US20100150885A1 - Methods and compositions for inducing brown adipogenesis - Google Patents

Methods and compositions for inducing brown adipogenesis Download PDF

Info

Publication number
US20100150885A1
US20100150885A1 US11/914,425 US91442506A US2010150885A1 US 20100150885 A1 US20100150885 A1 US 20100150885A1 US 91442506 A US91442506 A US 91442506A US 2010150885 A1 US2010150885 A1 US 2010150885A1
Authority
US
United States
Prior art keywords
bmp
cells
stem cells
mesenchymal stem
population
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
US11/914,425
Inventor
Yu-Hua Tseng
C. Ronald Kahn
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.)
Joslin Diabetes Center Inc
Original Assignee
Joslin Diabetes Center Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Joslin Diabetes Center Inc filed Critical Joslin Diabetes Center Inc
Priority to US11/914,425 priority Critical patent/US20100150885A1/en
Assigned to JOSLIN DIABETES CENTER, INC. reassignment JOSLIN DIABETES CENTER, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KAHN, C. RONALD, TSENG, YU-HUA
Publication of US20100150885A1 publication Critical patent/US20100150885A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • C07K14/475Growth factors; Growth regulators
    • C07K14/495Transforming growth factor [TGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0653Adipocytes; Adipose tissue
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0667Adipose-derived stem cells [ADSC]; Adipose stromal stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/01Modulators of cAMP or cGMP, e.g. non-hydrolysable analogs, phosphodiesterase inhibitors, cholera toxin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/02Compounds of the arachidonic acid pathway, e.g. prostaglandins, leukotrienes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/33Insulin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/385Hormones with nuclear receptors of the family of the retinoic acid recptor, e.g. RAR, RXR; Peroxisome proliferator-activated receptor [PPAR]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/395Thyroid hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • Obesity which is generally associated with an abnormal accumulation of fat cells, develops when energy intake exceeds energy expenditure.
  • Adipose tissues play an important role in obesity, insulin resistance and diabetes.
  • Two functionally different types of fat tissues are present in mammals: white adipose tissue (WAT), which is the primary site of depot of triglycerides and release of fatty acids, and brown adipose tissue (BAT), which is specialized in thermogenic energy expenditure through the expression of uncoupling protein-1 (UCP-1).
  • WAT white adipose tissue
  • BAT brown adipose tissue
  • the most commonly known fat cells are white fat cells, also known as white adipose tissue (WAT) cells, which have a thin ring of cytoplasm surrounding a lipid or fat droplet. WAT is found underneath the skin and provides heat insulation, cushioning against shock and jarring, and energy reserves. An average lean person has roughly 20 to 40 billion WAT cells. An obese person can have up to ten times more WAT than the average lean person.
  • WAT white adipose tissue
  • brown fat cells also known as brown adipose tissue (BAT) cells.
  • BAT brown adipose tissue
  • Energy expenditure for thermogenesis in BAT serves either to maintain body temperature in the cold or to waste food energy. It has roles in thermal balance and energy balance, and when defective, is usually associated with obesity. BAT is typically atrophied in obese animals. The importance of BAT in overall energy homeostasis is underscored by the finding that ablation of BAT in mice results in severe obesity accompanied by insulin resistance, hyperglycemia, hyperlipidemia, and hypercholesterolemia (Lowell at al., Nature 366(6457):740-2 (1993); Hamann et al., Diabetes.
  • BAT also features the presence of abundant and large mitochondria (Nedergaard et al., in Brown Adipose Tissue , Trayhurn and Nicholls, Eds. (Edward Arnold, Baltimore, 1986)), which serve as the center site for oxidative phosphorylation, intermediary metabolism, adaptive thermogenesis, generation of reactive oxygen species and apoptosis.
  • mitochondrial biogenesis has been long known to accompany brown adipocyte differentiation.
  • Adipose tissues contain a potential mitotic compartment, which can allow for growth and differentiation of WAT or BAT cells.
  • Adipose tissue can be readily assayed using routine techniques.
  • An exemplary assay for adipose cells is the Oil Red O lipophilic red dye assay. The dye is used to stain neutral lipids in cells. The amount of staining is directly proportional to the amount of lipid in the cell and can be measured spectrophotometrically. The amount of lipid accumulation is determined as a parameter of differentiation.
  • WAT and BAT can be distinguished by routine techniques, e.g., morphologic changes specific to WAT or BAT, or evaluation of WAT-specific or BAT-specific markers.
  • BAT cells can be identified by expression of uncoupling protein (UCP), e.g., UCP-1.
  • UCP uncoupling protein
  • Bone morphogenetic proteins belong to the TGF ⁇ superfamily. BMPs bind to specific type-I and -II serine/threonine kinase receptor complexes, RIa, RIb, and RII, which signal through SMAD proteins or the p38 mitogen-activated protein kinase (MAPK).
  • the BMPs are important regulators of key events in many aspects of tissue development and morphogenesis, including the processes of bone formation during embryogenesis, postnatal growth, remodeling and regeneration of the skeleton. Localization studies in both human and mouse tissues have demonstrated high levels of mRNA expression and protein synthesis for various BMPs in adipose, heart, lung, small intestine, limb bud and teeth.
  • BMP-2 has been implicated in early skeletal development, including BMP-2, -4, -5, -7, -14 (CDMP-1/GDF-5).
  • Other members such as BMP-3, -6, -7 and -13 (CDMP-2/GDF-6) may be involved in later stages of skeletal formation.
  • the present invention is based, in part, on the discovery that bone morphogenetic proteins (BMPs) play an important role in adipocyte differentiation.
  • BMPs bone morphogenetic proteins
  • BMPs 2, 4, 5, 6, and 7 markedly induce differentiation of brown preadipocytes, even in the absence of normally required induction cocktails.
  • treatment of stem cells with BMPs triggers commitment of these cells to the brown adipocyte lineage.
  • Implantation of BMP-7-treated stem cells into athymic mice leads to development of these cells into a tissue containing both brown and white adipocytes. In addition, these mice become more insulin-sensitive.
  • adenoviral-mediated BMP-7 expression in normal mice results in a significant increase in brown fat mass and energy expenditure, and a significant reduction of body weight in diet-induced obese C57BL/6 mice.
  • brown adipose tissue BAT
  • the methods described herein are useful for the treatment of obesity and related disorders, such as diabetes.
  • the methods can also be used to decrease fat stores in subjects including food animals, e.g., to improve the quality of the meat derived therefrom.
  • the invention features methods of modulating adipose tissue function or development, e.g., promoting BAT adipogenesis, in a subject.
  • the methods include administering to the subject a population of BMP-activated stem cells, e.g., pluripotent mesenchymal stem cells, as described herein, wherein said population of BMP-activated stem cells, or their progeny (i.e., daughter cells), undergo brown adipogenesis.
  • the invention features methods of treating a subject, e.g., decreasing fat stores or weight in a subject such as a human.
  • the methods include administering to the subject a population of BMP-activated stem cells, e.g., pluripotent mesenchymal stem cells, as described herein, wherein said population of BMP-activated stem cells, or their progeny, undergo brown adipogenesis.
  • the methods can optionally include identifying a subject in need of decreasing fat stores or weight.
  • the invention includes methods of enhancing insulin sensitivity in a subject, e.g., a subject that is insulin-resistant.
  • the methods include administering to the subject a population of BMP-activated stem cells, e.g., pluripotent mesenchymal stem cells, as described herein, wherein said population of BMP-activated stem cells, or their progeny, undergo brown adipogenesis.
  • the methods can optionally include identifying a subject in need of enhanced insulin sensitivity.
  • BMP-activated means that stem cell has an artificially enhanced level of BMP signalling, e.g., BMP-2, -4, -5, -6, and/or -7 signalling.
  • “Artificially” enhanced means that the level of BMP signalling has been increased by direct human intervention.
  • BMP signalling can be enhanced by any method described herein, e.g., by treating the cell with a compound that enhances BMP signalling as described herein, e.g., a BMP polypeptide or nucleic acid.
  • BMP signalling can be enhanced by any method described herein, e.g., by treating the cell with a compound that enhances BMP signalling as described herein, e.g., a BMP polypeptide or nucleic acid.
  • Populations of stem cells activated by methods described herein are also included within the present invention.
  • the cells can be autologous, allogeneic or xenogeneic.
  • methods described herein can include treating (e.g., contacting) a population of stem cells, e.g., pluripotent mesenchymal stem cells, with a compound in an amount sufficient to increase BMP signalling, thereby producing a population of BMP-activated cells.
  • stem cells e.g., pluripotent mesenchymal stem cells
  • methods described herein can include implanting a population of BMP-activated cells into a subject.
  • the BMP-activated cells can be implanted directly or can be administered in a scaffold, matrix, or other implantable device to which the cells can attach (examples include carriers made of, e.g., collagen, fibronectin, elastin, cellulose acetate, cellulose nitrate, polysaccharide, fibrin, gelatin, and combinations thereof).
  • the methods include implanting a population of BMP-activated cells comprising a sufficient number of cells to promote brown adipogenesis in the subject, e.g., to increase the amount of BAT in the subject by at least 1%, e.g., 2%, 5%, 7%, 10%, 15%, 20%, 25% or more.
  • the methods include providing a purified population of stem cells, e.g., a population of pluripotent mesenchymal stem cells, (e.g., a population of cells in which at least 60%, e.g., 70%, 80%, 90% or more of the cells are stem cells); and contacting the cells with a compound that increases expression, levels or activity of one or more of BMP-2, -4, -5, -6, and/or -7, as described herein, thereby activating the cells.
  • a purified population of stem cells e.g., a population of pluripotent mesenchymal stem cells, (e.g., a population of cells in which at least 60%, e.g., 70%, 80%, 90% or more of the cells are stem cells)
  • the methods include evaluating the level of BAT adipogenesis in the cell or cell population.
  • BAT differentiation can be evaluated by measuring any of, e.g., a BAT marker, such as uncoupling protein (UCP), e.g., UCP-1, expression; BAT morphology (e.g., using visual, e.g., microscopic, inspection of the cells); or BAT thermodynamics, e.g., cytochrome oxidase activity, Na+-K+-ATPase enzyme units, or other enzymes involved in BAT thermogenesis.
  • the methods include evaluating WAT differentiation, e.g., evaluating a WAT specific marker or WAT morphology.
  • a compound that increases BMP-2, -4, -5, -6, and/or -7 signaling can be, e.g., one or more of the following:
  • the compound is a BMP-2, -4, -5, -6, and/or -7 polypeptide or nucleic acid.
  • a “BMP-2, -4, -5, -6, and/or -7 polypeptide or nucleic acid” is a BMP-2, -4, -5, -6, and/or -7 polypeptide or nucleic acid as described herein, e.g., a mature human BMP-2, -4, -5, -6, and/or -7 polypeptide or active fragment thereof, or a nucleic acid encoding a mature human BMP-2, -4, -5, -6, and/or -7 polypeptide or active fragment thereof.
  • the compound is a BMP-2 polypeptide, e.g., human BMP-2, e.g., a mature BMP-2 polypeptide, e.g., a BMP-2 polypeptide that includes amino acids 283-396 of SEQ ID NO:1.
  • the polypeptide can be a recombinant polypeptide.
  • the compound is a BMP-4 polypeptide, e.g., human BMP-4, e.g., a mature BMP-4 polypeptide, e.g., a BMP-4 polypeptide that includes amino acids 293-408 of SEQ ID NO:2.
  • the polypeptide can be a recombinant polypeptide.
  • the compound is a BMP-5 polypeptide, e.g., human BMP-5, e.g., a mature BMP-5 polypeptide, e.g., a BMP-4 polypeptide that includes amino acids 323-454 of SEQ ID NO:3.
  • the polypeptide can be a recombinant polypeptide.
  • the compound is a BMP-6 polypeptide, e.g., human BMP-6, e.g., a mature BMP-6 polypeptide, e.g., a BMP-6 polypeptide that includes amino acids 374-513 of SEQ ID NO:4, amino acids 382-513 of SEQ ID NO:4, amino acids 388-513 of SEQ ID NO:4, or amino acids 412-513 of SEQ ID NO:4.
  • the polypeptide can be a recombinant polypeptide.
  • the compound is a BMP-7 polypeptide, e.g., human BMP-7, e.g., a mature BMP-7 polypeptide, e.g., a BMP-7 polypeptide that includes amino acids 293-431 of SEQ ID NO:5.
  • the polypeptide can be a recombinant polypeptide.
  • the compound is a nucleic acid encoding a BMP-2, -4, -5, -6, and/or -7 polypeptide, or a biologically active fragment or analog thereof.
  • a BMP nucleic acid can include: a BMP-2, -4, -5, -6, and/or -7 coding region; a promoter sequence, e.g., a promoter sequence from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene; an enhancer sequence; untranslated regulatory sequences, e.g., a 5′ untranslated region (UTR), e.g., a 5′UTR from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene, a 3′ UTR, e.g., a 3′UTR from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene; a polyadenylation site; an
  • the level of BMP-2, -4, -5, -6, and/or -7 protein is increased by increasing the level of expression of an endogenous BMP-2, -4, -5, -6, and/or -7 gene, e.g., by increasing transcription of the BMP-2, -4, -5, -6, and/or -7 gene or increasing BMP-2, -4, -5, -6, and/or -7 mRNA stability.
