.
  • The nucleic acid sequences described herein may be used to clone the respective genomic DNA sequences in order to study the genes' structure and regulation. The murine and human cDNA or genomic sequences can also be used to isolate the homologous genes from other mammalian species. The mammalian DNA sequences can be used to study the receptors' functions in various in vitro and in vivo model systems.
  • As exemplified below for ALK-5 cDNA, it is also recognised that, given the sequence information provided herein, the artisan could easily combine the molecules with a pertinent promoter in a vector, so as to produce a cloning vehicle for expression of the molecule. The promoter and coding molecule must be operably linked via any of the well-recognized and easily-practised methodologies for so doing. The resulting vectors, as well as the isolated nucleic acid molecules themselves, may be used to transform prokaryotic cells (e.g. E. coli), or transfect eukaryotes such as yeast (S. cerevisiae), PAE, COS or CHO cell lines. Other appropriate expression systems will also be apparent to the skilled artisan.
  • Several methods may be used to isolate the ligands for the ALKs. As shown for ALK-5 cDNA, cDNA clones encoding the active open reading frames can be subcloned into expression vectors and transfected into eukaryotic cells, for example COS cells. The transfected cells which can express the receptor can be subjected to binding assays for radioactively-labelled members of the TGF-β superfamily (TGF-β, activins, inhibins, bone morphogenic proteins and müllerian-inhibiting substances), as it may be expected that the receptors will bind members of the TGF-β superfamily. Various biochemical or cell-based assays can be designed to identify the ligands, in tissue extracts or conditioned media, for receptors in which a ligand is not known. Antibodies raised to the receptors may also be used to identify the ligands, using the immunoprecipitation of the cross-linked complexes. Alternatively, purified receptor could be used to isolate the ligands using an affinity-based approach. The determination of the expression patterns of the receptors may also aid in the isolation of the ligand. These studies may be carried out using ALK DNA or RNA sequences as probes to perform in situ hybridisation studies.
  • The use of various model systems or structural studies should enable the rational development of specific agonists and antagonists useful in regulating receptor function. It may be envisaged that these can be peptides, mutated ligands, antibodies or other molecules able to interact with the receptors.
  • The foregoing provides examples of the invention Applicants intend to claim which includes, inter alia, isolated nucleic acid molecules coding for activin receptor-like kinases (ALKs), as defined herein. These include such sequences isolated from mammalian species such as mouse, human, rat, rabbit and monkey.
  • The following description relates to specific embodiments. It will be understood that the specification and examples are illustrative but not limitative of the present invention and that other embodiments within the spirit and scope of the invention will suggest themselves to those skilled in the art.
  • Preparation of mRNA and Construction of a cDNA Library
  • For construction of a cDNA library, poly (A) RNA was isolated from a human erythroleukemia cell line (HEL 92.1.7) obtained from the American Type Culture Collection (ATCC TIB 180). These cells were chosen as they have been shown to respond to both activin and TGF-S. Moreover leukaemic cells have proved to be rich sources for the cloning of novel receptor tyrosine kinases (Partanen et al (1990) Proc. Natl. Acad. Sci. USA 87, 8913-8917 and (1992) Mol. Cell. Biol. 12, 1698-1707). (Total) RNA was prepared by the guanidinium isothiocyanate method (Chirgwin et al (1979) Biochemistry 18, 5294-5299). mRNA was selected using the poly-A or poly AT tract mRNA isolation kit (Promega, Madison, Wis., U.S.A.) as described by the manufacturers, or purified through an oligo (dT)-cellulose column as described by Aviv and Leder (1972) Proc. Natl. Acad. Sci. USA 69, 1408-1412. The isolated mRNA was used for the synthesis of random primed (Amersham) cDNA, that was used to make a λgt10 library with 1×105 independent cDNA clones using the Riboclone cDNA synthesis system (Promega) and λgt10 in vitro packaging kit (Amersham) according to the manufacturers' procedures. An amplified oligo (dT) primed human placenta λZAPII cDNA library of 5×105 independent clones was used. Poly (A)+ RNA isolated from AG1518 human foreskin fibroblasts was used to prepare a primary random primed λZAPII cDNA library of 1.5×106 independent clones using the RiboClone cDNA synthesis system and Gigapack Gold II packaging extract (Stratagene). In addition, a primary oligo (dT) primed human foreskin fibroblast λgt10 cDNA library (Claesson-Welsh et al (1989) Proc. Natl. Acad. Sci. USA. 86 4917-4912) was prepared. An amplified oligo (dT) primed HEL cell λgt11 cDNA library of 1.5×106 independent clones (Poncz et al (1987) Blood 69 219-223) was used. A twelve-day mouse embryo λEXIox cDNA library was obtained from Novagen (Madison, Wis., U.S.A.); a mouse placenta λZAPII cDNA library was also used.
  • Generation of cDNA Probes by PCR
  • For the generation of cDNA probes by PCR (Lee et al (1988) Science 239, 1288-1291) degenerate PCR primers were constructed based upon the amino-acid sequence similarity between the mouse activin type II receptor (Mathews and Vale (1991) Cell 65, 973-982) and daf-1 (George et al (1990) Cell 61, 635-645) in the kinase domains II and VIII. FIG. 1 shows the aligned serine/threonine kinase domains (I-VIII), of four related receptors of the TGF-β superfamily, i.e. hTβR-II, mActR-IIB, mActR-II and the daf-1 gene product, using the nomenclature of the subdomains according to Hanks et al (1988) Science 241, 45-52.
  • Several considerations were applied in the design of the PCR primers. The sequences were taken from regions of homology between the activin type II receptor and the daf-1 gene product, with particular emphasis on residues that confer serine/threonine specificity (see Table 2) and on residues that are shared by transmembrane kinase proteins and not by cytoplasmic kinases. The primers were designed so that each primer of a PCR set had an approximately similar GC composition, and so that self complementarity and complementarity between the 3′ ends of the primer sets were avoided. Degeneracy of the primers was kept as low as possible, in particular avoiding serine, leucine and arginine residues (6 possible codons), and human codon preference was applied. Degeneracy was particularly avoided at the 3′ end as, unlike the 5′ end, where mismatches are tolerated, mismatches at the 3′ end dramatically reduce the efficiency of PCR.
  • In order to facilitate directional subcloning, restriction enzyme sites were included at the 5′ end of the primers, with a GC clamp, which permits efficient restriction enzyme digestion. The primers utilised are shown in FIG. 2. Oligonucleotides were synthesized using Gene assembler plus (Pharmacia-LKB) according to the manufacturers instructions.
  • The mRNA prepared from HEL cells as described above was reverse-transcribed into cDNA in the presence of 50 mM Tris-HCl, pH 8.3, 8 mM MgCl2, 30 mM KCl, 10 mM dithiothreitol, 2 mM nucleotide triphosphates, excess oligo (dT) primers and 34 units of AMV reverse transcriptase at 42° C. for 2 hours in 40 μl of reaction volume. Amplification by PCR was carried out with a 7.5% aliquot (3 μl) of the reverse-transcribed mRNA, in the presence of 10 mM Tris-HCl, pH 8.3, 50 mM KCl, 1.5 M MgCl2, 0.01% gelatin, 0.2 mM nucleotide triphosphates, 1 μM of both sense and antisense primers and 2.5 units of Taq polymerase (Perkin Elmer Cetus) in 100 μl reaction volume. Amplifications were performed on a thermal cycler (Perkin Elmer Cetus) using the following program: first 5 thermal cycles with denaturation for 1 minute at 94° C., annealing for 1 minute at 50° C., a 2 minute ramp to 55° C. and elongation for 1 minute at 72° C., followed by 20 cycles of 1 minute at 94° C., 30 seconds at 55° C. and 1 minute at 72° C. A second round of PCR was performed with 3 μl of the first reaction as a template. This involved 25 thermal cycles, each composed of 94° C. (1 min), 55° C. (0.5 min), 72° C. (1 min)
  • General procedures such as purification of nucleic acids, restriction enzyme digestion, gel electrophoresis, transfer of nucleic acid to solid supports and subcloning were performed essentially according to established procedures as described by Sambrook et al, (1989), Molecular cloning: A Laboratory Manual, 2nd Ed. Cold Spring Harbor Laboratory (Cold Spring Harbor, N.Y., USA).
  • Samples of the PCR products were digested with BamHI and EcoRI and subsequently fractionated by low melting point agarose gel electrophoresis. Bands corresponding to the approximate expected sizes, (see Table 1: ≈460 bp for primer pair B3-S and E8-AS and ≈140 bp for primer pair B7-S and E8-AS) were excised from the gel and the DNA was purified. Subsequently, these fragments were ligated into pUC19 (Yanisch-Perron et al (1985) Gene 33, 103-119), which had been previously linearised with BamHI and EcoR1 and transformed into E. coli strain DH5α using standard protocols (Sambrook et al, supra). Individual clones were sequenced using standard double-stranded sequencing techniques and the dideoxynucleotide chain termination method as described by Sanger et al (1977) Proc. Natl. Acad. Sci. USA 74, 5463-5467, and T7 DNA polymerase.
  • Employing Reverse Transcriptase PCR on HEL mRNA with the primer pair B3-S and E8-AS, three PCR products were obtained, termed 11.1, 11.2 and 11.3, that corresponded to novel genes. Using the primer pair B7-S and E8-AS, an additional novel PCR product was obtained termed 5.2.
    TABLE 1
    SEQUENCE
    SIZE OF DNA IDENTITY
    FRAGMENT IN SEQUENCE BETWEEN
    mActRII/ IDENTITY WITH mActRII
    NAME INSERT hTβRII SEQUENCE and
    OF PCR SIZE CLONES mActRII/hTβRII TβR-II
    PRODUCT PRIMERS (bp) (bp) (%) (%)
    11.1 B3-S/E8-AS 460 460 46/40 42
    11.2 B3-S/E8-AS 460 460 49/44 47
    11.3 B3-S/E8-AS 460 460 44/36 48
    11.29 B3-S/E8-AS 460 460 ND/100 ND
     9.2 B1-S/E8-AS 800 795 100/ND ND
     5.2 B7-S/E8-AS 140 143 40/38 60