  • transcription of the BMP-2, -4, -5, -6, and/or -7 gene is increased by: altering the regulatory sequence of the endogenous BMP-2, -4, -5, -6, and/or -7 gene, e.g., by the addition of a positive regulatory element (such as an enhancer or a DNA-binding site for a transcriptional activator); the deletion of a negative regulatory element (such as a DNA-binding site for a transcriptional repressor) and/or replacement of the endogenous regulatory sequence, or elements therein, with that of another gene, thereby allowing the coding region of the BMP-2, -4, -5, -6, and/or -7 gene to be transcribed more efficiently.
  • a positive regulatory element such as an enhancer or a DNA-binding site for a transcriptional activator
  • a negative regulatory element such as a DNA-binding site for a transcriptional repressor
  • the nucleic acid encodes or increases transcription of BMP-7.
  • the methods include contacting, administering or expressing one or more other compounds in addition to the BMP, e.g., peroxisome proliferator-activated receptor gamma (PPAR ⁇ ), Retinoid X receptor, alpha (RxR ⁇ ), insulin, T3, a thiazolidinedione (TZD), retinoic acid, another BMP protein (e.g., BMP-1 or BMP-3), vitamin A, retinoic acid, insulin, glucocorticoid or agonist thereof, Wingless-type (Wnt), e.g., Wnt-1, Insulin-like Growth Factor-1 (IGF-1), or other growth factor, e.g., Epidermal growth factor (EGF), Fibroblast growth factor (FGF), Transforming growth factor (TGF)- ⁇ , TGF- ⁇ , Tumor necrosis factor alpha (TNF ⁇ ), Macrophage colony stimulating factor (MCSF), Vascular endothelial growth factor (VEGF) and/
  • the compound can be a BMP-2, -4, -5, -6, and/or -7 protein as described herein or a portion thereof linked with a heterologous polypeptide sequence, e.g., a second BMP protein, to form a chimeric molecule or fusion protein.
  • the methods include administering the compound in combination with a second treatment, e.g., a second treatment for obesity or a related disorder such as diabetes.
  • the second treatment can be insulin, orlistat, phendimetrazine, and/or phentermine.
  • the subject is a mammal.
  • the subject is a human subject, e.g., an obese human subject.
  • the subject is a non-human mammal, e.g., an experimental animal, a companion animal, or a food animal, e.g., a cow, pig, or sheep that is raised for food.
  • a BMP polypeptide or nucleic acid is used, the polypeptide or nucleic acid will be from the same species as the subject, e.g., human, cat, dog, cow, pig, or sheep.
  • the methods include evaluating the subject for one or more of: weight, adipose tissue stores, adipose tissue morphology, insulin levels, insulin metabolism, glucose levels, thermogenic capacity, and cold sensitivity.
  • the evaluation can be performed before, during, and/or after the administration of the compound. For example, the evaluation can be performed at least 1 day, 2 days, 4, 7, 14, 21, 30 or more days before and/or after the administration.
  • the methods include one or more additional rounds of implantation of BMP-activated mesenchymal stem cells, e.g., to increase brown adipogenesis, e.g., to maintain or further reduce obesity in the subject.
  • the invention features a population of BMP-activated stem cells, e.g., pluripotent mesenchymal stem cells.
  • the cells are genetically engineered to express increased levels of a BMP-2, -4, -5, -6, and/or -7 polypeptide, e.g., a BMP-2, -4, -5, -6, and/or -7 polypeptide described herein, either stably or transiently.
  • the cells can be, e.g., cultured mammalian cells, e.g., human cells.
  • the cells are genetically engineered to express at least one other protein as well, e.g., a non-BMP-2, -4, -5, -6, and/or -7 polypeptide, and/or a second (or more) BMP protein.
  • the expressed BMP-2, -4, -5, -6, and/or -7 polypeptide will generally be of the same species as the stem cells, e.g., a human BMP expressed in human cells.
  • the cells are immortalized, e.g., capable of self-renewal indefinitely in culture.
  • the cells used in the methods and compositions described herein express one or more BMP receptors, e.g., type I or II BMP receptors.
  • FIG. 1A is a bar graph illustrating mRNA levels of necdin, Wnt-10a, Pref-1, Gjb3, and PPAR ⁇ in cells treated with BMP-7 after 24 or 72 hours.
  • FIG. 1B is a bar graph illustrating mRNA levels of PGC ⁇ in cells treated with BMP-7 after 24 or 72 hours.
  • FIG. 2D is a bar graph illustrating the effect of administration of a BMP-7 adenoviral vector (black bars) or Lac Z adenoviral control construct (open bars) on UCP-1 gene expression as measured by Q-RT-PCR.
  • FIGS. 2E and 2F are bar graphs illustrating the effect of administration of a BMP-7 adenoviral vector (black bars) or Lac Z adenoviral control construct (open bars) on oxygen consumption VO 2 in 4-week old and 12-week old mice for light ( 2 E) and dark ( 2 F) cycles.
  • FIGS. 3A-3D are photomicrographs of sections of tissue showing the morphology of normal brown fat ( 3 A), normal white fat ( 3 B), implant-derived tissue with a brown fat-type morphology ( 3 C), and implant-derived tissue with a partially white fat-type morphology ( 3 D).
  • FIGS. 4A and 4B are bar graphs illustrating the effect of implantation of BMP-7 treated pluripotent mesenchymal stem cells on blood glucose ( 4 A) and blood insulin levels ( 4 B).
  • FIGS. 5A and 5B are each six panels of bar graphs illustrating expression levels of PGC-1 ⁇ , NRF-1, Tfam, PGC-1 ⁇ , NRF-2, and Cyto C after 3 days ( 5 A) or 8 days ( 5 B) of treatment with Insulin and T3 (light gray bars), Insulin, T3 and BMP-6 (mid gray bars), or Insulin, T3, and BMP-7 (black bars), as compared to control (white bars) in brown preadipocytes (left group of four bars in each panel) and white preadipocytes (right group of four bars in each panel).
  • FIGS. 5C and 5D are each pairs of photomicrographs of brown preadipocytes ( 5 C) and 3T3-L1 cells ( 5 D) treated with Insulin and T3 (left panel of each) or Insulin, T3 and BMP-7 (right panels of each), showing an increased number and size of mitochondria in the BMP-7 treated cells.
  • BMPs are members of the transforming growth factor- ⁇ superfamily that are involved in multiple key steps of embryonic development as well as throughout life (Kishigami and Mishina, Cytokine. Growth Factor. Rev. 16:265-278 (2005); Chen et al., Growth Factors. 22:233-241 (2004); Yamamoto and Oelgeschlager, Naturwissenschaften 91:519-534 (2004)). BMPs have been shown to play a role in two different stages of adipocyte development. First, BMP-2 and 4 stimulate differentiation of multipotent mesenchymal cells and bone marrow stromal cells into adipocytes under appropriate conditions (Butterwith et al., Biochem.
  • BMPs also stimulate the differentiation of committed white preadipocytes (Sottile and Seuwen, FEBS Lett. 475:201-204 (2000); Rebbapragada et al., Mol. Cell. Biol. 23:7230-7242 (2003)).
  • BMP-2, -4, -5, -6, and -7 are involved in adipocyte differentiation, and treatment of stem cells with BMP-2, -4, -5, -6, and/or -7 promotes brown adipogenesis.
  • BMP-2, -4, -5, -6, and/or -7 are thus therapeutic, diagnostic and drug discovery targets for adipose-related disorders, such as obesity and related disorders such as diabetes, insulin resistance, hyperglycemia, hyperlipidemia, and hypercholesterolemia.
  • the methods described herein include implanting a population of BMP-activated stem cells as described herein into a subject.
  • Some of the methods described herein include implanting stem cells that have been treated with an agent that increases BMP signalling.
  • the methods include treating (e.g., contacting) stem cells, e.g., pluripotent mesenchymal stem cells, with the compound in an amount sufficient to increase BMP signalling, and thereafter implanting the BMP-activated cells (e.g., at least one cell or a population of such cells) in a subject.
  • Suitable agents can be the BMPs themselves, e.g., recombinant proteins, or nucleic acids that encode the BMPs, to treat the stem cells.
  • treating the cells includes genetically engineered the cells in vitro to express a BMP-2, -4, -5, -6, and/or -7 polypeptide.
  • the cells are then administered to a subject.
  • Populations of such genetically engineered stem cells are also included within the scope of the present invention.
  • Other compounds are described herein.
  • Stem cells are progenitor cells that are capable of both self-renewal and differentiation into many different cell lineages. Suitable stem cells, and methods for isolating them, are known in the art. Stem cells can be pluripotent or totipotent. Embryonic or adult stem cells can be used. Embryonic stem cells are generally derived from embryos that are less than a week old, e.g., in vitro fertilized embryos.
  • Adult stem cells can include hematopoietic stem cells, generally isolated from bone marrow, peripheral blood, or umbilical cord blood; mesenchymal stem cells, e.g., from bone marrow or periosteum; intestinal (gut) stem cells, from the small intestine; skin stem cells; neuronal stem cells; and hepatic stem cells (oval cells), from the liver.
  • mesenchymal stem cells e.g., from bone marrow or periosteum
  • intestinal stem cells from the small intestine
  • skin stem cells e.g., skin stem cells
  • neuronal stem cells hepatic stem cells (oval cells)
  • primary pluripotent mesenchymal stem cells can be isolated from bone marrow (see, e.g., Halleux et al., J. Musculoskelet. Neuronal. Interact. 2(1):71-6 (2001)), connective tissue (Young et al., Dev Dyn. 202(2):137-44 (1995)), and other tissues.
  • the stem cells are isolated from an adipose tissue.
  • the cells are purified, e.g., a population of cells in which at least 60%, e.g., 70%, 80%, 90% or more of the cells are stem cells.
  • a purified population of stem cells is enriched by any method known in the art for cell enrichment, e.g., immunomagnetic cell sorting, fluorescence activated cell sorting (FACS), adherence to tissue culture plates and flasks, or culturing under conditions that favor the growth of the desired stem cells. Such methods are known in the art.
  • primary cell includes cells present in a suspension of cells isolated from a mammalian tissue source (prior to their being plated, i.e., attached to a tissue culture substrate such as a dish or flask), cells present in an explant derived from tissue, both of the previous types of cells plated for the first time, and cell suspensions derived from these plated cells.
  • tissue culture substrate such as a dish or flask
  • secondary cell or “cell strain” refers to cells at all subsequent steps in culturing. Secondary cells are cell strains that consist of secondary cells that have been passaged one or more times.
  • Primary and secondary stem cells can be obtained from a variety of tissues and include cell types which can be maintained and propagated in culture. Primary cells are preferably obtained from the individual to whom the BMP-activated cells are administered. However, primary cells can also be obtained from a donor (e.g., an individual other than the recipient, typically of the same species, preferably ab immunologically compatible individual). Methods for obtaining and culturing such cells are known in the art.
  • the methods can include allowing stem cells to undergo sufficient number doubling to produce either a clonal cell strain or a heterogeneous cell strain of desired size, e.g., a sufficient number to provide a therapeutic effect to an individual, or a sufficient number to establish a stable cell line, before or after BMP-activation.
  • the cells can be cultured for a time in the absence of the BMP, then in the presence of the BMP for a time (e.g., 1, 2, 3 or more days) before implantation into the subject.
  • the cells can be washed (e.g., in isotonic PBS) before implantation to remove any contaminants, including BMPs or components of growth media, before implantation.
  • the number of required cells is variable and depends on a variety of factors, including but not limited to, the use of the transfected cells, the functional level of the exogenous DNA in the transfected cells, the site of implantation of the transfected cells (for example, the number of cells that can be used is limited by the anatomical site of implantation), and the age, surface area, and clinical condition of the patient.
  • the population of BMP-activated stem cells includes at least 10 7 , 10 8 , 10 9 , or more cells.
  • BMP-activated stem cells are stem cells that have an enhanced level of BMP signalling, e.g., BMP-2, -4, -5, -6, and/or -7 signalling, wherein the level of BMP signalling has been increased by direct human intervention.
  • BMP signalling can be enhanced in the cells by any method known in the art or described herein, e.g., by treating the cell with a compound that enhances BMP signalling as described herein, e.g., a BMP polypeptide or nucleic acid.
  • a pharmaceutically acceptable carrier e.g., for storage or implantation.
  • the language “pharmaceutically acceptable carrier” is intended to include any and all solvents, media, antibacterial and antifungal agents, isotonic agents, and the like, compatible with pharmaceutical administration and viability of the cells. In general, the cells will be maintained in a sterile state.
  • the use of such media and agents for pharmaceutically active substances are known. Except insofar as any conventional media or agent is incompatible with the active compound, such media can be used in the compositions of the invention. Supplementary active compounds can also be incorporated into the compositions.
  • the cells can be autologous, allogeneic or xenogeneic.
  • methods described herein can include obtaining a population of stem cells from a subject, optionally culturing and/or enriching the stem cells to obtain a purified population of stem cells, treating the cells with an agent that enhances BMP signalling as described herein to activate the cells, and implanting the cells in the same subject from which they were removed.
  • the cells are allogeneic or xenogeneic; if necessary, immune suppression can be administered to prevent rejection of the cells.
  • BMP proteins have been used in the clinic in the treatment of bone and cartilage disorders or wounds. The effective clinical use of recombinant BMPs is discussed in Einhorn, J. Bone and Joint Surgery 85A:82-88 (2003), and Sandhu, Spine 28(15):S64-73 (2003).
  • a BMP polypeptide e.g., a mature BMP polypeptide
  • BMPs are small secreted proteins that are internalized into their target cells where they exert their activity.
  • homologous proteins from that species can also be used, e.g., cow, pig, sheep, or goat. Such homologous proteins can be identified, e.g., using methods known in the art, e.g., searching available databases for homologs identified in the target species, e.g., the homologene database.
  • BMP-2 is 396 amino acids in length, localized to chromosome 20p12 in human. The nucleotide and amino acid sequences of human BMP-2 are disclosed in Wozney et al., Science 242(4885):1528-1534 (1988).
  • BMP2 belongs to the transforming growth factor-beta (TGF ⁇ ) superfamily. Bone morphogenetic protein induces bone formation, and BMP2 is a candidate gene for the autosomal dominant disease of fibrodysplasia (myositis) ossificans progressive. Bone morphogenetic protein 2 regulates myogenesis through dosage-dependent PAX3 expression in pre-myogenic cells, and is expressed in mesoderm under SHM control through the SOX9.
  • TGF ⁇ transforming growth factor-beta
  • Amino acids 38-268 are the TGF ⁇ propeptide domain
  • 291-396 are the TGF ⁇ family N-terminal domain
  • Amino acids 283-396 are the mature peptide. The sequence is set forth in Wozney et al., Science 242:1528-1534 (1988).
  • BMP-4 induces cartilage and bone formation, and is important in mesoderm induction, tooth development, limb formation and fracture repair.
  • the sequence of the BMP-4 preproprotein is shown below.
  • Amino acids 41-276 are the TGF ⁇ propeptide domain, and 302-408 are the TGF ⁇ family N-terminal domain.
  • Amino acids 293-408 are the mature peptide. The sequence is set forth in Wozney et al., Science 242:1528-1534 (1988).
  • the mature form of BMP-4 contains four potential N-linked glycosylation sites per polypeptide chain.
  • the BMP-5 preproprotein is a 454 amino acid protein, as shown below. BMP-5 induces cartilage and bone formation. The sequence is set forth in Celeste et al., Proc. Natl. Acad. Sci. U.S.A., 87, 9843-9847, 1990.
  • the mature BMP-5 protein is believed to be amino acids 323-454 of SEQ ID NO:3, and has four potential N-linked glycosylation sites per polypeptide chain, and four potential disulfide bridges. See UniProt Accession Nos. P22003; Q9H547; or Q9NTM5; HomoloGene: 22412.
  • BMP-6 is an autocrine stimulator of chondrocyte differentiation, and is involved in the development of embryonic neural, and urinary systems, as well as growth and differentiation of liver and keratinocytes. BMP-6 knockout mice are viable and show a slight delay in ossification of the sternum. BMP-6 (precursor) is a 57 kD protein, 513 amino acids in length, localized to chromosome 6p24 in human. The nucleotide and amino acid sequence of human BMP-6 is disclosed in U.S. Pat. No. 5,187,076.
  • BMP-6 is predicted to be synthesized as a precursor molecule which is cleaved to yield a 132 amino acid mature polypeptide with a calculated molecular weight of approximately 15 Kd.
  • the mature form of BMP-6 contains three potential N-linked glycosylation sites per polypeptide chain.
  • the active BMP-6 protein molecule is likely a dimer. Processing of BMP-6 into the mature form involves dimerization and removal of the N-terminal region in a manner analogous to the processing of the related protein TGF ⁇ (Gentry et al., Molec. Cell. Biol. 8:4162 (1988); Dernyck et al., Nature 316:701 (1985)).
  • the human BMP-6 precursor is shown below.
  • the mature polypeptide is believed to include amino acids 374-513 of SEQ ID NO:4.
  • Other active BMP-6 polypeptides include polypeptides including amino acids 382-513, 388-513 and 412-513 of SEQ ID NO:4.
  • the human BMP-6 promoter has been characterized (See Tamada et al., Biochim Biophys Acta. 1998, 1395(3):247-51), and can be used in methods described herein. See UniProt Accession No. P22004; HomoloGene:1300.
  • BMP-7 also belongs to the TGF ⁇ superfamily. It induces cartilage and bone formation, and may be the osteoinductive factor responsible for the phenomenon of epithelial osteogenesis. BMP-7 plays a role in calcium regulation and bone homeostasis, and in the regulation of anti-inflammatory response in the adult gut tissue. The sequence of BMP-7 is shown below:
  • Amino acids 1-29 are a potential signal sequence; 30-431 are the prepropeptide, and 293-431 are the mature protein.
  • the mature form of BMP-7 contains four potential N-linked glycosylation sites per polypeptide chain, and four potential disulfide bridges. See UniProt Accession No. P 18075; HomoloGene: 20410.
  • Modifications can be made to a protein compound that result in pharmacokinetic properties of the protein which are desirable for use in protein therapy. For example, such modifications can result in an increase in cellular uptake, circulatory half-life, rate of clearance and reduced immunogenicity.
  • a protein compound e.g., a compound described herein such as a BMP-2, -4, -5, -6, and/or -7 polypeptide.
  • the choice of expression system can influence pharmacokinetic characteristics. Differences between expression systems in post-translational processing can lead to recombinant proteins of varying molecular size and charge, which can affect, for example, cellular uptake, circulatory half-life, rate of clearance and immunogenicity.
  • the pharmacokinetic properties of the protein may be optimized by the appropriate selection of an expression system, such as selection of a bacterial, viral, or mammalian expression system.
  • Exemplary mammalian cell lines useful in expression systems for therapeutic proteins are Chinese hamster ovary, (CHO) cells, the monkey COS-1 cell line and the CV-1 cell line.
  • a protein can be chemically altered to enhance the pharmacokinetic properties while maintaining activity.
  • the protein can be covalently linked to a variety of moieties, altering the molecular size and charge of the protein and consequently its pharmacokinetic characteristics.
  • the moieties are preferably non-toxic and biocompatible.
  • polyethylene glycol (PEG) can be covalently attached to the protein (PEGylation). See, e.g., Poly ( ethylene glycol ): Chemistry and Biological Applications , Harris and Zalipsky, eds., ACS Symposium Series, No. 680, 1997; Harris et al., Clinical Pharmacokinetics 40:7, 485-563 (2001)).
  • the protein can be similarly linked to oxidized dextrans via an amino group. (See Sheffield, Current Drug Targets—Cardiovas. and Haemat. Dis. 1:1, 1-22 (2001)).
  • the protein compounds can be chemically linked to another protein.
  • the protein can be cross-linked carrier protein to form a larger molecular weight complex with improved cellular uptake.
  • the carrier protein can be a serum protein, such as albumin.
  • the protein can be attached to one or more albumin molecules via a bifunctional cross-linking recompound.
  • the cross-linking recompound may be homo- or heterofunctional.
  • the protein can cross-link with itself to form a homodimer, trimer, or higher analog. Again, either heterobifunctional or homobifunctional cross-linking recompounds can be used to form the dimers or trimers. (See Stykowski et al., Proc. Natl. Acad. Sci. USA, 95, 1184-1188 (1998)).
  • the stem cells of mammalian origin can be, for example, activated by transfection with an exogenous nucleic acid that includes a heterologous nucleotide sequence, e.g., encoding BMP-2, -4, -5, -6, and/or -7, or an agonist thereof, with or without a nucleotide sequence encoding a signal peptide, and produce the encoded product either transiently or stably, over extended periods of time.
  • a heterologous amino acid can also be a regulatory sequence, e.g., a promoter, which causes expression, e.g., constitutive or inducible expression or upregulation, of an endogenous BMP-2, -4, -5, -6, and/or -7 sequence.
  • An exogenous nucleic acid sequence can be introduced into a primary or secondary cell by homologous recombination as described, for example, in U.S. Pat. No. 5,641,670, the contents of which are incorporated herein by reference.
  • the transfected cells can also include DNA encoding a selectable marker that confers a selectable phenotype upon them, facilitating their identification and isolation.
  • the compound that enhances BMP signalling as described herein includes, e.g., a BMP nucleic acid, e.g., a BMP-2, -4, -5, -6, and/or -7 encoding sequence or active fragment thereof, and any of: a promoter sequence, e.g., a promoter sequence from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene; an enhancer sequence, e.g., 5′ untranslated region (UTR), e.g., a 5′ UTR from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene, a 3′ UTR, e.g., a 3′ UTR from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene; a polyadenylation site; an insulator sequence; or another sequence that enhances the expression of BMP-2, -4,
  • nucleic acids described herein e.g., a nucleic acid encoding a BMP-2, -4, -5, -6, and/or -7 polypeptide as described herein, can be incorporated into a gene construct.
  • the methods described herein can use such expression vectors for in vitro transfection and expression of a BMP-2, -4, -5, -6, and/or -7 polypeptide described herein in particular cell types, e.g., stem cells, e.g., pluripotent mesenchymal stem cells.
  • Expression constructs of such components can be administered in any biologically effective carrier, e.g., any formulation or composition capable of effectively delivering the component gene to cells in vivo.
  • Approaches include insertion of a subject gene in viral vectors including recombinant retroviruses, adenovirus, adeno-associated virus, and herpes simplex virus-1, or recombinant bacterial or eukaryotic plasmids.
  • Viral vectors transfect cells directly, and infection of cells with a viral vector generally has the advantage that a large proportion of the targeted cells can receive the nucleic acid. Additionally, molecules encoded within the viral vector, e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells which have taken up viral vector nucleic acid. Retroviral vectors, adenovirus-derived vectors, and adeno-associated virus vectors can also be used as a recombinant gene delivery system for the transfer of exogenous genes. These vectors provide efficient delivery of genes into cells, and the transferred nucleic acids are generally stably integrated into the chromosomal DNA of the host.
  • Non-viral methods can also be employed to cause expression of an nucleic acid compound described herein (e.g., a BMP-2, -4, -5, -6, and/or -7 polypeptide encoding nucleic acid) into a cell.
  • an nucleic acid compound described herein e.g., a BMP-2, -4, -5, -6, and/or -7 polypeptide encoding nucleic acid
  • Most nonviral methods of gene transfer rely on normal mechanisms used by mammalian cells for the uptake and intracellular transport of macromolecules.
  • non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject gene by the targeted cell.
  • Plasmid DNA can be delivered with the help of, for example, cationic liposomes (e.g., LIPOFECTINTM) or derivatized (e.g., antibody conjugated), polylysine conjugates, gramicidin S, artificial viral envelopes or other such intracellular carriers, as well as direct injection of the gene construct or CaPO 4 precipitation.
  • cationic liposomes e.g., LIPOFECTINTM
  • derivatized e.g., antibody conjugated
  • polylysine conjugates e.g., antibody conjugated
  • gramicidin S e.g., gramicidin S
  • artificial viral envelopes or other such intracellular carriers as well as direct injection of the gene construct or CaPO 4 precipitation.
  • Other embodiments include plasmid injection systems such as are described in Meuli et al., J. Invest. Dermatol. 116(1):131-135 (2001); Cohen et al., Gene Ther 7(22):1896-905
  • the BMP compounds described herein can be formulated in any suitable manner, e.g., in a carrier system, for use in contacting with the populations of cells.
  • the carrier can be a colloidal system.
  • the colloidal system can be liposome, a phospholipid bilayer vehicle.
  • the protein is encapsulated in a liposome while maintaining protein integrity.
  • there are a variety of methods to prepare liposomes See Lichtenberg et al., Methods Biochem Anal, 33:337-462 (1988), LIPOSOME TECHNOLOGY Anselem et al., CRC Press, 1993).
  • Liposomes can be prepared from an assortment of phospholipids varying in size and substitution, and may also contain additional components with low toxicity, such as cholesterol.
  • the liposome can be formulated and isolated in a variety of shapes and sizes.
  • moieties may attached to the surface of the liposome to further enhance the pharmacokinetic properties of the carrier.
  • the moieties may be attached to phospholipid or cholesterol molecules, and the percentage of the moiety incorporated on the surface may be adjusted for optimal liposome stability and pharmacokinetic characteristics.
  • One embodiment comprises a liposome with poly-ethylene glycol (PEG) added to the surface.
  • PEG poly-ethylene glycol
  • Liposomal formulations can delay clearance and increase cellular uptake. (See Reddy, Annals of Pharmacotherapy, 34(7/8):915-923 (2000)).
  • the carrier can also be a polymer, e.g., a biodegradable, biocompatible polymer matrix.
  • the protein can be embedded in the polymer matrix while maintaining protein integrity.
  • the polymer may be natural, such as polypeptides, proteins or polysaccharides, or synthetic, such as poly( ⁇ -hydroxy) acids. Examples include carriers made of e.g., collagen, fibronectin, elastin, cellulose acetate, cellulose nitrate, polysaccharide, fibrin, gelatin, and combinations thereof.
  • the polymer is poly-lactic acid (PLA) or co-polylactic/glycolic acid (PGLA).
  • the polymeric matrices can be prepared and isolated in a variety of forms and sizes, including microspheres and nanospheres. Polymer formulations can lead to prolonged duration of therapeutic effect. (See Reddy, Annals of Pharmacotherapy, 34(7/8):915-923 (2000)). A polymer formulation for human growth hormone (hGH) has been used in clinical trials. (See Kozarich and Rich, Chemical Biology 2:548-552 (1998)).