    Isolation of cDNA Clones
  • The PCR products obtained were used to screen various cDNA libraries described supra. Labelling of the inserts of PCR products was performed using random priming method (Feinberg and Vogelstein (1983) Anal. Biochem, 132 6-13) using the Megaprime DNA labelling system (Amersham). The oligonucleotide derived from the sequence of the PCR product 5.2 was labelled by phosphorylation with T4 polynucleotide kinase following standard protocols (Sambrook et al, supra). Hybridization and purification of positive bacteriophages were performed using standard molecular biological techniques.
  • The double-stranded DNA clones were all sequenced using the dideoxynucleotide chain-termination method as described by Sanger et al, supra, using T7 DNA polymerase (Pharmacia-LKB) or Sequenase (U.S. Biochemical Corporation, Cleveland, Ohio, U.S.A.). Compressions of nucleotides were resolved using 7-deaza-GTP (U.S. Biochemical Corp.) DNA sequences were analyzed using the DNA STAR computer program (DNA STAR Ltd. U.K.). Analyses of the sequences obtained revealed the existence of six distinct putative receptor serine/threonine kinases which have been named ALK 1-6.
  • To clone cDNA for ALK-1 the oligo (dT) primed human placenta cDNA library was screened with a radiolabelled insert derived from the PCR product 11.3; based upon their restriction enzyme digestion patternS, three different types of clones with approximate insert sizes. of 1.7 kb, 2 kb & 3.5 kb were identified. The 2 kb clone, named HP57, was chosen as representative of this class and subjected to complete sequencing. Sequence analysis of ALK-1 revealed a sequence of 1984 nucleotides including a poly-A tail (SEQ ID No. 1). The longest open reading frame encodes a protein of 503 amino-acids, with high sequence similarity to receptor serine/threonine kinases (see below). The first methionine codon, the putative translation start site, is at nucleotide 283-285 and is preceded by an in-frame stop codon. This first ATG is in a more favourable context for translation initiation (Kozak (1987) Nucl. Acids Res., 15, 8125-8148) than the second and third in-frame ATG at nucleotides 316-318 and 325-327. The putative initiation codon is preceded by a 5′ untranslated sequence of 282 nucleotides that is GC-rich (80% GC), which is not uncommon for growth factor receptors (Kozak (1991) J. Cell Biol., 115, 887-903). The 3′ untranslated sequence comprises 193 nucleotides and ends with a poly-A tail. No bona fide poly-A addition signal is found, but there is a sequence (AATACA), 17-22 nucleotides upstream of the poly-A tail, which may serve as a poly-A addition signal.
  • ALK-2 cDNA was cloned by screening an amplified oligo (dT) primed human placenta cDNA library with a radiolabelled insert derived from the PCR product 11.2. Two clones, termed HP53 and HP64, with insert sizes of 2.7 kb and 2.4 kb respectively, were identified and their sequences were determined. No sequence difference in the overlapping clones was found, suggesting they are both derived from transcripts of the same gene.
  • Sequence analysis of cDNA clone HP53 (SEQ ID No. 3) revealed a sequence of 2719 nucleotides with a poly-A tail. The longest open reading frame encodes a protein of 509 amino-acids. The first ATG at nucleotides 104-106 agrees favourably with Kozak's consensus sequence with an A at position 3. This ATG is preceded in-frame by a stop codon. There are four ATG codons in close proximity further downstream, which agree with the Kozak's consensus sequence (Kozak, supra), but according to Kozak's scanning model the first ATG is predicted to be the translation start site. The 5′ untranslated sequence is 103 nucleotides. The 3′ untranslated sequence of 1089 nucleotides contains a polyadenylation signal located 9-14 nucleotides upstream from the poly-A tail. The cDNA clone HP64 lacks 498 nucleotides from the 5′ end compared to HP53, but the sequence extended at the 3′ end with 190 nucleotides and poly-A tail is absent. This suggests that different polyadenylation sites occur for ALK-2. In Northern blots, however, only one transcript was detected (see below).
  • The cDNA for human ALK-3 was cloned by initially screening an oligo (dT) primed human foreskin fibroblast cDNA library with an oligonucleotide (SEQ ID No. 23) derived from the PCR product 5.2. One positive cDNA clone with an insert size of 3 kb, termed ON11, was identified. However, upon partial sequencing, it appeared that this clone was incomplete; it encodes only part of the kinase domain and lacks the extracelluar domain. The most 5′ sequence of ON11, a 540 nucleotide XbaI restriction fragment encoding a truncated kinase domain, was subsequently used to probe a random primed fibroblast cDNA library from which one cDNA clone with an insert size of 3 kb, termed ONF5, was isolated (SEQ ID No. 5). Sequence analysis of ONF5 revealed a sequence of 2932 nucleotides without a poly-A tail, suggesting that this clone was derived by internal priming. The longest open reading frame codes for a protein of 532 amino-acids. The first ATG codon which is compatible with Kozak's consensus sequence (Kozak, supra), is at 310-312 nucleotides and is preceded by an in-frame stop codon. The 5′ and 3′ untranslated sequences are 309 and 1027 nucleotides long, respectively.
  • ALK-4 cDNA was identified by screening a human oligo (dT) primed human erythroleukemia cDNA library with the radiolabelled insert of the PCR product 11.1 as a probe. One cDNA clone, termed 11H8, was identified with an insert size of 2 kb (SEQ ID No. 7). An open reading frame was found encoding a protein sequence of 383 amino-acids encoding a truncated extracellular domain with high similarity to receptor serine/threonine kinases. The 3′ untranslated sequence is 818 nucleotides and does not contain a poly-A tail, suggesting that the cDNA was internally primed. cDNA encoding the complete extracellular domain (nucleotides 1-366) was obtained from HEL cells by RT-PCR with 5′ primer (SEQ ID No. 24) derived in part from sequence at translation start site of SKR-2 (a cDNA sequence deposited in GenBank data base, accesion number L10125, that is identical in part to ALK-4) and 3′ primer (SEQ ID No. 25) derived from 11H8 cDNA clone.
  • ALK-5 was identified by screening the random primed HEL cell λgt 10 cDNA library with the PCR product 11.1 as a probe. This yielded one positive clone termed EMBLA (insert size of 5.3 kb with 2 internal EcoRI sites). Nucleotide sequencing revealed an open reading frame of 1509 bp, coding for 503 amino-acids. The open reading frame was flanked by a 5′ untranslated sequence of 76 bp, and a 3′ untranslated sequence of 3.7 kb which was not completely sequenced. The nucleotide and deduced amino-acid sequences of ALK-5 are shown in SEQ ID Nos. 9 and 10. In the 5′ part of the open reading frame, only one ATG codon was found; this codon fulfils the rules of translation initiation (Kozak, supra). An in-frame stop codon was found at nucleotides (−54)−(−52) in the 5′ untranslated region. The predicted ATG start codon is followed by a stretch of hydrophobic amino-acid residues which has characteristics of a cleavable signal sequence. Therefore, the first ATG codon is likely to be used as a translation initiation site. A preferred cleavage site for the signal peptidase, according to von Heijne (1986) Nucl. Acid. Res. 14, 4683-4690, is located between amino-acid residues 24 and 25. The calculated molecular mass of the primary translated product of the ALK-5 without signal sequence is 53,646 Da.
  • Screening of the mouse embryo λEX Iox cDNA library using PCR, product 11.1 as a probe yielded 20 positive clones. DNAs from the positive clones obtained from this library were digested with EcoRI and HindIII, electrophoretically separated on a 1.3% agarose gel and transferred to nitrocellulose filters according to established procedures as described by Sambrook et al, supra. The filters were then hybridized with specific probes for human ALK-1 (nucleotide 288-670), ALK-2 (nucleotide 1-581), ALK-3 (nucleotide 79-824) or ALK-4 nucleotide 1178-1967). Such analyses revealed that a clone termed ME-7 hybridised with the human ALK-3 probe. However, nucleotide sequencing revealed that this clone was incomplete, and lacked the 5′ part of the translated region. Screening the same cDNA library with a probe corresponding to the extracelluar domain of human ALK-3 (nucleotides 79-824) revealed the clone ME-D. This clone was isolated and the sequence was analyzed. Although this clone was incomplete in the 3′ end of the translated region, ME-7 and ME-D overlapped and together covered the complete sequence of mouse ALK-3. The predicted amino-acid sequence of mouse ALK-3 is very similar to the human sequence; only 8 amino-acid residues differ (98% identity; see SEQ ID No. 14) and the calculated molecular mass of the primary translated product without the putative signal sequence is 57,447 Da.
  • Of the clones obtained from the initial library screening with PCR product 11.1, four clones hybridized to the probe corresponding to the conserved kinase domain of ALK-4 but not to probes from more divergent parts of ALK-1 to -4. Analysis of these clones revealed that they have an identical sequence which differs from those of ALK-1 to -5 and was termed ALK-6. The longest clone ME6 with a 2.0 kb insert was completely sequenced yielding a 1952 bp fragment consisting of an open reading frame of 1506 bp (502 amino-acids), flanked by a 5′ untranslated sequence of 186 bp, and a 3′ untranslated sequence of 160 bp. The nucleotide and predicted amino-acid sequences of mouse ALK-6 are shown in SEQ ID Nos. 17 and 18. No polyadenylation signal was found in the 3′ untranslated region of ME6, indicating that the cDNA was internally primed in the 3′ end. Only one ATG codon was found in the 5′ part of the open reading frame, which fulfils the rules for translation initiation (Kozak, supra), and was preceded by an in-frame stop codon at nucleotides 163-165. However, a typical hydrophobic leader sequence was not observed at the N terminus of the translated region. Since there is no ATG codon and putative hydrophobic leader sequence, this ATG codon is likely to be used as a translation initiation site. The calculated molecular mass of the primary translated product with the putative signal sequence is 55,576 Da.
  • Mouse ALK-1 (clone AM6 with 1.9 kb insert) was obtained from the mouse placenta λZAPII cDNA library using human ALK-1 cDNA as a probe (see SEQ ID No. 11). Mouse ALK-4 (clone 8al with 2.3 kb insert) was also obtained from this library using human ALK-4 cDNA library as a probe (SEQ ID No. 15).
  • To summarise, clones HP22, HP57, ONF1, ONF3, ONF4 and HP29 encode the same gene, ALK-1. Clone AM6 encodes mouse ALK-1. HP53, HP64 and HP84 encode the same gene, ALK-2. ONF5, ONF2 and ON11 encode the same gene ALK-3. ME-7 and ME-D encode the mouse counterpart of human ALK-3. 11H8 encodes a different gene ALK-4, whilst 8al encodes the mouse equivalent. EMBLA encodes ALK-5, and ME-6 encodes ALK-6.
  • The sequence alignment between the 6 ALK genes and TβR-II, mActR-II and ActR-IIB is shown in FIG. 3. These molecules have a similar domain structure; an N-terminal predicted hydrophobic signal sequence (von Heijne (1986) Nucl. Acids Res. 14: 4683-4690) is followed by a relatively small extracellular cysteine-rich ligand binding domain, a single hydrophobic transmembrane region (Kyte & Doolittle (1982) J. Mol. Biol. 157, 105-132) and a C-terminal intracellular portion, which consists almost entirely of a kinase domain (FIGS. 3 and 4).
  • The extracelluar domains of these receptors have cysteine-rich regions, but they show little sequence similarity; for example, less than 20% sequence identity is found between Daf-1, ActR-II, TβR-II and ALK-5. The ALKs appear to form a subfamily as they show higher sequence similarities (15-47% identity) in their extracellular domains. The extracellular domains of ALK-5 and ALK-4 have about 29% sequence identity. In addition, ALK-3 and ALK-6 share a high degree of sequence similarity in their extracellular domains (46% identity).
  • The positions of many of the cysteine residues in all receptors can be aligned, suggesting that the extracellular domains may adopt a similar structural configuration. See FIG. 5 for ALKs-1,-2,-3 & -5. Each of the ALKs (except ALK-6) has a potential N-linked glycosylation site, the position of which is conserved between ALK-1 and ALK-2, and between ALK-3, ALK-4 and ALK-5 (see FIG. 4).
  • The sequence similarities in the kinase domains between daf-1, ActR-II, TβR-II and ALK-5 are approximately 40%, whereas the sequence similarity between the ALKs 1 to 6 is higher (between 59% and 90%; see FIG. 6). Pairwise comparison using the Jutun-Hein sequence alignment program (Hein (1990) Meth, Enzymol., 183, 626-645), between all family members, identifies the ALKs as a separate subclass among serine/threonine kinases (FIG. 7).
  • The catalytic domains of kinases can be divided into 12 subdomains with stretches of conserved amino-acid residues. The key motifs are found in serine/threonine kinase receptors suggesting that they are functional kinases. The consensus sequence for the binding of ATP (Gly-X-Gly-X-X-Gly in subdomain I followed by a Lys residue further downstream in subdomain II) is found in all the ALKs.
  • The kinase domains of daf-1, ActR-II, and ALKs show approximately equal sequence similarity with tyrosine and serine/threonine protein kinases. However analysis of the amino-acid sequences in subdomains VI and VIII, which are the most useful to distinguish a specificity for phosphorylation of tyrosine residues versus serine/threonine residues (Hanks et al (1988) Science 241 42-52) indicates that these kinases are serine/threonine kinases; refer to Table 2.
    TABLE 2
    SUBDOMAINS
    KINASE VIB VIII
    Serine/threonine kinase consensus DLKPEN G (T/S) XX
    (Y/F) X
    Tyrosine kinase consensus DLAARN XP (I/V)
    (K/R) W
    (T/M)
    Act R-II DIKSKN GTRRYM
    Act R-IIB DFKSKN GTRRYM
    TβR-II DLKSSN GTARYM
    ALK-I DFKSRN GTKRYM
    ALK −2, −3, −4, −5, & −6 DLKSKN GTKRYM
  • The sequence motifs DLKSKN (Subdomain VIB) and GTKRYM (Subdomain VIII), that are found in most of the serine/threonine kinase receptors, agree well with the consensus sequences for all protein serine/threonine kinase receptors in these regions. In addition, these receptors, except for ALK-1, do not have a tyrosine residue surrounded by acidic residues between subdomains VII and VIII, which is common for tyrosine kinases. A unique characteristic of the members of the ALK serine/threonine kinase receptor family is the presence of two short inserts in the kinase domain between subdomains VIA and VIB and between subdomains X and XI. In the intracellular domain, these regions, together with the juxtamembrane part and C-terminal tail, are the most divergent between family members (see FIGS. 3 and 4). Based on the sequence similarity with the type II receptors for TGF-β and activin, the C termini of the kinase domains of ALKs -1 to -6 are set at Ser-495, Ser-501, Ser-527, Gln-500, Gln-498 and Ser-497, respectively.
  • mRNA Expression
  • The distribution of ALK-1, -2, -3, -4 was determined by Northern blot analysis. A Northern blot filter with mRNAs from different human tissues was obtained from Clontech (Palo Alto, Calif.). The filters were hybridized with 32P-labelled probes at 42° C. overnight in 50% formaldehyde, 5× standard saline citrate (SSC; 1×SSC is 50 mM sodium citrate, pH 7.0, 150 mM NaCl), 0.1% SDS, 50 mM sodium phosphate, 5× Denhardt's solution and 0.1 mg/ml salmon sperm DNA. In order to minimize cross-hybridization, probes were used that did not encode part of the kinase domains, but corresponded to the highly diverged sequences of either 5′ untranslated and ligand-binding regions (probes for ALK-1, -2 and -3) or 3′ untranslated sequences (probe for ALK-4). The probes were labelled by random priming using the Multiprime (or Mega-prime) DNA labelling system and [α-32P] dCTP (Feinberg & Vogelstein (1983) Anal. Biochem. 132: 6-13). Unincorporated label was removed by Sephadex G-25 chromatography. Filters were washed at 65° C., twice for 30 minutes in 2.5×SSC, 0.1% SDS and twice for 30 minutes in 0.3×SSC, 0.1% SDS before being exposed to X-ray film. Stripping of blots was performed by incubation at 90-100° C. in water for 20 minutes.
  • Our further analysis suggest ALK-1 is endothelial cell specific.
  • The ALK-5 mRNA size and distribution were determined by Northern blot analysis as above. An EcoR1 fragment of 980 bp of the full length ALK-5 cDNA clone, corresponding to the C-terminal part of the kinase domain and 3′ untranslated region (nucleotides 1259-2232 in SEQ ID No. 9) was used as a probe. The filter was washed twice in 0.5×SSC, 0.1% SDS at 55° C. for 15 minutes.