  • hGH human growth hormone
  • polymer microsphere sustained release formulations are described in PCT publication WO 99/15154 (Tracy et al.), U.S. Pat. Nos. 5,674,534 and 5,716,644 (both to Zale et al.), PCT publication WO 96/40073 (Zale et al.), and PCT publication WO 00/38651 (Shah et al.).
  • U.S. Pat. Nos. 5,674,534 and 5,716,644 and PCT publication WO 96/40073 describe a polymeric matrix containing particles of erythropoietin that are stabilized against aggregation with a salt.
  • Methods described herein can include implanting a population of BMP-activated stem cells, e.g., as described herein, into a subject to be treated, wherein said population of BMP-activated stem cells, or their progeny (i.e., daughter cells), undergo brown adipogenesis. Once implanted, the stem cells will generally undergo adipogenesis, generating BAT in the subject.
  • These cell therapy methods are useful, e.g., for the treatment of obesity and insulin resistance in a subject, or for treating a disease associated with a lack of mitochondria, e.g., diabetes, cancer, neurodegeneration, and aging.
  • the delivery system can include a reservoir containing a population of BMP-activated pluripotent mesenchymal stem cells, and a needle in fluid communication with the reservoir.
  • the population of BMP-activated stem cells will be in a pharmaceutically acceptable carrier, with or without a scaffold, matrix, or other implantable device to which the cells can attach (examples include carriers made of, e.g., collagen, fibronectin, elastin, cellulose acetate, cellulose nitrate, polysaccharide, fibrin, gelatin, and combinations thereof).
  • Such delivery systems are also within the scope of the invention. Generally, such delivery systems are maintained in a sterile manner.
  • Various routes of administration and various sites e.g., renal sub capsular, subcutaneous, central nervous system (including intrathecal), intravascular, intrahepatic, intrasplanchnic, intraperitoneal (including intraomental), intramuscularly implantation
  • the cells will be implanted into the subject subcutaneously.
  • the population of BMP-activated stem cells that is implanted includes at least 10 7 , 10 8 , 10 9 , or more cells.
  • an immunosuppressive compound e.g., a drug or antibody
  • an immunosuppressive compound can be administered to the recipient subject at a dosage sufficient to achieve inhibition of rejection of the cells.
  • Dosage ranges for immunosuppressive drugs are known in the art. See, e.g., Freed et al., N. Engl. J. Med. 327:1549 (1992); Spencer et al., N. Engl. J. Med. 327:1541 (1992); Widner et al., N. Engl. J. Med. 327:1556 (1992)).
  • Dosage values may vary according to factors such as the disease state, age, sex, and weight of the individual.
  • Adipose tissue plays an active role in energy balance.
  • Two functionally different types of fat tissue are present in mammals: white adipose tissue, which is the primary site of triglycerides storage and release of fatty acids, and brown adipose tissue, which is specialized for energy expenditure via thermogenesis.
  • white adipose tissue which is the primary site of triglycerides storage and release of fatty acids
  • brown adipose tissue which is specialized for energy expenditure via thermogenesis.
  • the regulation of differentiation and balance of function between the two types of adipose tissue is critical to whole body energy homeostasis.
  • the role of BMPs in this regulation was explored in the experiments described herein.
  • BMPs pluripotent C3H10T1/2 mesenchymal stem cells
  • the cells were treated with BAT induction cocktail (1 nM T3, 20 nM insulin, 0.5 mM isobutylmethylxanthine (IBMX), 0.5 mM dexamethazone, and 0.125 mM indomethacin) or WAT induction cocktail (1 nM T3, 0.5 mM isobutylmethylxanthine (IBMX), 0.5 mM dexamethazone, and 0.125 mM indomethacin).
  • BAT induction cocktail (1 nM T3, 20 nM insulin, 0.5 mM isobutylmethylxanthine (IBMX), 0.5 mM dexamethazone, and 0.125 mM indomethacin
  • WAT induction cocktail 1 nM T3, 0.5 mM isobutylmethylxanthine (IBMX), 0.5 mM dexamethazone, and 0.125 mM indomethacin.
  • BMP-2,4,6, and 7, but not BMP-3 bone morphogenetic protein-2,4,6, and 7, but not BMP-3
  • UCP-1 brown fat specific marker uncoupling protein-1
  • PPAR ⁇ peroxisome proliferator-activated receptor gamma
  • FES fatty acid synthase
  • BMP-6 and BMP-7 in combination with hormone induction cocktail and rosiglitazone produced similar effects on a mouse embryonic fibroblast (MEF) cell line generated using the 3T3 protocol 41, with increased lipid accumulation and expression of PPAR ⁇ , FAS, and the brown fat specific protein UCP-1 in these cells.
  • MEF mouse embryonic fibroblast
  • BMP-6 and -7 can not only trigger commitment of mesenchymal stem cells to a brown adipocyte lineage, but also act in concert with other differentiating agents to induce characteristics of brown fat, including UCP-1, in more primitive fibroblastic cells.
  • stromo-vascular fraction (SVF) cells were isolated from interscapular BAT and nearby subcutaneous WAT close to BAT, and induced to undergo adipocyte differentiation in a serum free differentiation medium containing transferrin, dexamethasone, insulin and T3 supplemented with rosiglitazone (1 ⁇ g/ml), BMP-7 (3.3 nM) or vehicle. While rosiglitazone induced similar levels of adipocyte differentiation in SVFs derived from both fat depots within 3 days, BMP-7 had a specific effect on induction of differentiation only in the brown precursor cells at this time point. Thus, the effect of BMP-7 on promoting brown adipogenesis was not due to immortalization or some other factor unique to the brown preadipocyte cell lines, but occurred even in primary brown preadipocytes or their precursors present in the stromo-vascular factions.
  • adenoviral constructs for the expression of BMP-7 or LacZ (as a control) 5 ⁇ 10 8 plaque-forming units per gram body weight via tail veins were injected into 4-week and 12-week old C57BL/6 mice via the tail vein. These mice were sacrificed 15 days after adenoviral injection. Various adipose depots were collected and weighed.
  • adenoviral-mediated expression of BMP-7 in C57BL/6 mice results in a significant increase in brown, but not white, fat mass relative to total body weight and energy expenditure.
  • adenoviral-mediated expression of BMP-7 in vivo leads to an increase in energy expenditure.
  • the BMP-7 adenovirally-treated mice showed a significant increase in energy expenditure in both light and dark cycles ( FIGS. 2E and 2F ).
  • BAT mass in 4-week old mice was not due to an increase in the size of the brown fat cells, but to an increase in cell number (data not shown), consistent with the hypothesis that BMP-7 can act as a growth and differentiation factor on the brown fat precursor cells. This could involve recruitment of pluripotent mesenchymal stem cells and/or promoting differentiation of existing committed preadipocytes present in the SVF, as suggested by the experiments described above.
  • UCP-1 expression in BAT in 12-week old animals suggests that BMPs can directly increase thermogenic function of mature brown adipocytes in adult mammals.
  • C3H10T1/2 cells were treated with 3.3 nM recombinant BMP-7 for 3 days, and then injected into athymic mice.
  • 1.5 ⁇ 10 7 BMP-7 treated cells were injected subcutaneously into the sternum/thoracic regions of 5-w old BALB/c athymic mice; this region was chosen because it is generally free of any fat deposits. Mice were sacrificed 6 weeks after implantation.
  • PGC-1 ⁇ stimulates expression of nuclear respiratory factor (NRF)-1 and NRF-2, and coactivates the transcriptional function of these factors on expression of mitochondrial transcription factor A (Tfam), which is a direct regulator of mitochondrial replication and transcription (Wu et al., 1999, supra).
  • NRF nuclear respiratory factor
  • Tfam mitochondrial transcription factor A
  • BMP-6 and BMP-7 caused a 50% reduction in PGC-1 ⁇ and NRF-2 gene expression and had no effect on expression of PGC-1 ⁇ , NRF-1, Tfam and Cyto C.
  • expression of genes involved in mitochondrial biogenesis in brown preadipocytes was further increased by BMP-6 and BMP-7 ( FIG. 5B ).
  • BMP-6 also induced some increase in expression of PGC-1 ⁇ , PGC-1 ⁇ , Tfam and Cyto C in 3T3-L1 white preadipocytes, but with no effect on UCP-1.
  • BMP-6 and BMP-7 have major effects to stimulate differentiation, mitochondrial biogenesis, and UCP-1 expression in brown preadipocytes, but only have minimal effect in 3T3-L1 white preadipocytes; and have no effect on induction of UCP-1 expression in the latter cells.

Abstract

Methods and compositions for treating obesity and related disorders. The methods include the use of stem cells treated with BMP-2, -4, -5, -6 and/or -7.

Description

    CLAIM OF PRIORITY
  • This application claims the benefit under 35 USC §119(e) of U.S. Provisional Patent Application Ser. No. 60/686,542, filed on Jun. 1, 2005, the entire contents of which are hereby incorporated by reference.
  • FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • The U.S. Government has certain rights in this invention pursuant to Grants No. DK63696, DK33201, R21DK70722, and P30 DK46200-13, awarded by the National Institutes of Health.
  • BACKGROUND
  • Obesity, and disorders associated with obesity such as diabetes, are a major global health concern. Obesity, which is generally associated with an abnormal accumulation of fat cells, develops when energy intake exceeds energy expenditure. Adipose tissues play an important role in obesity, insulin resistance and diabetes. Two functionally different types of fat tissues are present in mammals: white adipose tissue (WAT), which is the primary site of depot of triglycerides and release of fatty acids, and brown adipose tissue (BAT), which is specialized in thermogenic energy expenditure through the expression of uncoupling protein-1 (UCP-1).
  • The most commonly known fat cells are white fat cells, also known as white adipose tissue (WAT) cells, which have a thin ring of cytoplasm surrounding a lipid or fat droplet. WAT is found underneath the skin and provides heat insulation, cushioning against shock and jarring, and energy reserves. An average lean person has roughly 20 to 40 billion WAT cells. An obese person can have up to ten times more WAT than the average lean person.
  • The less common fat cells are the brown fat cells, also known as brown adipose tissue (BAT) cells. Energy expenditure for thermogenesis in BAT serves either to maintain body temperature in the cold or to waste food energy. It has roles in thermal balance and energy balance, and when defective, is usually associated with obesity. BAT is typically atrophied in obese animals. The importance of BAT in overall energy homeostasis is underscored by the finding that ablation of BAT in mice results in severe obesity accompanied by insulin resistance, hyperglycemia, hyperlipidemia, and hypercholesterolemia (Lowell at al., Nature 366(6457):740-2 (1993); Hamann et al., Diabetes. 44(11):1266-73 (1995); Hamann et al., Endocrinology 137(1):21-9 (1996). Increasing the relative proportion and function of BAT may increase whole body energy expenditure, preventing the development of obesity. In fact, the role of BAT as a defense against obesity has been clearly demonstrated through targeted ablation of this tissue in mice and the BAT-less mice become more susceptible to diet-induced obesity, diabetes, and hyperlipidemia (Lowell et al., Nature 366:740-742 (1993); Hamann et al., Endocrinology 137:21-29 (1996).
  • BAT also features the presence of abundant and large mitochondria (Nedergaard et al., in Brown Adipose Tissue, Trayhurn and Nicholls, Eds. (Edward Arnold, Baltimore, 1986)), which serve as the center site for oxidative phosphorylation, intermediary metabolism, adaptive thermogenesis, generation of reactive oxygen species and apoptosis. In BAT, mitochondrial biogenesis has been long known to accompany brown adipocyte differentiation. During the past decade, it has become increasingly evident that the integrity of mitochondria contribute to a variety of human diseases, including obesity, diabetes, cancer, neurodegeneration, and aging (Duchen, Diabetes 53 (Suppl 1): S96-102 (2004); Taylor and Turnbull, Nat. Rev. Genet. 6:389-402 (2005); Lowell and Shulman, Science 307:384-387 (2005)).
  • Adipose tissues contain a potential mitotic compartment, which can allow for growth and differentiation of WAT or BAT cells. Adipose tissue can be readily assayed using routine techniques. An exemplary assay for adipose cells is the Oil Red O lipophilic red dye assay. The dye is used to stain neutral lipids in cells. The amount of staining is directly proportional to the amount of lipid in the cell and can be measured spectrophotometrically. The amount of lipid accumulation is determined as a parameter of differentiation. WAT and BAT can be distinguished by routine techniques, e.g., morphologic changes specific to WAT or BAT, or evaluation of WAT-specific or BAT-specific markers. For example, BAT cells can be identified by expression of uncoupling protein (UCP), e.g., UCP-1.
  • Bone morphogenetic proteins (BMPs) belong to the TGFβ superfamily. BMPs bind to specific type-I and -II serine/threonine kinase receptor complexes, RIa, RIb, and RII, which signal through SMAD proteins or the p38 mitogen-activated protein kinase (MAPK). The BMPs are important regulators of key events in many aspects of tissue development and morphogenesis, including the processes of bone formation during embryogenesis, postnatal growth, remodeling and regeneration of the skeleton. Localization studies in both human and mouse tissues have demonstrated high levels of mRNA expression and protein synthesis for various BMPs in adipose, heart, lung, small intestine, limb bud and teeth.
  • Several BMPs have been implicated in early skeletal development, including BMP-2, -4, -5, -7, -14 (CDMP-1/GDF-5). Other members, such as BMP-3, -6, -7 and -13 (CDMP-2/GDF-6) may be involved in later stages of skeletal formation.