  • Using the probe for ALK-1, two transcripts of 2.2 and 4.9 kb were detected. The ALK-1 expression level varied strongly between different tissues, high in placenta and lung, moderate in heart, muscle and kidney, and low (to not detectable) in brain, liver and pancreas. The relative ratios between the two transcripts were similar in most tissues; in kidney, however, there was relatively more of the 4.9 kb transcript. By reprobing the blot with a probe for ALK-2, one transcript of 4.0 kb was detected with a ubiquitous expression pattern. Expression was detected in every tissue investigated and was highest in placenta and skeletal muscle. Subsequently the blot was reprobed for ALK-3. One major transcript of 4.4 kb and a minor transcript of 7.9 kb were detected. Expression was high in skeletal muscle, in which also an additional minor transcript of 10 kb was observed. Moderate levels of ALK-3 mRNA were detected in heart, placenta, kidney and pancreas, and low (to not detectable) expression was found in brain, lung and liver. The relative ratios between the different transcripts were similar in the tested tissues, the 4.4 kb transcript being the predominant one, with the exception for brain where both transcripts were expressed at a similar level. Probing the blot with ALK-4 indicated the presence of a transcript with the estimated size of 5.2 kb and revealed an ubiquitous expression pattern. The results of Northern blot analysis using the probe for ALK-5 showed that a 5.5 kb transcript is expressed in all human tissues tested, being most abundant in placenta and least abundant in brain and heart.
  • The distribution of mRNA for mouse ALK-3 and -6 in various mouse tissues was also determined by Northern blot analysis. A multiple mouse tissue blot was obtained from Clontech, Palo Alto, Calif., U.S.A. The filter was hybridized as described above with probes for mouse ALK-3 and ALK-6. The EcoRI-PstI restriction fragment, corresponding to nucleotides 79-1100 of ALK-3, and the SacI-HpaI fragment, corresponding to nucleotides 57-720 of ALK-6, were used as probes. The filter was washed at 65° C. twice for 30 minutes in 2.5×SSC, 0.1% SDS and twice for 30 minutes with 0.3×SSC, 0.1% SDS and then subjected to autoradiography.
  • Using the probe for mouse ALK-3, a 1.1 kb transcript was found only in spleen. By reprobing the blot with the ALK-6 specific probe, a transcript of 7.2 kb was found in brain and a weak signal was also seen in lung. No other signal was seen in the other tissues tested, i.e. heart, liver, skeletal muscle, kidney and testis.
  • All detected transcript sizes were different, and thus no cross-reaction between mRNAs for the different ALKs was observed when the specific probes were used. This suggests that the multiple transcripts of ALK-1 and ALK-3 are coded from the same gene. The mechanism for generation of the different transcripts is unknown at present; they may be formed by alternative mRNA splicing, differential polyadenylation, use of different promotors, or by a combination of these events. Differences in mRNA splicing in the regions coding for the extracellular domains may lead to the synthesis of receptors with different affinities for ligands, as was shown for mActR-IIB (Attisano et al (1992) Cell 68, 97-108) or to the production of soluble binding protein.
  • The above experiments describe the isolation of nucleic acid sequences coding for new family of human receptor kinases. The cDNA for ALK-5 was then used to determine the encoded protein size and binding properties.
  • Properties of the ALKs cDNA Encoded Proteins
  • To study the properties of the proteins encoded by the different ALK cDNAs, the cDNA for each ALK was subcloned into a eukaryotic expression vector and transfected into various cell types and then subjected to immunoprecipitation using a rabbit antiserum raised against a synthetic peptide corresponding to part of the intracellular juxtamembrane region. This region is divergent in sequence between the various serine/threonine kinase receptors. The following amino-acid residues were used:
    ALK-1 145-166
    ALK-2 151-172
    ALK-3 181-202
    ALK-4 153-171
    ALK-5 158-179
    ALK-6 151-168
  • The rabbit antiserum against ALK-5 was designated VPN.
  • The peptides were synthesized with an Applied Biosystems 430A Peptide Synthesizer using t-butoxycarbonyl chemistry and purified by reversed-phase high performance liquid chromatography. The peptides were coupled to keyhole limpet haemocyanin (Calbiochem-Behring) using glutaraldehyde, as described by Guillick et al (1985) EMBO J. 4, 2869-2877. The coupled peptides were mixed with Freunds adjuvant and used to immunize rabbits.
  • Transient Transfection of the ALK-5 cDNA
  • COS-1 cells (American Type Culture Collection) and the R mutant of Mv1Lu cells (for references, see below) were cultured in Dulbecco's modified Eagle's medium containing 10% fetal bovine serum (FBS) and 100 units/ml penicillin and 50 μg 1 ml streptomycin in 5% CO2 atmosphere at 37° C. The ALK-5 cDNA (nucleotides (-76)-2232), which includes the complete coding region, was cloned in the pSV7d vector (Truett et al, (1985) DNA 4, 333-349), and used for transfection. Transfection into COS-1 cells was performed by the calcium phosphate precipitation method (Wigler et al (1979) Cell 16, 777-785). Briefly, cells were seeded into 6-well cell culture plates at a density of 5×105 cells/well, and transfected the following day with 10 μg of recombinant plasmid. After overnight incubation, cells were washed three times with a buffer containing 25 mM Tris-HCl, pH 7.4, 138 mM NaCl, 5 mM KCl, 0.7 mM CaCl2, 0.5 mM MgCl2 and 0.6 mM Na2HPO4, and then incubated with Dulbecco's modified Eagle's medium containing FBS and antibiotics. Two days after transfection, the cells were metabolically labelled by incubating the cells for 6 hours in methionine and cysteine-free MCDB 104 medium with 150 μCi/ml of [35S]-methionine and [35S]-cysteine (in vivo labelling mix; Amersham). After labelling, the cells were washed with 150 mM NaCI, 25 mM Tris-HCl, pH 7.4, and then solubilized with a buffer containing 20 mM Tris-HCl, pH 7.4, 150 mM NaCl, 10 mM EDTA, 1% Triton X-100, 1% deoxycholate, 1.5% Trasylol (Bayer) and 1 mM phenylmethylsulfonylfluoride (PMSF; Sigma). After 15 minutes on ice, the cell lysates were pelleted by centrifugation, and the supernatants were then incubated with 7 μl of preimmune serum for 1.5 hours at 4° C. Samples were then given 50 μl of protein A-Sepharose (Pharmacia-LKB) slurry (50% packed beads in 150 mM NaCl, 20 mM Tris-HCl, pH 7.4, 0.2% Triton X100) and incubated for 45 minutes at 4° C. The beads were spun down by centrifugation, and the supernatants (1 ml) were then incubated with either 7 μl of preimmune serum or the VPN antiserum for 1.5 hours at 4° C. For blocking, 10 μg of peptide was added together with the antiserum. Immune complexes were then given 50 μl of protein A-Sepharose (Pharmacia-LKB) slurry (50% packed beads in 150 mM NaCl, 20 mM Tris-HCl, pH 7.4, 0.2% Triton X-100) and incubated for 45 minutes at 4° C. The beads were spun down and washed four times with a washing buffer (20 mM Tris-HCl, pH 7.4, 500 mM NaCI, 1% Triton X-100, 1% deoxycholate and 0.2% SDS), followed by one wash in distilled water. The immune complexes were eluted by boiling for 5 minutes in the SDS-sample buffer (100 mM Tris-HCl, pH 8.8, 0.01% bromophenol blue, 36% glycerol, 4% SDS) in the presence of 10 mM DTT, and analyzed by SDS-gel electrophoresis using 7-15% polyacrylamide gels (Blobel and Dobberstein, (1975) J. Cell Biol. 67, 835-851). Gels were fixed, incubated with Amplify (Amersham) for 20 minutes, and subjected to fluorography. A component of 53Da was seen. This component was not seen when preimmune serum was used, or when 10 μg blocking peptide was added together with the antiserum. Moreover, it was not detectable in samples derived from untransfected COS-1 cells using either preimmune serum or the antiserum.
  • Digestion with Endoglycosidase F
  • Samples immunoprecipitated with the VPN antisera obtained as described above were incubated with 0.5 U of endoglycosidase F (Boehringer Mannheim Biochemica) in a buffer containing 100 mM sodium phosphate, pH 6.1, 50 mM EDTA, 1% Triton X-100, 0.1% SDS and 1% β-mercaptoethanol at 37° C. for 24 hours. Samples were eluted by boiling for 5 minutes in the SDS-sample buffer, and analyzed by SDS-polyacrylamide gel electrophoresis as described above. Hydrolysis of N-linked carbohydrates by endoglycosidase F shifted the 53 kDa band to 51 kDa. The extracelluar domain of ALK-5 contains one potential acceptor site for N-glycosylation and the size of the deglycosylated protein is close to the predicted size of the core protein.
  • Establishment of PAE Cell Lines Expressing ALK-5
  • In order to investigate whether the ALK-5 cDNA encodes a receptor for TGF-β, porcine aortic endothelial (PAE) cells were transfected with an expression vector containing the ALK-5 cDNA, and analyzed for the binding of 125I-TGF-β1.
  • PAE cells were cultured in Ham's F-12 medium supplemented with 10% FBS and antibiotics (Miyazono et al., (1988) J. Biol. Chem. 263, 6407-6415). The ALK-5 cDNA was cloned into the cytomegalovirus (CMV)-based expression vector pcDNA I/NEO (Invitrogen), and transfected into PAE cells by electroporation. After 48 hours, selection was initiated by adding Geneticin (G418 sulphate; Gibco-BRL) to the culture medium at a final concentration of 0.5 mg/ml (Westermark et al., (1990) Proc. Natl. Acad. Sci. USA 87, 128-132). Several clones were obtained, and after analysis by immunoprecipitation using the VPN antiserum, one clone denoted PAE/TβR-1 was chosen and further analyzed.
  • Iodination of TGF-β1, Binding and Affinity Crosslinking
  • Recombinant human TGF-β1 was iodinated using the chloramine T method according to Frolik et al., (1984) J. Biol. Chem. 259, 10995-11000. Cross-linking experiments were performed as previously described (Ichijo et al., (1990) Exp. Cell Res. 187, 263-269). Briefly, cells in 6-well plates were washed with binding buffer (phosphate-buffered saline containing 0.9 mM CaCl2, 0.49 mM MgCl2 and 1 mg/ml bovine serum albumin (BSA)), and incubated on ice in the same buffer with 125I-TGF-β1 in the presence or absence of excess unlabelled TGF-β1 for 3 hours. Cells were washed and cross-linking was done in the binding buffer without BSA together with 0.28 mM disuccinimidyl suberate (DSS; Pierce Chemical Co.) for 15 minutes on ice. The cells were harvested by the addition of 1 ml of detachment buffer (10 mM Tris-HC1, pH 7.4, 1 mM EDTA, 10% glycerol, 0.3 mM PMSF). The cells were pelleted by centrifugation, then resuspended in 50 μl of solubilization buffer (125 mM NaCl, 10 mM Tris-HCl, pH 7.4, 1 mM EDTA, 1% Triton X-100, 0.3 mM PMSF, 1% Trasylol) and incubated for 40 minutes on ice. Cells were centrifuged again and supernatants were subjected to analysis by SDS-gel electrophoresis using 4-15% polyacrylamide gels, followed by autoradiography. 125I-TGF-β1 formed a 70 kDa cross-linked complex in the transfected PAE cells (PAE/TβR-I cells). The size of this complex was very similar to that of the TGF-β type I receptor complex observed at lower amounts in the untransfected cells. A concomitant increase of 94 kDa TGF-β type II receptor complex could also be observed in the PAE/TβR-I cells. Components of 150-190 kDa, which may represent crosslinked complexes between the type I and type II receptors, were also observed in the PAE/TβR-I cells.
  • In order to determine whether the cross-linked 70 kDa complex contained the protein encoded by the ALK-5 cDNA, the affinity cross-linking was followed by immunoprecipitation using the VPN antiserum. For this, cells in 25 cm2 flasks were used. The supernatants obtained after cross-linking were incubated with 7 μl of preimmune serum or VPN antiserum in the presence or absence of 10 μg of peptide for 1.5 h at 4° C. Immune complexes were then added to 50 μl of protein A-Sepharose slurry and incubated for 45 minutes at 4° C. The protein A-Sepharose beads were washed four times with the washing buffer, once with distilled water, and the samples were analyzed by SDS-gel electrophoresis using 4-15% polyacrylamide gradient gels and autoradiography. A 70 kDa cross-linked complex was precipitated by the VPN antiserum in PAE/TβR-1 cells, and a weaker band of the same size was also seen in the untransfected cells, indicating that the untransfected PAE cells contained a low amount of endogenous ALK-5. The 70 kDa complex was not observed when preimmune serum was used, or when immune serum was blocked by 10 μg of peptide. Moreover, a coprecipitated 94 kDa component could also be observed in the PAE/TβR-I cells. The latter component is likely to represent a TGF-β type II receptor complex, since an antiserum, termed DRL, which was raised against a synthetic peptide from the C-terminal part of the TGF-β type II receptor, precipitated a 94 kDa TGF-β type II receptor complex, as well as a 70 kDa type I receptor complex from PAE/TβR-I cells.
  • The carbohydrate contents of ALK-5 and the TGF-β type II receptor were characterized by deglycosylation using endoglycosidase F as described above and analyzed by SDS-polyacrylamide gel electrophoresis and autoradiography. The ALK-5 cross-linked complex shifted from 70 kDa to 66 kDa, whereas that of the type II receptor shifted from 94 kDa to 82 kDa. The observed larger shift of the type II receptor band compared with that of the ALK-5 band is consistent with the deglycosylation data of the type I and type II receptors on rat liver cells reported previously (Cheifetz et al (1988) J. Biol. Chem. 263, 16984-16991), and fits well with the fact that the porcine TGF-β type II receptor has two N-glycosylation sites (Lin et al (1992) Cell 68, 775-785), whereas ALK-5 has only one (see SEQ ID No. 9).
  • Binding of TGF-β1 to the type I receptor is known to be abolished by transient treatment of the cells with dithiothreitol (DTT) (Cheifetz and Massague (1991) J. Biol. Chem. 266, 20767-20772; Wrana et al (1992) Cell 71, 1003-1014). When analyzed by affinity cross-linking, binding of 125I-TGF-β1 to ALK-5, but not to the type II receptor, was completely abolished by DTT treatment of PAE/TβR-1 cells. Affinity cross-linking followed by immunoprecipitation by the VPN antiserum showed that neither the ALK-5 nor the type II receptor complexes was precipitated after DTT treatment, indicating that the VPN antiserum reacts only with ALK-5. The data show that the VPN antiserum recognizes a TGF-β type I receptor, and that the type I and type II receptors form a heteromeric complex.
  • 125I-TGF-β1 Binding & Affinity Crosslinking of Transfected COS Cells
  • Transient expression plasmids of ALKs-1 to -6 and TβR-II were generated by subcloning into the pSV7d expression vector or into the pcDNA I expression vector (Invitrogen). Transient transfection of COS-1 cells and iodination of TGF-β1 were carried out as described above. Crosslinking and immunoprecipitation were performed as described for PAE cells above.
  • Transfection of cDNAs for ALKs into COS-1 cells did not show any appreciable binding of 125I-TGFβ1, consistent with the observation that type I receptors do not bind TGF-β in the absence of type II receptors. When the TβR-II cDNA was co-transfected with cDNAs for the different ALKs, type I receptor-like complexes were seen, at different levels, in each case. COS-1 cells transfected with TβR-II and ALK cDNAs were analyzed by affinity crosslinking followed by immunoprecipitation using the DRL antisera or specific antisera against ALKs. Each one of the ALKs bound 125I-TGF-β1 and was coimmunoprecipitated with the TβR-II complex using the DRL antiserum. Comparison of the efficiency of the different ALKs to form heteromeric complexes with TβR-II, revealed that ALK-5 formed such complexes more efficiently than the other ALKs. The size of the crosslinked complex was larger for ALK-3 than for other ALKs, consistent with its slightly larger size.
  • Expression of the ALK Protein in Different Cell Types
  • Two different approaches were used to elucidate which ALK's are physiological type I receptors for TGF-β.