  • SUMMARY OF THE INVENTION
  • The present invention is based, in part, on the discovery that bone morphogenetic proteins (BMPs) play an important role in adipocyte differentiation. In particular, it has been found that BMPs 2, 4, 5, 6, and 7 markedly induce differentiation of brown preadipocytes, even in the absence of normally required induction cocktails. Furthermore, treatment of stem cells with BMPs triggers commitment of these cells to the brown adipocyte lineage. Implantation of BMP-7-treated stem cells into athymic mice leads to development of these cells into a tissue containing both brown and white adipocytes. In addition, these mice become more insulin-sensitive. Finally, adenoviral-mediated BMP-7 expression in normal mice results in a significant increase in brown fat mass and energy expenditure, and a significant reduction of body weight in diet-induced obese C57BL/6 mice. Since brown adipose tissue (BAT) is specialized for energy expenditure, the methods described herein are useful for the treatment of obesity and related disorders, such as diabetes. The methods can also be used to decrease fat stores in subjects including food animals, e.g., to improve the quality of the meat derived therefrom.
  • Accordingly, in one aspect, the invention features methods of modulating adipose tissue function or development, e.g., promoting BAT adipogenesis, in a subject. The methods include administering to the subject a population of BMP-activated stem cells, e.g., pluripotent mesenchymal stem cells, as described herein, wherein said population of BMP-activated stem cells, or their progeny (i.e., daughter cells), undergo brown adipogenesis.
  • In another aspect, the invention features methods of treating a subject, e.g., decreasing fat stores or weight in a subject such as a human. The methods include administering to the subject a population of BMP-activated stem cells, e.g., pluripotent mesenchymal stem cells, as described herein, wherein said population of BMP-activated stem cells, or their progeny, undergo brown adipogenesis. The methods can optionally include identifying a subject in need of decreasing fat stores or weight.
  • In a further aspect, the invention includes methods of enhancing insulin sensitivity in a subject, e.g., a subject that is insulin-resistant. The methods include administering to the subject a population of BMP-activated stem cells, e.g., pluripotent mesenchymal stem cells, as described herein, wherein said population of BMP-activated stem cells, or their progeny, undergo brown adipogenesis. The methods can optionally include identifying a subject in need of enhanced insulin sensitivity.
  • As used herein, “BMP-activated” means that stem cell has an artificially enhanced level of BMP signalling, e.g., BMP-2, -4, -5, -6, and/or -7 signalling. “Artificially” enhanced means that the level of BMP signalling has been increased by direct human intervention. BMP signalling can be enhanced by any method described herein, e.g., by treating the cell with a compound that enhances BMP signalling as described herein, e.g., a BMP polypeptide or nucleic acid. Populations of stem cells activated by methods described herein are also included within the present invention. The cells can be autologous, allogeneic or xenogeneic.
  • In some embodiments, methods described herein can include treating (e.g., contacting) a population of stem cells, e.g., pluripotent mesenchymal stem cells, with a compound in an amount sufficient to increase BMP signalling, thereby producing a population of BMP-activated cells.
  • In some embodiments, methods described herein can include implanting a population of BMP-activated cells into a subject. The BMP-activated cells can be implanted directly or can be administered in a scaffold, matrix, or other implantable device to which the cells can attach (examples include carriers made of, e.g., collagen, fibronectin, elastin, cellulose acetate, cellulose nitrate, polysaccharide, fibrin, gelatin, and combinations thereof). In general, the methods include implanting a population of BMP-activated cells comprising a sufficient number of cells to promote brown adipogenesis in the subject, e.g., to increase the amount of BAT in the subject by at least 1%, e.g., 2%, 5%, 7%, 10%, 15%, 20%, 25% or more.
  • In some embodiments, the methods include providing a purified population of stem cells, e.g., a population of pluripotent mesenchymal stem cells, (e.g., a population of cells in which at least 60%, e.g., 70%, 80%, 90% or more of the cells are stem cells); and contacting the cells with a compound that increases expression, levels or activity of one or more of BMP-2, -4, -5, -6, and/or -7, as described herein, thereby activating the cells.
  • In some embodiments, the methods include evaluating the level of BAT adipogenesis in the cell or cell population. BAT differentiation can be evaluated by measuring any of, e.g., a BAT marker, such as uncoupling protein (UCP), e.g., UCP-1, expression; BAT morphology (e.g., using visual, e.g., microscopic, inspection of the cells); or BAT thermodynamics, e.g., cytochrome oxidase activity, Na+-K+-ATPase enzyme units, or other enzymes involved in BAT thermogenesis. In other embodiments, the methods include evaluating WAT differentiation, e.g., evaluating a WAT specific marker or WAT morphology.
  • A compound that increases BMP-2, -4, -5, -6, and/or -7 signaling can be, e.g., one or more of the following:
      • (a) a BMP-2, -4, -5, -6, and/or -7 polypeptide or a functional fragment or variant thereof, preferably an active (e.g., BMPR-I and/or BMPR-II activating) BMP-2, -4, -5, -6, and/or -7 polypeptide or a functional fragment or analog thereof (e.g., a mature BMP-2, -4, -5, -6, and/or -7 polypeptide, e.g., a mature BMP-2, -4, -5, -6, and/or -7 polypeptide described herein);
      • (b) a peptide or protein agonist of BMP-2, -4, -5, -6, and/or -7 that increases the activity, e.g., the BMPR-I and/or BMPR-II activating activity of BMP-2, -4, -5, -6, and/or -7 (e.g., by increasing or stabilizing binding of BMP-2, -4, -5, -6, and/or -7 to its receptor);
      • (c) a small molecule or protein mimetic that mimics BMP-2, -4, -5, -6, and/or -7 signaling activity, e.g., BMPR-I and/or BMPR-II binding activity, or SMAD phosphorylating activity;
      • (d) a small molecule that increases expression of BMP-2, -4, -5, -6, and/or -7, e.g., by binding to the promoter region of a BMP-2, -4, -5, -6, and/or -7 gene;
      • (e) an antibody, e.g., an antibody that binds to and stabilizes or assists the binding of BMP-2, -4, -5, -6, and/or -7 to a BMP-2, -4, -5, -6, and/or -7 binding partner (e.g., a BMP-2, -4, -5, -6, and/or -7 receptor described herein). In some embodiments, the antibody that binds the BMP-2, -4, -5, -6, and/or -7 is a monoclonal antibody, e.g., a humanized chimeric or human monoclonal antibody; or
      • (f) a nucleic acid encoding a BMP-2, -4, -5, -6, and/or -7 polypeptide or functional fragment or analog thereof. The nucleic acid can be a genomic sequence or a cDNA sequence.
  • In some embodiments, the compound is a BMP-2, -4, -5, -6, and/or -7 polypeptide or nucleic acid. As used herein, a “BMP-2, -4, -5, -6, and/or -7 polypeptide or nucleic acid” is a BMP-2, -4, -5, -6, and/or -7 polypeptide or nucleic acid as described herein, e.g., a mature human BMP-2, -4, -5, -6, and/or -7 polypeptide or active fragment thereof, or a nucleic acid encoding a mature human BMP-2, -4, -5, -6, and/or -7 polypeptide or active fragment thereof.
  • In some embodiments, the compound is a BMP-2 polypeptide, e.g., human BMP-2, e.g., a mature BMP-2 polypeptide, e.g., a BMP-2 polypeptide that includes amino acids 283-396 of SEQ ID NO:1. The polypeptide can be a recombinant polypeptide.
  • In some embodiments, the compound is a BMP-4 polypeptide, e.g., human BMP-4, e.g., a mature BMP-4 polypeptide, e.g., a BMP-4 polypeptide that includes amino acids 293-408 of SEQ ID NO:2. The polypeptide can be a recombinant polypeptide.
  • In some embodiments, the compound is a BMP-5 polypeptide, e.g., human BMP-5, e.g., a mature BMP-5 polypeptide, e.g., a BMP-4 polypeptide that includes amino acids 323-454 of SEQ ID NO:3. The polypeptide can be a recombinant polypeptide.
  • In some embodiments, the compound is a BMP-6 polypeptide, e.g., human BMP-6, e.g., a mature BMP-6 polypeptide, e.g., a BMP-6 polypeptide that includes amino acids 374-513 of SEQ ID NO:4, amino acids 382-513 of SEQ ID NO:4, amino acids 388-513 of SEQ ID NO:4, or amino acids 412-513 of SEQ ID NO:4. The polypeptide can be a recombinant polypeptide.
  • In some embodiments, the compound is a BMP-7 polypeptide, e.g., human BMP-7, e.g., a mature BMP-7 polypeptide, e.g., a BMP-7 polypeptide that includes amino acids 293-431 of SEQ ID NO:5. The polypeptide can be a recombinant polypeptide.
  • In some embodiments, the compound is a nucleic acid encoding a BMP-2, -4, -5, -6, and/or -7 polypeptide, or a biologically active fragment or analog thereof. A BMP nucleic acid can include: a BMP-2, -4, -5, -6, and/or -7 coding region; a promoter sequence, e.g., a promoter sequence from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene; an enhancer sequence; untranslated regulatory sequences, e.g., a 5′ untranslated region (UTR), e.g., a 5′UTR from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene, a 3′ UTR, e.g., a 3′UTR from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene; a polyadenylation site; an insulator sequence. In another embodiment, the level of BMP-2, -4, -5, -6, and/or -7 protein is increased by increasing the level of expression of an endogenous BMP-2, -4, -5, -6, and/or -7 gene, e.g., by increasing transcription of the BMP-2, -4, -5, -6, and/or -7 gene or increasing BMP-2, -4, -5, -6, and/or -7 mRNA stability. In some embodiments, transcription of the BMP-2, -4, -5, -6, and/or -7 gene is increased by: altering the regulatory sequence of the endogenous BMP-2, -4, -5, -6, and/or -7 gene, e.g., by the addition of a positive regulatory element (such as an enhancer or a DNA-binding site for a transcriptional activator); the deletion of a negative regulatory element (such as a DNA-binding site for a transcriptional repressor) and/or replacement of the endogenous regulatory sequence, or elements therein, with that of another gene, thereby allowing the coding region of the BMP-2, -4, -5, -6, and/or -7 gene to be transcribed more efficiently.
  • In some embodiments, the nucleic acid encodes or increases transcription of BMP-7.
  • In some embodiments, the methods include contacting, administering or expressing one or more other compounds in addition to the BMP, e.g., peroxisome proliferator-activated receptor gamma (PPARγ), Retinoid X receptor, alpha (RxRα), insulin, T3, a thiazolidinedione (TZD), retinoic acid, another BMP protein (e.g., BMP-1 or BMP-3), vitamin A, retinoic acid, insulin, glucocorticoid or agonist thereof, Wingless-type (Wnt), e.g., Wnt-1, Insulin-like Growth Factor-1 (IGF-1), or other growth factor, e.g., Epidermal growth factor (EGF), Fibroblast growth factor (FGF), Transforming growth factor (TGF)-α, TGF-β, Tumor necrosis factor alpha (TNFα), Macrophage colony stimulating factor (MCSF), Vascular endothelial growth factor (VEGF) and/or Platelet-derived growth factor (PDGF). In other embodiments, the compound can be a BMP-2, -4, -5, -6, and/or -7 protein as described herein or a portion thereof linked with a heterologous polypeptide sequence, e.g., a second BMP protein, to form a chimeric molecule or fusion protein. In some embodiments, the methods include administering the compound in combination with a second treatment, e.g., a second treatment for obesity or a related disorder such as diabetes. For example, the second treatment can be insulin, orlistat, phendimetrazine, and/or phentermine.
  • In general, the subject is a mammal. In some embodiments, the subject is a human subject, e.g., an obese human subject. In some embodiments, the subject is a non-human mammal, e.g., an experimental animal, a companion animal, or a food animal, e.g., a cow, pig, or sheep that is raised for food. Generally, where a BMP polypeptide or nucleic acid is used, the polypeptide or nucleic acid will be from the same species as the subject, e.g., human, cat, dog, cow, pig, or sheep.
  • In some embodiments, the methods include evaluating the subject for one or more of: weight, adipose tissue stores, adipose tissue morphology, insulin levels, insulin metabolism, glucose levels, thermogenic capacity, and cold sensitivity. The evaluation can be performed before, during, and/or after the administration of the compound. For example, the evaluation can be performed at least 1 day, 2 days, 4, 7, 14, 21, 30 or more days before and/or after the administration.
  • In some embodiments, the methods include one or more additional rounds of implantation of BMP-activated mesenchymal stem cells, e.g., to increase brown adipogenesis, e.g., to maintain or further reduce obesity in the subject.
  • In another aspect, the invention features a population of BMP-activated stem cells, e.g., pluripotent mesenchymal stem cells. In some embodiments, the cells are genetically engineered to express increased levels of a BMP-2, -4, -5, -6, and/or -7 polypeptide, e.g., a BMP-2, -4, -5, -6, and/or -7 polypeptide described herein, either stably or transiently. The cells can be, e.g., cultured mammalian cells, e.g., human cells. In some embodiments, the cells are genetically engineered to express at least one other protein as well, e.g., a non-BMP-2, -4, -5, -6, and/or -7 polypeptide, and/or a second (or more) BMP protein. The expressed BMP-2, -4, -5, -6, and/or -7 polypeptide will generally be of the same species as the stem cells, e.g., a human BMP expressed in human cells. In some embodiments, the cells are immortalized, e.g., capable of self-renewal indefinitely in culture.