  • Firstly, several cell lines were tested for the expression of the ALK proteins by cross-linking followed by immunoprecipitation using the specific antiseras against ALKs and the TGF-β type II receptor. The mink lung epithelial cell line, Mv1Lu, is widely used to provide target cells for TGF-β action and is well characterized regarding TGF-β receptors (Laiho et al (1990) J. Biol. Chem. 265, 18518-18524; Laiho et al (1991) J. Biol. Chem. 266, 9108-9112). Only the VPN antiserum efficiently precipitated both type I and type II TGF-β receptors in the wild type Mv1Lu cells. The DRL antiserum also precipitated components with the same size as those precipitated by the VPN antiserum. A mutant cell line (R mutant) which lacks the TGF-β type I receptor and does not respond to TGF-β (Laiho et al, supra) was also investigated by cross-linking followed by immunoprecipitation. Consistent with the results obtained by Laiho et al (1990), supra the type III and type II TGF-β receptor complexes, but not the type I receptor complex, were observed by affinity crosslinking. Crosslinking followed by immunoprecipatition using the DRL antiserum revealed only the type II receptor complex, whereas neither the type I nor type II receptor complexes was seen using the VPN antiserum. When the cells were metabolically labelled and subjected to immunoprecipitation using the VPN antiserum, the 53 kDa ALK-5 protein was precipitated in both the wild-type and R mutant Mv1Lu cells. These results suggest that the type I receptor expressed in the R mutant is ALK-5, which has lost the affinity for binding to TGF-β after mutation.
  • The type I and type II TGF-β receptor complexes could be precipitated by the VPN and DRL antisera in other cell lines, including human foreskin fibroblasts (AG1518), human lung adenocarcinoma cells (A549), and human oral squamous cell carcinoma cells (HSC-2). Affinity cross-linking studies revealed multiple TGF-β type I receptor-like complexes of 70-77 kDa in these cells. These components were less efficiently competed by excess unlabelled TGF-β1 in HSC-2 cells. Moreover, the type II receptor complex was low or not detectable in A549 and HSC-2 cells. Cross-linking followed by immunoprecipitation revealed that the VPN antiserum precipitated only the 70 kDa complex among the 70-77 kDa components. The DRL antiserum precipitated the 94 kDa type II receptor complex as well as the 70 kDa type I receptor complex in these cells, but not the putative type I receptor complexes of slightly larger sizes. These results suggest that multiple type I TGF-β receptors may exist and that the 70 kDa complex containing ALK-5 forms a heteromeric complex with the TGF-β type II receptor cloned by Lin et al (1992) Cell 68, 775-785, more efficiently that the other species. In rat pheochromocytoma cells (PC12) which have been reported to have no TGF-β receptor complexes by affinity cross-linking (Massague et al (1990) Ann. N.Y. Acad. Sci. 593, 59-72), neither VPN nor DRL antisera precipitated the TGF-β receptor complexes. The antisera against ALKs-1 to -4 and ALK6 did not efficiently immunoprecipitate the crosslinked receptor complexes in porcine aortic endothelial (PAE) cells or human foreskin fibroblasts.
  • Next, it was investigated whether ALKs could restore responsiveness to TGF-β in the R mutant of Mv1Lu cells, which lack the ligand-binding ability of the TGF-β type I receptor but have intact type II receptor. Wild-type Mv1Lu cells and mutant cells were transfected with ALK cDNA and were then assayed for the production of plasminogen activator inhibitor-1 (PAI-1) which is produced as a result of TGF-β receptor activation as described previously by Laiho et al (1991) Mol. Cell Biol. 11, 972-978. Briefly, cells were added with or without 10 ng/ml of TGF-β1 for 2 hours in serum-free MCDB 104 without methionine. Thereafter, cultures were labelled with [35S] methionine (40 μCi/ml) for 2 hours. The cells were removed by washing on ice once in PBS, twice in 10 mM Tris-HCl (pH 8.0), 0.5% sodium deoxycholate, 1 mM PMSF, twice in 2 mM Tris-HCl (pH 8.0), and once in PBS. Extracellular matrix proteins were extracted by scraping cells into the SDS-sample buffer containing DTT, and analyzed by SDS-gel electrophoresis followed by fluorography using Amplify. PAI-1 can be identified as a characteristic 45kDa band (Laiho et al (1991) Mol. Cell Biol. 11, 972-978). Wild-type Mv1Lu cells responded to TGF-β and produced PAI-1, whereas the R mutant clone did not, even after stimulation by TGF-β1. Transient transfection of the ALK-5 cDNA into the R mutant clone led to the production of PAI-1 in response to the stimulation by TGF-β1, indicating that the ALK-5 cDNA encodes a functional TGF-β type I receptor. In contrast, the R mutant cells that were transfected with other ALKs did not produce PAI-1 upon the addition of TGF-β1.
  • Using similar approaches as those described above for the identification of TGF-β-binding ALKs, the ability of ALKs to bind activin in the presence of ActRII was examined. COS-1 cells were co-transfected as described above. Recombinant human activin A was iodinated using the chloramine T method (Mathews and Vale (1991) Cell 65, 973-982). Transfected COS-1 cells were analysed for binding and crosslinking of 125I-activin A in the presence or absence of excess unlabelled activin A. The crosslinked complexes were subjected to immunoprecipitation using DRL antisera or specific ALK antisera.
  • All ALKs appear to bind activin A in the presence of Act R-II. This is more clearly demonstrated by affinity cross-linking followed by immunopreciptation. ALK-2 and ALK-4 bound 125I-activin A and were coimmunoprecipitated with ActR-II. Other ALKs also bound 125I-activin A but with a lower efficiency compared to ALK-2 and ALK-4.
  • In order to investigate whether ALKs are physiological activin type I receptors, activin responsive cells were examined for the expression of endogenous activin type I receptors. Mv1Lu cells, as well as the R mutant, express both type I and type II receptors for activin, and the R mutant cells produce PAI-1 upon the addition of activin A. Mv1Lu cells were labeled with 125I-activin A, cross-linked and immunoprecipitated by the antisera against ActR-II or ALKs as described above.
  • The type I and type II receptor complexes in Mv1Lu cells were immunoprecipitated only by the antisera against ALK-2, ALK-4 and ActR-II. Similar results were obtained using the R mutant cells. PAE cells do not bind activin because of the lack of type II receptors for activin, and so cells were transfected with a chimeric receptor, to enable them to bind activin, as described herein. A plasmid (chim A) containing the extracelluar domain and C-terminal tail of Act R-II (amino-acids -19 to 116 and 465 to 494, respectively (Mathews and Vale (1991) Cell, 65, 973-982)) and the kinase domain of TβR-II (amino-acids 160-543) (Lin et al (1992) Cell, 68, 775-785) was constructed and transfected into pcDNA/neo (Invitrogen). PAE cells were stably transfected with the chim A plasmid by electroporation, and cells expressing the chim A protein were established as described previously. PAE/Chim A cells were then subjected to 125I-activin A labelling crosslinking and immunoprecipitation as described above.
  • Similar to Mv1Lu cells, activin type I receptor complexes in PAE/Chim A cells were immunoprecipitated by the ALK-2 and ALK-4 antisera. These results show that both ALK-2 and ALK-4 serve as high affinity type I receptors for activin A in these cells.
  • ALK-1, ALK-3 and ALK-6 bind TGF-β1 and activin A in the presence of their respective type II receptors, but the functional consequences of the binding of the ligands remains to be elucidated.
  • The experiments described supra suggested further experiments. Specifically, it is known that TGF-β family members acts as ligands in connection with specific type I and type II receptors, with resulting complexes interacting with members of the Smad family. See Heldin et al., Nature 390: 465-471 (1997), incorporated by reference. The Smad molecules are homologs of molecules found in Drosophila (“Mad”), and C. elegans (Sma), hence, the acronym “Smad”. These are involved in signal transduction pathways downstream of serine/threonine kinase receptors. See Massagué et al., Trends Cell Biol. 2: 187-192 (1997). The different members of the family have different signaling roles. Smad1, for example, as well as Smad 2 and 3, and perhaps Smad 5, became phosphorylated via specific type 1 serine/threonine kinase receptors, and act in pathway restricted fashion. For example, Xenopus Mad1 induces ventral mesoderm, in the presence of BMP. The human Smad1 has been shown to have ventralizing activity. See Liu et al., Nature 381: 620-623 (1996); Kretzschmer et al., Genes Dev 11: 984-995 (1997). There is also some evidence that TGF-β phosphorylates Smad1. See Lechleider et al., J. Biol. Chem. 271: 17617-17620 (1996); Yingling et al., Proc. Natl. Acad. Sci. USA 93: 8940-8944 (1996). Given what was known regarding this complex signaling pathway, the role of ALK-1 was studied.
  • COS-7 cells, which do not express ALK-1, were transfected with cDNA encoding tagged ALK-1. The tag was hemagluttinin (hereafter “HA”), and a commercially available lipid containing transfecting agent was used. In parallel experiments, porcine aortic endothelial (PAE) cells were also used, because these cells express TGFβ type II receptors, and ALK-5, but not ALK-1. Hence, PAE cells were either transfected, or not. Transfection protocols are given, supra.
  • The cells were then contacted with 125I labelled TGF-β1, and were then contacted with ALK-1 specific antisera, to ascertain whether cross linking had occurred. See the experiments, supra, as well as ten Dijke et al., Science 264: 101-104 (1994), incorporated by reference. Antisera to ALK-5 were also used.
  • The results indicated that the ALK-1 antiserum immunoprecipitated complexes of the appropriate size from the transfected COS-7 and PAE cells, but not those which were not transfected, thereby establishing that ALK-1 is a receptor for TGF-β.
  • This was confirmed in experiments on human umbilical vein endothelial cells (HUVEC). These cells are known to express ALK-1 endogenously, as well as ALK-5. The ALK-5 antiserum and the ALK-1 antiserum both immunoprecipitated type I and type II receptor cross linked complexes. The ALK-1 antiserum immunoprecipitated band migrated slightly more slowly than the band immunprecipitated by the ALK-5 antiserum (see, e.g., FIG. 8). This is in agreement with the difference in size of ALK-1 and ALK-5, and it indicates that both ALK-1 and ALK-5 bind TGF-β in HUVECS.
  • Further, it shows that ALK-1 acts as a co-called “type I” TGF-β receptor in an endogenous, physiological setting.
  • Once it was determined that TGF-β and ALK-1 interact, studies were carried out to determine whether or not activation of ALK-1 resulted in phosphorylation of Smads. To test this, COS-7 cells were transfected in the same manner described supra with either Flag tagged Smad1, Flag tagged Smad2 or Flag tagged Smad-5 together with either a constitutively active form of ALK-1, or a constitutively active form of ALK-5. Specifically, the variant of ALK-1 is Q201D, and that of ALK-5 is T204D. Constitutively active ALK-1 was used to avoid the need for an additional transfection step. To elaborate, it is known that for the TGF-β pathway to function adequately, a complex of two, type I receptors, and two, type II receptors must interact, so as to activate the receptors. Constitutively active receptors, such as what was used herein, do not require the presence of the type II receptor to function. See Wieser et al., EMBO J 14: 2199-2208 (1995). In order to determine if the resulting transfected cells produced phosphorylated Smads, Smads were determined using a Flag specific antibody, which precipitated them, and phosphorylation was determined using the antiphosphoserine antibody of Nishimura et al., J. Biol. Chem. 273: 1872-1879 (1998). It was determined, when the data were analyzed, that Smad1 and Smad-5 (an intracellular signalling molecule which is structurally highly similar to Smad1) were phosphorylated following interaction with activated ALK-1, but not following interaction of TGF-β and ALK-5. Conversely, the interaction of TGF-β and ALK-5 led to phosphorylation of Smad 2, but not Smad 1. This supports a conclusion that ALK-1 transduces signal in a manner similar to BMPs.
  • FIG. 8 depicts the phosphorylation of Smad-5 following interaction with ALK-1 but not ALK-5. Phosphorylation of both Smad-5 and Smad1 has been shown for BMP type I receptors suggesting ALK-1 is functionally very similar to ALK3 (BMPR-IA) and (ALK6 BMPR-IB).
  • Additional experiments were then carried out to study the interaction of ALK-1 with Smad-1. Specifically, COS-7 cells were transfected with cDNA which encoded the wild type form of the TGFβ type II receptor (TBR-II), a kinase inactive form of ALK-1, and Flag tagged Smad-1. Kinase inactive ALK-1 was used, because the interaction of Smad-1 and receptors is known to be transient, as once Smads are phosphorylated they dissociate from the type I receptor. See Marcias-Silva et al., Cell 87: 1215-1224 (1996); Nakao et al., EMBO J 16: 5353-5362 (1997). Affinity cross-linking, using 125I-TGF-β1, and immunoprecipitation with Flag antibody was carried out, as discussed supra. The expression of ALK-1 was determined using anti-HA antibody, since the vector used to express ALK-1 effectively tagged it with HA.
  • The immunoprecipitating of Smad1 resulted in coprecipitation of a cross linked TBR-II/ALK-1 complex, suggesting a direct association of Smad1 with ALK-1.
  • These examples show that one can identify molecule which inhibit, or enhance expression of a gene whose expression is regulated by phosphorylated Smad1. To elaborate, as ALK-1 has been identified as a key constituent of the pathway by which Smad1 is phosphorylated, one can contact cells which express both Smad1 and ALK-1 with a substance of interest, and then determine if the Smad1 becomes phosphorylated. The cells can be those which inherently express both ALK-1 and Smad1, or which have been transformed or transfected with DNA encoding one or both of these. One can determine the phosphorylation via, e.g., the use of anti phosphorylated serine antibodies, as discussed supra. In an especially preferred embodiment, the assay can be carried out using TGF-β, as a competing agent. The TGF-β, as has been shown, does bind to ALK-1, leading to phosphorylation of Smad1. Hence, by determining a value with TGF-β alone, one can then compare a value determined with amounts of the substance to be tested, in the presence of TGF-β. Changes in phosphorylation levels can thus be attributed to the test substance.
  • In this type of system, it must be kept in mind that both type I receptors and type II receptors must be present; however, as indicated, supra, one can eliminate the requirement for a type II receptor by utilizing a constitutively active form of ALK-1, such as the form described supra. Additional approaches to inhibiting this system will be clear to the skilled artisan. For example, since it is known that there is interaction between Smad1 and the ALK-1 receptor, one can test for inhibition via the use of small molecules which inhibit the receptor/Smad interaction. Heldin et al., supra, mention Smad6 and Smad7 as Smad1 inhibitors, albeit in the context of a different system. Hence one can test for inhibition, or inhibit the interaction, via adding a molecule to be tested or for actual inhibition to a cell, wherein the molecule is internalized by the cell, followed by assaying for phosphorylation, via a method such as is discussed supra.
  • In a similar way, one can assay for inhibitors of type I/type II receptor interaction, by testing the molecule of interest in a system which includes both receptors, and then assaying for phorphorylation.
  • Conversely, activators or agonists can also be tested for, or utilized, following the same type of procedures.
  • Via using any of these systems, one can identify any gene or genes which are activated by phosphorylated Smad1. To elaborate, the art is very familiar with systems of expression analysis, such as differential display PCR, subtraction hybridization, and other systems which combine driver and testes populations of nucleic acids, whereby transcripts which are expressed or not expressed can be identified. By simply using an activator/inhibitor of the system disclosed herein, on a first sample, and a second sample where none is used, one can then carry out analysis of transcript, thereby determining the transcripts of interest.
  • Also a part of the invention is the regulation of a phosphorylation of Smad-1 or Smad-5, with inhibitors, such as antibodies against the extracellular domain of ALK-1 or TGF-β, or enhancers, such as TGF-β itself, or those portions of the TGF-β molecule which are necessary for binding. Indeed, by appropriate truncation, one can also determine what portions of ALK-1 are required for phosphorylation of Smad1 or Smad-5 to take place.
  • The invention has been described by way of example only, without restriction of its scope. The invention is defined by the subject matter herein, including the claims that follow the immediately following full Sequence Listings.