  • In some embodiments, the cells used in the methods and compositions described herein express one or more BMP receptors, e.g., type I or II BMP receptors.
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
  • Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A is a bar graph illustrating mRNA levels of necdin, Wnt-10a, Pref-1, Gjb3, and PPARγ in cells treated with BMP-7 after 24 or 72 hours.
  • FIG. 1B is a bar graph illustrating mRNA levels of PGCα in cells treated with BMP-7 after 24 or 72 hours.
  • FIG. 1C is a set of ten line graphs illustrating the results of Quantitative RT-PCR analysis for C/EBPδ, C/EBPβ, C/EBPα, PPARγ, aP2, PGC-1α, PGC-1β, NRF-1, NRF-2, Tfam, and Cyto C using total RNAs isolated from C3H10T1/2 cells before (day 0) or after 3 days of treatment with BMP-7 or vehicle and at day 6 and day 10 of differentiation. Data are presented as mean±SEM (n=3).
  • FIGS. 2A-2C are bar graphs illustrating the effect of administration of a BMP-7 adenoviral construct (black bars) or Lac Z adenoviral control construct (open bars) on body weight (2A), epididymal WAT (2B), and interscapular BAT (2C) in 4- and 12-week old mice. Weights of epididymal WAT and interscapular BAT are presented as percentage of total body weight. All data for FIGS. 2A-2F are presented as mean±SEM. Asterisks depict statistically significant differences between Adeno-lacZ and Adeno-BMP 7 for each age group by ANOVA (*=P<0.05, **=P<0.01, ***=P<0.001).
  • FIG. 2D is a bar graph illustrating the effect of administration of a BMP-7 adenoviral vector (black bars) or Lac Z adenoviral control construct (open bars) on UCP-1 gene expression as measured by Q-RT-PCR.
  • FIGS. 2E and 2F are bar graphs illustrating the effect of administration of a BMP-7 adenoviral vector (black bars) or Lac Z adenoviral control construct (open bars) on oxygen consumption VO2 in 4-week old and 12-week old mice for light (2E) and dark (2F) cycles.
  • FIGS. 3A-3D are photomicrographs of sections of tissue showing the morphology of normal brown fat (3A), normal white fat (3B), implant-derived tissue with a brown fat-type morphology (3C), and implant-derived tissue with a partially white fat-type morphology (3D).
  • FIGS. 4A and 4B are bar graphs illustrating the effect of implantation of BMP-7 treated pluripotent mesenchymal stem cells on blood glucose (4A) and blood insulin levels (4B).
  • FIGS. 5A and 5B are each six panels of bar graphs illustrating expression levels of PGC-1α, NRF-1, Tfam, PGC-1β, NRF-2, and Cyto C after 3 days (5A) or 8 days (5B) of treatment with Insulin and T3 (light gray bars), Insulin, T3 and BMP-6 (mid gray bars), or Insulin, T3, and BMP-7 (black bars), as compared to control (white bars) in brown preadipocytes (left group of four bars in each panel) and white preadipocytes (right group of four bars in each panel).
  • FIGS. 5C and 5D are each pairs of photomicrographs of brown preadipocytes (5C) and 3T3-L1 cells (5D) treated with Insulin and T3 (left panel of each) or Insulin, T3 and BMP-7 (right panels of each), showing an increased number and size of mitochondria in the BMP-7 treated cells.
  • DETAILED DESCRIPTION
  • BMPs are members of the transforming growth factor-β superfamily that are involved in multiple key steps of embryonic development as well as throughout life (Kishigami and Mishina, Cytokine. Growth Factor. Rev. 16:265-278 (2005); Chen et al., Growth Factors. 22:233-241 (2004); Yamamoto and Oelgeschlager, Naturwissenschaften 91:519-534 (2004)). BMPs have been shown to play a role in two different stages of adipocyte development. First, BMP-2 and 4 stimulate differentiation of multipotent mesenchymal cells and bone marrow stromal cells into adipocytes under appropriate conditions (Butterwith et al., Biochem. Soc Trans 24:163 S (1996); Chen et al., J. Cell. Biol. 142:295-305 (1998); Chen et al., J Cell Biochem. 82:187-199 (2001); Tang et al., Proc Natl. Acad Sci U S.A. 101:9607-9611 (2004)). In addition, BMPs also stimulate the differentiation of committed white preadipocytes (Sottile and Seuwen, FEBS Lett. 475:201-204 (2000); Rebbapragada et al., Mol. Cell. Biol. 23:7230-7242 (2003)). However, other studies showed that BMP-2 suppressed adipogenic differentiation and promoted osteogenesis in multipotent mesenchymal progenitors via homeobox gene, Msx2 (Ichida et al., J. Biol. Chem. 279:34015-34022 (2004); S. L. Cheng et al., J. Biol. Chem. 278:45969-45977 (2003)).
  • As described herein, BMP-2, -4, -5, -6, and -7 are involved in adipocyte differentiation, and treatment of stem cells with BMP-2, -4, -5, -6, and/or -7 promotes brown adipogenesis. BMP-2, -4, -5, -6, and/or -7 are thus therapeutic, diagnostic and drug discovery targets for adipose-related disorders, such as obesity and related disorders such as diabetes, insulin resistance, hyperglycemia, hyperlipidemia, and hypercholesterolemia. In general, the methods described herein include implanting a population of BMP-activated stem cells as described herein into a subject.
  • Some of the methods described herein include implanting stem cells that have been treated with an agent that increases BMP signalling. In general, the methods include treating (e.g., contacting) stem cells, e.g., pluripotent mesenchymal stem cells, with the compound in an amount sufficient to increase BMP signalling, and thereafter implanting the BMP-activated cells (e.g., at least one cell or a population of such cells) in a subject. Suitable agents can be the BMPs themselves, e.g., recombinant proteins, or nucleic acids that encode the BMPs, to treat the stem cells. In some embodiments, treating the cells includes genetically engineered the cells in vitro to express a BMP-2, -4, -5, -6, and/or -7 polypeptide. The cells are then administered to a subject. Populations of such genetically engineered stem cells are also included within the scope of the present invention. Other compounds are described herein.
  • BMP-Activated Stem Cells
  • The methods described herein include the use of BMP-activated stem cells. Stem cells are progenitor cells that are capable of both self-renewal and differentiation into many different cell lineages. Suitable stem cells, and methods for isolating them, are known in the art. Stem cells can be pluripotent or totipotent. Embryonic or adult stem cells can be used. Embryonic stem cells are generally derived from embryos that are less than a week old, e.g., in vitro fertilized embryos. Adult stem cells can include hematopoietic stem cells, generally isolated from bone marrow, peripheral blood, or umbilical cord blood; mesenchymal stem cells, e.g., from bone marrow or periosteum; intestinal (gut) stem cells, from the small intestine; skin stem cells; neuronal stem cells; and hepatic stem cells (oval cells), from the liver. See, e.g., Tuan et al., Arthritis Res. Ther. 5:32-45 (2003); Prockop et al., Proc. Natl. Acad. Sci. USA 100 Suppl 1:11917-23 (2003); Bianco and Gehron Robey, J. Clin. Invest. 105(12):1663-8 (2000). As one example, primary pluripotent mesenchymal stem cells can be isolated from bone marrow (see, e.g., Halleux et al., J. Musculoskelet. Neuronal. Interact. 2(1):71-6 (2001)), connective tissue (Young et al., Dev Dyn. 202(2):137-44 (1995)), and other tissues. In some embodiments, the stem cells are isolated from an adipose tissue.
  • In some embodiments, the cells are purified, e.g., a population of cells in which at least 60%, e.g., 70%, 80%, 90% or more of the cells are stem cells. A purified population of stem cells is enriched by any method known in the art for cell enrichment, e.g., immunomagnetic cell sorting, fluorescence activated cell sorting (FACS), adherence to tissue culture plates and flasks, or culturing under conditions that favor the growth of the desired stem cells. Such methods are known in the art.
  • The term “primary cell” includes cells present in a suspension of cells isolated from a mammalian tissue source (prior to their being plated, i.e., attached to a tissue culture substrate such as a dish or flask), cells present in an explant derived from tissue, both of the previous types of cells plated for the first time, and cell suspensions derived from these plated cells. The term “secondary cell” or “cell strain” refers to cells at all subsequent steps in culturing. Secondary cells are cell strains that consist of secondary cells that have been passaged one or more times.
  • Primary and secondary stem cells can be obtained from a variety of tissues and include cell types which can be maintained and propagated in culture. Primary cells are preferably obtained from the individual to whom the BMP-activated cells are administered. However, primary cells can also be obtained from a donor (e.g., an individual other than the recipient, typically of the same species, preferably ab immunologically compatible individual). Methods for obtaining and culturing such cells are known in the art.
  • The methods can include allowing stem cells to undergo sufficient number doubling to produce either a clonal cell strain or a heterogeneous cell strain of desired size, e.g., a sufficient number to provide a therapeutic effect to an individual, or a sufficient number to establish a stable cell line, before or after BMP-activation. Where the cells are not transfected but rather treated with a BMP, the cells can be cultured for a time in the absence of the BMP, then in the presence of the BMP for a time (e.g., 1, 2, 3 or more days) before implantation into the subject. The cells can be washed (e.g., in isotonic PBS) before implantation to remove any contaminants, including BMPs or components of growth media, before implantation. The number of required cells is variable and depends on a variety of factors, including but not limited to, the use of the transfected cells, the functional level of the exogenous DNA in the transfected cells, the site of implantation of the transfected cells (for example, the number of cells that can be used is limited by the anatomical site of implantation), and the age, surface area, and clinical condition of the patient. In some embodiments, the population of BMP-activated stem cells includes at least 107, 108, 109, or more cells.
  • BMP-activated stem cells are stem cells that have an enhanced level of BMP signalling, e.g., BMP-2, -4, -5, -6, and/or -7 signalling, wherein the level of BMP signalling has been increased by direct human intervention. BMP signalling can be enhanced in the cells by any method known in the art or described herein, e.g., by treating the cell with a compound that enhances BMP signalling as described herein, e.g., a BMP polypeptide or nucleic acid. Populations of stem cells activated by methods described herein are also included within the present invention. Optionally, the population of BMP-activated cells can be suspended in a pharmaceutically acceptable carrier, e.g., for storage or implantation. As used herein, the language “pharmaceutically acceptable carrier” is intended to include any and all solvents, media, antibacterial and antifungal agents, isotonic agents, and the like, compatible with pharmaceutical administration and viability of the cells. In general, the cells will be maintained in a sterile state. The use of such media and agents for pharmaceutically active substances are known. Except insofar as any conventional media or agent is incompatible with the active compound, such media can be used in the compositions of the invention. Supplementary active compounds can also be incorporated into the compositions.
  • The cells can be autologous, allogeneic or xenogeneic. In some embodiments, methods described herein can include obtaining a population of stem cells from a subject, optionally culturing and/or enriching the stem cells to obtain a purified population of stem cells, treating the cells with an agent that enhances BMP signalling as described herein to activate the cells, and implanting the cells in the same subject from which they were removed. In some embodiments, the cells are allogeneic or xenogeneic; if necessary, immune suppression can be administered to prevent rejection of the cells.
  • BMP Proteins
  • BMP proteins have been used in the clinic in the treatment of bone and cartilage disorders or wounds. The effective clinical use of recombinant BMPs is discussed in Einhorn, J. Bone and Joint Surgery 85A:82-88 (2003), and Sandhu, Spine 28(15):S64-73 (2003). A BMP polypeptide (e.g., a mature BMP polypeptide) is itself is a viable therapeutic compound because BMPs are small secreted proteins that are internalized into their target cells where they exert their activity. Although the human proteins are described herein, one of skill in the art will appreciate that when another species is the intended recipient of the treated cells, homologous proteins from that species can also be used, e.g., cow, pig, sheep, or goat. Such homologous proteins can be identified, e.g., using methods known in the art, e.g., searching available databases for homologs identified in the target species, e.g., the homologene database.
  • BMP-2
  • BMP-2 is 396 amino acids in length, localized to chromosome 20p12 in human. The nucleotide and amino acid sequences of human BMP-2 are disclosed in Wozney et al., Science 242(4885):1528-1534 (1988). BMP2 belongs to the transforming growth factor-beta (TGFβ) superfamily. Bone morphogenetic protein induces bone formation, and BMP2 is a candidate gene for the autosomal dominant disease of fibrodysplasia (myositis) ossificans progressive. Bone morphogenetic protein 2 regulates myogenesis through dosage-dependent PAX3 expression in pre-myogenic cells, and is expressed in mesoderm under SHM control through the SOX9.
  • The human BMP-2 is shown below. Amino acids 38-268 are the TGFβ propeptide domain, and 291-396 are the TGFβ family N-terminal domain. Amino acids 283-396 are the mature peptide. The sequence is set forth in Wozney et al., Science 242:1528-1534 (1988).
  • (SEQ ID NO: 1)
      1 MVAGTRCLLA LLLPQVLLGG AAGLVPELGR RKFAAASSGR PSSQPSDEVL SEFELRLLSM
     61 FGLKQRPTPS RDAVVPPYML DLYRRHSGQP GSPAPDHRLE RAASRANTVR SFHHEESLEE
    121 LPETSGKTTR RFFFNLSSIP TEEFITSAEL QVFREQMQDA LGNNSSFHHR INIYEIIKPA
    181 TANSKFPVTR LLDTRLVNQN ASRWESFDVT PAVMRWTAQG HANHGFVVEV AHLEEKQGVS
    241 KRHVRISRSL HQDEHSWSQI RPLLVTFGHD GKGHPLHKRE KRQAKHKQRK RLKSSCKRHP
    301 LYVDFSDVGW NDWIVAPPGY HAFYCHGECP FPLADHLNST NHAIVQTLVN SVNSKIPKAC
    361 CVPTELSAIS MLYLDENEKV VLKNYQDMVV EGCGCR

    The mature form of BMP-2 contains four potential N-linked glycosylation sites per polypeptide chain, and four potential disulfide bridges. See UniProt entry No. P 12643; HomoloGene:926.
  • BMP-4
  • BMP-4 induces cartilage and bone formation, and is important in mesoderm induction, tooth development, limb formation and fracture repair. The sequence of the BMP-4 preproprotein is shown below. Amino acids 41-276 are the TGFβ propeptide domain, and 302-408 are the TGFβ family N-terminal domain. Amino acids 293-408 are the mature peptide. The sequence is set forth in Wozney et al., Science 242:1528-1534 (1988).
  • (SEQ ID NO: 2)
      1 MIPGNRMLMV VLLCQVLLGG ASHASLIPET GKKKVAEIQG HAGGRRSGQS HELLRDFEAT
     61 LLQMFGLRRR PQPSKSAVIP DYMRDLYRLQ SGEEEEEQIH STGLEYPERP ASRANTVRSF
    121 HHEEHLENIP GTSENSAFRF LFNLSSIPEN EAISSAELRL FREQVDQGPD WERGFHRINI
    181 YEVMKPPAEV VPGHLITRLL DTRLVHHNVT RWETFDVSPA VLRWTREKQP NYGLAIEVTH
    241 LHQTRTHQGQ HVRISRSLPQ GSGNWAQLRP LLVTFGHDGR GHALTRRRRA KRSPKHHSQR
    301 ARKKNKNCRR HSLYVDFSDV GWNDWIVAPP GYQAFYCHGD CPFPLADHLN STNHAIVQTL
    361 VNSVNSSIPK ACCVPTELSA ISMLYLDEYD KVVLKNYQEM VVEGCGCR
  • The mature form of BMP-4 contains four potential N-linked glycosylation sites per polypeptide chain. A variant exists in which V152 is an A. See UniProt Accession No. P12644; HomoloGene:7247.
  • BMP-5
  • The BMP-5 preproprotein is a 454 amino acid protein, as shown below. BMP-5 induces cartilage and bone formation. The sequence is set forth in Celeste et al., Proc. Natl. Acad. Sci. U.S.A., 87, 9843-9847, 1990.
  • (SEQ ID NO: 3)
      1 MHLTVFLLKG IVGFLWSCWV LVGYAKGGLG DNHVHSSFIY RRLRNHERRE IQREILSILG
     61 LPHRPRPFSP GKQASSAPLF MLDLYNAMTN EENPEESEYS VRASLAEETR GARKGYPASP
    121 NGYPRRIQLS RTTPLTTQSP PLASLHDTNF LNDADMVMSF VNLVERDKDF SHQRRHYKEF
    181 RFDLTQIPHG EAVTAAEFRI YKDRSNNRFE NETIKISIYQ IIKEYTNRDA DLFLLDTRKA
    241 QALDVGWLVF DITVTSNHWV INPQNNLGLQ LCAETGDGRS INVKSAGLVG RQGPQSKQPF
    301 MVAFFKASEV LLRSVRAANK RKNQNRNKSS SHQDSSRMSS VGDYNTSEQK QACKKHELYV
    361 SFRDLGWQDW IIAPEGYAAF YCDGECSFPL NAHMNATNHA IVQTLVHLMF PDHVPKPCCA
    421 PTKLNAISVL YFDDSSNVIL KKYRNMVVRS CGCH
  • The mature BMP-5 protein is believed to be amino acids 323-454 of SEQ ID NO:3, and has four potential N-linked glycosylation sites per polypeptide chain, and four potential disulfide bridges. See UniProt Accession Nos. P22003; Q9H547; or Q9NTM5; HomoloGene: 22412.
  • BMP-6
  • BMP-6 is an autocrine stimulator of chondrocyte differentiation, and is involved in the development of embryonic neural, and urinary systems, as well as growth and differentiation of liver and keratinocytes. BMP-6 knockout mice are viable and show a slight delay in ossification of the sternum. BMP-6 (precursor) is a 57 kD protein, 513 amino acids in length, localized to chromosome 6p24 in human. The nucleotide and amino acid sequence of human BMP-6 is disclosed in U.S. Pat. No. 5,187,076. BMP-6 is predicted to be synthesized as a precursor molecule which is cleaved to yield a 132 amino acid mature polypeptide with a calculated molecular weight of approximately 15 Kd. The mature form of BMP-6 contains three potential N-linked glycosylation sites per polypeptide chain. The active BMP-6 protein molecule is likely a dimer. Processing of BMP-6 into the mature form involves dimerization and removal of the N-terminal region in a manner analogous to the processing of the related protein TGFβ (Gentry et al., Molec. Cell. Biol. 8:4162 (1988); Dernyck et al., Nature 316:701 (1985)). The human BMP-6 precursor is shown below. The mature polypeptide is believed to include amino acids 374-513 of SEQ ID NO:4. Other active BMP-6 polypeptides include polypeptides including amino acids 382-513, 388-513 and 412-513 of SEQ ID NO:4.
  • (SEQ ID NO: 4)
    MPGLGRRAQW LCWWWGLLCS CCGPPPLRPP LPAAAAAAAG GQLLGDGGSP GRTEQPPPSP  61
    QSSSGFLYRR LKTQEKREMQ KEILSVLGLP HRPRPLHGLQ QPQPPALRQQ EEQQQQQQLP 121
    RGEPPPGRLK SAPLFMLDLY NALSADNDED GASEGERQQS WPHEAASSSQ RRQPPPGAAH 181
    PLNRKSLLAP GSGSGGASPL TSAQDSAFLN DADMVMSFVN LVEYDKEFSP RQRHHKEFKF 241
    NLSQIPEGEV VTAAEFRIYK DCVMGSFKNQ TFLISIYQVL QEHQHRDSDL FLLDTRVVWA 301
    SEEGWLEFDI TATSNLWVVT PQHNMGLQLS VVTRDGVHVH PRAAGLVGRD GPYDKQPFMV 361
    AFFKVSEVHV RTTRSASSRR RQQSRNRSTQ SQDVARVSSA SDYNSSELKT ACRKHELYVS 421
    FQDLGWQDWI IAPKGYAANY CDGECSFPLN AHMNATNHAI VQTLVHLMNP EYVPKPCCAP 481
    TKLNAISVLY FDDNSNVILK KYRNMVVRAC GCH
  • The human BMP-6 promoter has been characterized (See Tamada et al., Biochim Biophys Acta. 1998, 1395(3):247-51), and can be used in methods described herein. See UniProt Accession No. P22004; HomoloGene:1300.
  • Administration, antisense treatment, and quantitation of BMP-6 are described in Boden et al. (Endocrinology Vol. 138, No. 7 2820-2828).
  • BMP-7
  • BMP-7 also belongs to the TGFβ superfamily. It induces cartilage and bone formation, and may be the osteoinductive factor responsible for the phenomenon of epithelial osteogenesis. BMP-7 plays a role in calcium regulation and bone homeostasis, and in the regulation of anti-inflammatory response in the adult gut tissue. The sequence of BMP-7 is shown below:
  • (SEQ ID NO: 5)
      1 MHVRSLRAAA PHSFVALWAP LFLLRSALAD FSLDNEVHSS FIHRRLRSQE RREMQREILS
     61 ILGLPHRPRP HLQGKHNSAP MFMLDLYNAM AVEEGGGPGG QGFSYPYKAV FSTQGPPLAS
    121 LQDSHFLTDA DMVMSFVNLV EHDKEFFHPR YHHREFRFDL SKIPEGEAVT AAEFRIYKDY
    181 IRERFDNETF RISVYQVLQE HLGRESDLFL LDSRTLWASE EGWLVFDITA TSNHWVVNPR
    241 HNLGLQLSVE TLDGQSINPK LAGLIGRHGP QNKQPFMVAF FKATEVHFRS IRSTGSKQRS
    301 QNRSKTPKNQ EALRMANVAE NSSSDQRQAC KKHELYVSFR DLGWQDWIIA PEGYAAYYCE
    361 GECAFPLNSY MNATNHAIVQ TLVHFINPET VPKPCCAPTQ LNAISVLYFD DSSNVILKKY
    421 RNNVVRACGC H
  • Amino acids 1-29 are a potential signal sequence; 30-431 are the prepropeptide, and 293-431 are the mature protein. The mature form of BMP-7 contains four potential N-linked glycosylation sites per polypeptide chain, and four potential disulfide bridges. See UniProt Accession No. P 18075; HomoloGene: 20410.
  • Pharmacokinetic Properties and Therapeutic Activity
  • Modifications can be made to a protein compound that result in pharmacokinetic properties of the protein which are desirable for use in protein therapy. For example, such modifications can result in an increase in cellular uptake, circulatory half-life, rate of clearance and reduced immunogenicity. Several art-recognized approaches are known that are useful to optimize the therapeutic activity of a protein compound, e.g., a compound described herein such as a BMP-2, -4, -5, -6, and/or -7 polypeptide.
  • Expression System
  • For recombinant proteins, the choice of expression system can influence pharmacokinetic characteristics. Differences between expression systems in post-translational processing can lead to recombinant proteins of varying molecular size and charge, which can affect, for example, cellular uptake, circulatory half-life, rate of clearance and immunogenicity. The pharmacokinetic properties of the protein may be optimized by the appropriate selection of an expression system, such as selection of a bacterial, viral, or mammalian expression system. Exemplary mammalian cell lines useful in expression systems for therapeutic proteins are Chinese hamster ovary, (CHO) cells, the monkey COS-1 cell line and the CV-1 cell line.
  • Chemical Modification
  • A protein can be chemically altered to enhance the pharmacokinetic properties while maintaining activity. The protein can be covalently linked to a variety of moieties, altering the molecular size and charge of the protein and consequently its pharmacokinetic characteristics. The moieties are preferably non-toxic and biocompatible. In some embodiments, polyethylene glycol (PEG) can be covalently attached to the protein (PEGylation). See, e.g., Poly(ethylene glycol): Chemistry and Biological Applications, Harris and Zalipsky, eds., ACS Symposium Series, No. 680, 1997; Harris et al., Clinical Pharmacokinetics 40:7, 485-563 (2001)). In another embodiment, the protein can be similarly linked to oxidized dextrans via an amino group. (See Sheffield, Current Drug Targets—Cardiovas. and Haemat. Dis. 1:1, 1-22 (2001)).
  • Furthermore, the protein compounds can be chemically linked to another protein. The protein can be cross-linked carrier protein to form a larger molecular weight complex with improved cellular uptake. In some embodiments, the carrier protein can be a serum protein, such as albumin. The protein can be attached to one or more albumin molecules via a bifunctional cross-linking recompound. The cross-linking recompound may be homo- or heterofunctional. In another embodiment, the protein can cross-link with itself to form a homodimer, trimer, or higher analog. Again, either heterobifunctional or homobifunctional cross-linking recompounds can be used to form the dimers or trimers. (See Stykowski et al., Proc. Natl. Acad. Sci. USA, 95, 1184-1188 (1998)).
  • BMP Nucleic Acids
  • The stem cells of mammalian origin can be, for example, activated by transfection with an exogenous nucleic acid that includes a heterologous nucleotide sequence, e.g., encoding BMP-2, -4, -5, -6, and/or -7, or an agonist thereof, with or without a nucleotide sequence encoding a signal peptide, and produce the encoded product either transiently or stably, over extended periods of time. A heterologous amino acid can also be a regulatory sequence, e.g., a promoter, which causes expression, e.g., constitutive or inducible expression or upregulation, of an endogenous BMP-2, -4, -5, -6, and/or -7 sequence. An exogenous nucleic acid sequence can be introduced into a primary or secondary cell by homologous recombination as described, for example, in U.S. Pat. No. 5,641,670, the contents of which are incorporated herein by reference. The transfected cells can also include DNA encoding a selectable marker that confers a selectable phenotype upon them, facilitating their identification and isolation.
  • In some embodiments, the compound that enhances BMP signalling as described herein includes, e.g., a BMP nucleic acid, e.g., a BMP-2, -4, -5, -6, and/or -7 encoding sequence or active fragment thereof, and any of: a promoter sequence, e.g., a promoter sequence from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene; an enhancer sequence, e.g., 5′ untranslated region (UTR), e.g., a 5′ UTR from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene, a 3′ UTR, e.g., a 3′ UTR from a BMP-2, -4, -5, -6, and/or -7 gene or from another gene; a polyadenylation site; an insulator sequence; or another sequence that enhances the expression of BMP-2, -4, -5, -6, and/or -7.