Claims (15)

1-28. (cancelled)
29 A method for determining if a substance inhibits binding of TGF-β to Alk-1 comprising contacting a cell that expresses an Alk-1 with said substance in the presence of TGF-β and determining if said substance inhibits binding of TGF-β to Alk-1.
30 The method of claim 29, wherein said substance is an antibody that binds to TGF-β.
31 The method of claim 29, wherein said substance is an antibody that binds to the extracellular domain of Alk-1.
32 The method of claim 29 wherein said cells that express Alk-1 are transfected with a nucleic acid molecule which encodes Alk-1.
33 The method of claim 29 wherein said Alk-1 is a constitutively active Alk-1.
34 The method of claim 29, wherein said Alk-1 is a kinase inactive Alk-1.
35 The method of claim 32, wherein said Alk-1 is an Alk-1 fusion polypeptide.
36 The method of claim 35, wherein said Alk-1 is fused to hemagglutinin.
37 The method of claim 29, wherein said cells that express Alk-1 are transfected with a nucleic acid molecule which encodes a Smad1 or a nucleic acid molecule which encodes a Smad5.
38 The method of claim 37, wherein said Smad1 is a Smad1 fusion polypeptide.
39 The method of claim 38, wherein said Smad1 is fused to Flag.
40 The method of claim 37, wherein said Smad5 is a Smad5 fusion polypeptide.
41 The method of claim 40, wherein said Smad5 is fused to Flag.
42 The method of claim 29, wherein inhibition of Smad1 or Smad5 phosphorylation in said cells that express an Alk-1 indicates inhibition of binding of TGF-β to Alk-1.
US10/739,413 1992-11-17 2003-12-19 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use Abandoned US20050048607A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/739,413 US20050048607A1 (en) 1992-11-17 2003-12-19 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use
US11/980,023 US20080131910A1 (en) 1992-11-17 2007-10-30 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use

Applications Claiming Priority (18)

Application Number Priority Date Filing Date Title
GB9224057.1 1992-11-17
GB929224057A GB9224057D0 (en) 1992-11-17 1992-11-17 Novel protein kinase receptors
GB939304677A GB9304677D0 (en) 1993-03-08 1993-03-08 Protein kinases
GB9304680.3 1993-03-08
GB939304680A GB9304680D0 (en) 1993-03-08 1993-03-08 Protein kinases
GB9304677.9 1993-03-08
GB939311047A GB9311047D0 (en) 1993-05-28 1993-05-28 Protein kinases
GB9311047.6 1993-05-28
GB939313763A GB9313763D0 (en) 1993-07-02 1993-07-02 Protein kinases
GB9313763.6 1993-07-02
GB9136099.2 1993-08-03
GB939316099A GB9316099D0 (en) 1993-08-03 1993-08-03 Protein kinases
GB939321344A GB9321344D0 (en) 1993-10-15 1993-10-15 Protein kinases
GB9321344.5 1993-10-15
US08/436,265 US6316217B1 (en) 1992-11-17 1993-11-17 Activin receptor-like kinases, proteins having serine threonine kinase domains and polynucleotides encoding same
US3917798A 1998-03-13 1998-03-13
US09/267,963 US6692925B1 (en) 1992-11-17 1999-03-12 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use
US10/739,413 US20050048607A1 (en) 1992-11-17 2003-12-19 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/267,963 Division US6692925B1 (en) 1992-11-17 1999-03-12 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/980,023 Continuation US20080131910A1 (en) 1992-11-17 2007-10-30 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use