  • The nucleic acids described herein, e.g., a nucleic acid encoding a BMP-2, -4, -5, -6, and/or -7 polypeptide as described herein, can be incorporated into a gene construct. The methods described herein can use such expression vectors for in vitro transfection and expression of a BMP-2, -4, -5, -6, and/or -7 polypeptide described herein in particular cell types, e.g., stem cells, e.g., pluripotent mesenchymal stem cells. Expression constructs of such components can be administered in any biologically effective carrier, e.g., any formulation or composition capable of effectively delivering the component gene to cells in vivo. Approaches include insertion of a subject gene in viral vectors including recombinant retroviruses, adenovirus, adeno-associated virus, and herpes simplex virus-1, or recombinant bacterial or eukaryotic plasmids.
  • Viral vectors transfect cells directly, and infection of cells with a viral vector generally has the advantage that a large proportion of the targeted cells can receive the nucleic acid. Additionally, molecules encoded within the viral vector, e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells which have taken up viral vector nucleic acid. Retroviral vectors, adenovirus-derived vectors, and adeno-associated virus vectors can also be used as a recombinant gene delivery system for the transfer of exogenous genes. These vectors provide efficient delivery of genes into cells, and the transferred nucleic acids are generally stably integrated into the chromosomal DNA of the host. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel et al. (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14, and other standard laboratory manuals.
  • Non-viral methods can also be employed to cause expression of an nucleic acid compound described herein (e.g., a BMP-2, -4, -5, -6, and/or -7 polypeptide encoding nucleic acid) into a cell. Most nonviral methods of gene transfer rely on normal mechanisms used by mammalian cells for the uptake and intracellular transport of macromolecules. In some embodiments, non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject gene by the targeted cell. Plasmid DNA can be delivered with the help of, for example, cationic liposomes (e.g., LIPOFECTIN™) or derivatized (e.g., antibody conjugated), polylysine conjugates, gramicidin S, artificial viral envelopes or other such intracellular carriers, as well as direct injection of the gene construct or CaPO4 precipitation. Other embodiments include plasmid injection systems such as are described in Meuli et al., J. Invest. Dermatol. 116(1):131-135 (2001); Cohen et al., Gene Ther 7(22):1896-905 (2000); or Tam et al., Gene Ther. 7(21):1867-74 (2000).
  • Formulations
  • The BMP compounds described herein can be formulated in any suitable manner, e.g., in a carrier system, for use in contacting with the populations of cells. The carrier can be a colloidal system. The colloidal system can be liposome, a phospholipid bilayer vehicle. In some embodiments, the protein is encapsulated in a liposome while maintaining protein integrity. As one skilled in the art would appreciate, there are a variety of methods to prepare liposomes. (See Lichtenberg et al., Methods Biochem Anal, 33:337-462 (1988), LIPOSOME TECHNOLOGY Anselem et al., CRC Press, 1993). Liposomes can be prepared from an assortment of phospholipids varying in size and substitution, and may also contain additional components with low toxicity, such as cholesterol. The liposome can be formulated and isolated in a variety of shapes and sizes. Additionally, moieties may attached to the surface of the liposome to further enhance the pharmacokinetic properties of the carrier. The moieties may be attached to phospholipid or cholesterol molecules, and the percentage of the moiety incorporated on the surface may be adjusted for optimal liposome stability and pharmacokinetic characteristics. One embodiment comprises a liposome with poly-ethylene glycol (PEG) added to the surface. Liposomal formulations can delay clearance and increase cellular uptake. (See Reddy, Annals of Pharmacotherapy, 34(7/8):915-923 (2000)).
  • The carrier can also be a polymer, e.g., a biodegradable, biocompatible polymer matrix. In some embodiments, the protein can be embedded in the polymer matrix while maintaining protein integrity. The polymer may be natural, such as polypeptides, proteins or polysaccharides, or synthetic, such as poly(α-hydroxy) acids. Examples include carriers made of e.g., collagen, fibronectin, elastin, cellulose acetate, cellulose nitrate, polysaccharide, fibrin, gelatin, and combinations thereof. In some embodiments, the polymer is poly-lactic acid (PLA) or co-polylactic/glycolic acid (PGLA). The polymeric matrices can be prepared and isolated in a variety of forms and sizes, including microspheres and nanospheres. Polymer formulations can lead to prolonged duration of therapeutic effect. (See Reddy, Annals of Pharmacotherapy, 34(7/8):915-923 (2000)). A polymer formulation for human growth hormone (hGH) has been used in clinical trials. (See Kozarich and Rich, Chemical Biology 2:548-552 (1998)).
  • Examples of polymer microsphere sustained release formulations are described in PCT publication WO 99/15154 (Tracy et al.), U.S. Pat. Nos. 5,674,534 and 5,716,644 (both to Zale et al.), PCT publication WO 96/40073 (Zale et al.), and PCT publication WO 00/38651 (Shah et al.). U.S. Pat. Nos. 5,674,534 and 5,716,644 and PCT publication WO 96/40073 describe a polymeric matrix containing particles of erythropoietin that are stabilized against aggregation with a salt.
  • Cell Therapy
  • Methods described herein can include implanting a population of BMP-activated stem cells, e.g., as described herein, into a subject to be treated, wherein said population of BMP-activated stem cells, or their progeny (i.e., daughter cells), undergo brown adipogenesis. Once implanted, the stem cells will generally undergo adipogenesis, generating BAT in the subject.
  • These cell therapy methods are useful, e.g., for the treatment of obesity and insulin resistance in a subject, or for treating a disease associated with a lack of mitochondria, e.g., diabetes, cancer, neurodegeneration, and aging.
  • Methods for implanting the populations of BMP-activated stem cells are known in the art, e.g., using a delivery system configured to allow the introduction of cells into a subject. In general, the delivery system can include a reservoir containing a population of BMP-activated pluripotent mesenchymal stem cells, and a needle in fluid communication with the reservoir. Typically, the population of BMP-activated stem cells will be in a pharmaceutically acceptable carrier, with or without a scaffold, matrix, or other implantable device to which the cells can attach (examples include carriers made of, e.g., collagen, fibronectin, elastin, cellulose acetate, cellulose nitrate, polysaccharide, fibrin, gelatin, and combinations thereof). Such delivery systems are also within the scope of the invention. Generally, such delivery systems are maintained in a sterile manner. Various routes of administration and various sites (e.g., renal sub capsular, subcutaneous, central nervous system (including intrathecal), intravascular, intrahepatic, intrasplanchnic, intraperitoneal (including intraomental), intramuscularly implantation) can be used. Generally, the cells will be implanted into the subject subcutaneously. In some embodiments, the population of BMP-activated stem cells that is implanted includes at least 107, 108, 109, or more cells.
  • Where non immunologically compatible cells are used, an immunosuppressive compound e.g., a drug or antibody, can be administered to the recipient subject at a dosage sufficient to achieve inhibition of rejection of the cells. Dosage ranges for immunosuppressive drugs are known in the art. See, e.g., Freed et al., N. Engl. J. Med. 327:1549 (1992); Spencer et al., N. Engl. J. Med. 327:1541 (1992); Widner et al., N. Engl. J. Med. 327:1556 (1992)). Dosage values may vary according to factors such as the disease state, age, sex, and weight of the individual.
  • The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.
  • EXAMPLES Example 1 BMPs Induce Differentiation of Pluripotent C3H10T1/2 Mesenchymal Stem Cells and Stromo-Vascular Fraction (SVF) Cells into Brown Adipocytes
  • Obesity is the result of imbalance between energy intake and energy expenditure. Adipose tissue plays an active role in energy balance. Two functionally different types of fat tissue are present in mammals: white adipose tissue, which is the primary site of triglycerides storage and release of fatty acids, and brown adipose tissue, which is specialized for energy expenditure via thermogenesis. The regulation of differentiation and balance of function between the two types of adipose tissue is critical to whole body energy homeostasis. The role of BMPs in this regulation was explored in the experiments described herein.
  • To evaluate the effect of BMPs on pluripotent C3H10T1/2 mesenchymal stem cells (ATCC), 33 nM recombinant BMPs (obtained from R&D Systems) were added to the normal culture medium (Dulbecco's Modified Earle's Medium High supplemented with 10% Fetal Bovine Serum) after the cells reached confluence; fresh medium was changed every 2 days. After 3 days of treatment with BMP-4, -6, or -7, the cells were treated with BAT induction cocktail (1 nM T3, 20 nM insulin, 0.5 mM isobutylmethylxanthine (IBMX), 0.5 mM dexamethazone, and 0.125 mM indomethacin) or WAT induction cocktail (1 nM T3, 0.5 mM isobutylmethylxanthine (IBMX), 0.5 mM dexamethazone, and 0.125 mM indomethacin). To visualize lipid accumulation, dishes were washed twice with phosphate-buffered saline and fixed with 10% buffered formalin for at least 1 hour at room temperature. Cells were then stained for 2 hours at room temperature with a filtered Oil Red O solution (0.5% Oil Red O in isopropyl alcohol), washed twice with distilled water, and visualized.
  • Cells treated with BMP-4, -6, or -7 plus the BAT induction cocktail showed a substantial increase in lipid accumulation as monitored by Oil Red O staining; cells treated with BMP-4, -6, or -7 plus the WAT induction cocktail showed a minor increase in lipid accumulation as monitored by Oil Red O staining; BMP-2, but not BMP-3, had a similar effect. Cells treated with BMP-6 or -7 showed a minor increase in lipid accumulation even in the absence of any induction cocktail. Expression of UCP-1 was detected by methods described in Klein et al., J. Biol. Chem. 274:34795-34802 (1999).
  • Cells treated with BMP-4, -6, or -7 in the presence of either WAT or BAT induction cocktail markedly induced the expression of FAS protein; strong upregulation of UCP-1 and PPARγ mRNA protein was seen in the cells treated with the BAT induction cocktail, but some expression of PPARγ and UCP-1 was seen in cells treated with the WAT induction cocktail, indicating the genesis of some BAT during the course of differentiation. Again, BMP-2, but not BMP-3, had a similar effect.
  • Treatment of the pluripotent C3H10T1/2 mesenchymal stem cells with bone morphogenetic protein (BMP)-2,4,6, and 7, but not BMP-3, triggers commitment of these cells to the brown adipocyte lineage as monitored by increases in lipid accumulation and expression of the brown fat specific marker uncoupling protein-1 (UCP-1; FIGS. 1A-B). This is accompanied by induction of other adipogenic markers peroxisome proliferator-activated receptor gamma (PPARγ) and fatty acid synthase (FAS), and increases in expression of the PPARγ coactivator-1 alpha (PGC-1α) (FIGS. 1A-B).
  • Analysis of gene expression indicated that the C3H10T1/2 cells had become committed to the brown adipocyte lineage after 3 days of BMP-7 treatment (FIG. 1C). At this time, BMP-7 pre-treatment had already increased expression of C/EBPδ, the first transcription factor to appear during adipocyte differentiation 39, by 4-fold. At day 6, as the cells further progressed in brown adipocyte differentiation, C/EBPδ levels remained high, and expression levels of C/EBPβ, C/EBPα, PPARγ and aP2 were greatly increased, as previously described in committed white and brown preadipocytes during in vitro differentiation 27,40, by 2.9-, 4.2-, 2.0-, and 1.6-fold, respectively, in BMP-7-pretreated cells (FIG. 1C, left panels). Interestingly, BMP-7 pretreatment also caused a transient induction of PGC-1α expression at day 3 and a significant increase in expression of NRF-1 and Tfam at both day 3 and day 6, followed by a later increase of Cyto C expression (FIG. 1C, right panels), indicating an increased mitochondrial biogenesis in C3H10T1/2 cells pretreated with BMP-7 followed by induction to brown adipocyte lineage.
  • Moreover, BMP-6 and BMP-7 in combination with hormone induction cocktail and rosiglitazone produced similar effects on a mouse embryonic fibroblast (MEF) cell line generated using the 3T3 protocol 41, with increased lipid accumulation and expression of PPARγ, FAS, and the brown fat specific protein UCP-1 in these cells.
  • These data suggest that BMP-6 and -7 can not only trigger commitment of mesenchymal stem cells to a brown adipocyte lineage, but also act in concert with other differentiating agents to induce characteristics of brown fat, including UCP-1, in more primitive fibroblastic cells.
  • Expression of Wnt-10a and necdin, two inhibitors of early adipogenesis, was significantly decreased by BMP-7 during the commitment phase in C3H10T1/2 cells (see FIGS. 1A-B). In addition, BMP-6 or -7 markedly induced differentiation of brown preadipocytes even in the absence of normally required induction cocktails. By contrast, under the same conditions, 3T3-L1, a white preadipocyte cell line, differentiated poorly in response to both BMP-6 and 7.
  • To determine if the effect of BMPs on brown preadipocyte differentiation could be observed in a primary culture system, stromo-vascular fraction (SVF) cells were isolated from interscapular BAT and nearby subcutaneous WAT close to BAT, and induced to undergo adipocyte differentiation in a serum free differentiation medium containing transferrin, dexamethasone, insulin and T3 supplemented with rosiglitazone (1 μg/ml), BMP-7 (3.3 nM) or vehicle. While rosiglitazone induced similar levels of adipocyte differentiation in SVFs derived from both fat depots within 3 days, BMP-7 had a specific effect on induction of differentiation only in the brown precursor cells at this time point. Thus, the effect of BMP-7 on promoting brown adipogenesis was not due to immortalization or some other factor unique to the brown preadipocyte cell lines, but occurred even in primary brown preadipocytes or their precursors present in the stromo-vascular factions.
  • Example 2 Effects of Exogenous BMP-7 Expression in Vivo