Publications (1)

Publication Number Publication Date
US20050048607A1 true US20050048607A1 (en) 2005-03-03

Family

ID=31192585

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/267,963 Expired - Lifetime US6692925B1 (en) 1992-11-17 1999-03-12 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use
US10/739,413 Abandoned US20050048607A1 (en) 1992-11-17 2003-12-19 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use
US11/980,023 Abandoned US20080131910A1 (en) 1992-11-17 2007-10-30 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use
US12/803,779 Expired - Fee Related US8012704B2 (en) 1992-11-17 2010-07-06 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/267,963 Expired - Lifetime US6692925B1 (en) 1992-11-17 1999-03-12 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/980,023 Abandoned US20080131910A1 (en) 1992-11-17 2007-10-30 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use
US12/803,779 Expired - Fee Related US8012704B2 (en) 1992-11-17 2010-07-06 Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use

Country Status (1)

Country Link
US (4) US6692925B1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070065444A1 (en) * 2005-09-07 2007-03-22 Amgen Fremont Inc. Human monoclonal antibodies to activin receptor-like kinase-1
US20080131910A1 (en) * 1992-11-17 2008-06-05 Kohei Miyazono Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use
US20080175844A1 (en) * 2006-11-02 2008-07-24 Acceleron Pharma, Inc. ALK1 receptor and ligand antagonist and uses thereof
US8158584B2 (en) 2008-05-02 2012-04-17 Acceleron Pharma, Inc. Pharmaceutical preparations comprising an ALK1-Fc fusion protein
US8642031B2 (en) 2006-11-02 2014-02-04 Acceleron Pharma, Inc. Antagonists of BMP9, BMP10, ALK1 and other ALK1 ligands, and uses thereof
US10059756B2 (en) 2006-11-02 2018-08-28 Acceleron Pharma Inc. Compositions comprising ALK1-ECD protein

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2332977T3 (en) * 2004-07-23 2016-02-29 Acceleron Pharma Inc ActRII receptor polypeptides
US7897581B2 (en) 2005-02-24 2011-03-01 Massachusetts Eye & Ear Infirmary Methods and compounds for promoting vessel regression
AU2012203432C1 (en) * 2005-09-07 2013-08-29 Amgen Fremont Inc. Human Monoclonal Antibodies to Activin Receptor-like Kinase-1
AU2014218362B2 (en) * 2005-09-07 2017-02-02 Amgen Fremont Inc. Human Monoclonal Antibodies to Activin Receptor-like Kinase-1
AU2013205016B2 (en) * 2005-09-07 2014-06-05 Amgen Fremont Inc. Human Monoclonal Antibodies to Activin Receptor-like Kinase-1
KR101557375B1 (en) 2005-11-23 2015-10-08 악셀레론 파마 인코포레이티드 Activin-actrπa antagonists and uses for promoting bone growth
US8128933B2 (en) * 2005-11-23 2012-03-06 Acceleron Pharma, Inc. Method of promoting bone growth by an anti-activin B antibody
KR102223231B1 (en) * 2006-12-18 2021-03-08 악셀레론 파마 인코포레이티드 Activin-actrii antagonists and uses for increasing red blood cell levels
US20100028332A1 (en) * 2006-12-18 2010-02-04 Acceleron Pharma Inc. Antagonists of actriib and uses for increasing red blood cell levels
US8895016B2 (en) * 2006-12-18 2014-11-25 Acceleron Pharma, Inc. Antagonists of activin-actriia and uses for increasing red blood cell levels
EP2111229B1 (en) * 2007-02-01 2013-04-10 Acceleron Pharma, Inc. Pharmaceutical compositions comprising Activin-ActRIIA antagonists for use in preventing or treating breast cancer metastasis or breast cancer related bone loss
TW201803890A (en) 2007-02-02 2018-02-01 艾瑟勒朗法瑪公司 Variants derived from ActRIIB and uses therefor
KR20160129095A (en) * 2007-02-09 2016-11-08 악셀레론 파마 인코포레이티드 Activin-actriia antagonists and uses for promoting bone growth in cancer patients
CN101861161B (en) 2007-09-18 2017-04-19 阿塞勒隆制药公司 Activin-ACTRIIA antagonists and uses for decreasing or inhibiting FSH secretion
KR20180073706A (en) * 2008-06-26 2018-07-02 악셀레론 파마 인코포레이티드 Methods for dosing an activin-actriia antagonist and monitoring of treated patients
PT3494986T (en) 2008-08-14 2020-07-14 Acceleron Pharma Inc Gdf traps for use to treat anemia
US8216997B2 (en) 2008-08-14 2012-07-10 Acceleron Pharma, Inc. Methods for increasing red blood cell levels and treating anemia using a combination of GDF traps and erythropoietin receptor activators
AU2010204985A1 (en) * 2009-01-13 2011-08-04 Acceleron Pharma Inc. Methods for increasing adiponectin
US8178488B2 (en) 2009-06-08 2012-05-15 Acceleron Pharma, Inc. Methods for increasing thermogenic adipocytes
KR20190090049A (en) 2009-06-12 2019-07-31 악셀레론 파마 인코포레이티드 TRUNCATED ActRIIB-FC FUSION PROTEINS
EP3202459B1 (en) * 2009-09-09 2021-04-14 Acceleron Pharma Inc. Actriib antagonists and dosing and uses thereof for treating obesity or type 2 diabetes by regulating body fat content
EP3260130B1 (en) * 2009-11-03 2021-03-10 Acceleron Pharma Inc. Methods for treating fatty liver disease
CA2781152A1 (en) * 2009-11-17 2011-05-26 Acceleron Pharma Inc. Actriib proteins and variants and uses therefore relating to utrophin induction for muscular dystrophy therapy
AU2011326586A1 (en) 2010-11-08 2013-05-30 Acceleron Pharma, Inc. ActRIIA binding agents and uses thereof
RU2018145985A (en) 2012-11-02 2019-02-18 Селджин Корпорейшн ACTIVIN-ACTRII ANTAGONISTS AND THEIR APPLICATION FOR TREATMENT OF BONE TISSUE DISORDERS AND OTHER DISORDERS
WO2015152183A1 (en) * 2014-03-31 2015-10-08 大日本住友製薬株式会社 Prophylactic agent and therapeutic agent for fibrodysplasia ossificans progressiva
US9850298B2 (en) 2014-06-13 2017-12-26 Acceleron Pharma Inc. Methods for treating ulcers in thalassemia syndrome with an ActRIIB polypeptide
MA41052A (en) 2014-10-09 2017-08-15 Celgene Corp TREATMENT OF CARDIOVASCULAR DISEASE USING ACTRII LIGAND TRAPS
TWI730949B (en) 2014-12-03 2021-06-21 美商西建公司 Activin-actrii antagonists and uses for treating anemia

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5420263A (en) * 1992-04-07 1995-05-30 The Johns Hopkins University Amplification of human MDM2 gene in human tumors
US5521295A (en) * 1993-06-07 1996-05-28 Amgen Inc. Nucleic acids encoding hybrid receptor molecules
US5538892A (en) * 1992-03-18 1996-07-23 The General Hospital Corporation Nucleic acids encoding a TGF-β type 1 receptor
US5614609A (en) * 1994-10-20 1997-03-25 Carlos F. Ibanez Serine threonine kinase receptor

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6692925B1 (en) * 1992-11-17 2004-02-17 Ludwig Institute For Cancer Research Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use
US6291206B1 (en) 1993-09-17 2001-09-18 Genetics Institute, Inc. BMP receptor proteins
US6248554B1 (en) 1993-11-24 2001-06-19 The Procter & Gamble Company DNA sequence coding for a BMP receptor
US5968752A (en) 1995-08-14 1999-10-19 Creative Biomolecules, Inc. Method for identifying an OP-1 analog which binds an ALK-1 receptor

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5538892A (en) * 1992-03-18 1996-07-23 The General Hospital Corporation Nucleic acids encoding a TGF-β type 1 receptor
US5420263A (en) * 1992-04-07 1995-05-30 The Johns Hopkins University Amplification of human MDM2 gene in human tumors
US5521295A (en) * 1993-06-07 1996-05-28 Amgen Inc. Nucleic acids encoding hybrid receptor molecules
US5614609A (en) * 1994-10-20 1997-03-25 Carlos F. Ibanez Serine threonine kinase receptor

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080131910A1 (en) * 1992-11-17 2008-06-05 Kohei Miyazono Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use
US20070065444A1 (en) * 2005-09-07 2007-03-22 Amgen Fremont Inc. Human monoclonal antibodies to activin receptor-like kinase-1
US7537762B2 (en) 2005-09-07 2009-05-26 Amgen Fremont, Inc. Human monoclonal antibodies to activin receptor-like kinase-1
US8080646B2 (en) 2005-09-07 2011-12-20 Amgen Fremont, Inc. Human monoclonal antibodies to activin receptor-like kinase-1
US9221915B2 (en) 2005-09-07 2015-12-29 Pfizer Inc. Human monoclonal antibodies to activin receptor-like kinase-1
US20080175844A1 (en) * 2006-11-02 2008-07-24 Acceleron Pharma, Inc. ALK1 receptor and ligand antagonist and uses thereof
US8455428B2 (en) 2006-11-02 2013-06-04 Acceleron Pharma, Inc. ALK1 receptor and ligand antagonist and uses thereof
US8642031B2 (en) 2006-11-02 2014-02-04 Acceleron Pharma, Inc. Antagonists of BMP9, BMP10, ALK1 and other ALK1 ligands, and uses thereof
US9452197B2 (en) 2006-11-02 2016-09-27 Acceleron Pharma, Inc. Antagonists of BMP9, BMP10, ALK1 and other ALK1 ligands, and uses thereof
US10059756B2 (en) 2006-11-02 2018-08-28 Acceleron Pharma Inc. Compositions comprising ALK1-ECD protein
US8158584B2 (en) 2008-05-02 2012-04-17 Acceleron Pharma, Inc. Pharmaceutical preparations comprising an ALK1-Fc fusion protein

Also Published As

Publication number Publication date
US6692925B1 (en) 2004-02-17
US20080131910A1 (en) 2008-06-05
US20100317011A1 (en) 2010-12-16
US8012704B2 (en) 2011-09-06

Similar Documents

Publication Publication Date Title
US8012704B2 (en) Proteins having serine/threonine kinase domains, corresponding nucleic acid molecules, and their use
US6982319B2 (en) Antibodies which bind specifically to activin receptor-like kinases
US8080639B2 (en) Antibodies which bind specifically to activin receptor like kinase 3
US5707632A (en) Receptors for fibroblast growth factors
US7071159B2 (en) Inhibitor of vascular endothelial cell growth factor
EP0538404B1 (en) Fibroblast growth factor receptors
WO1990008822A1 (en) Erythropoietin receptor
US5863888A (en) Human Bek Fibroblast growth factor receptor
EP2078752A2 (en) ALK-1 responds to TGF-beta and signals through SMAD-1 and SMAD-5
IE920318A1 (en) Human platelet-derived growth factor receptors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION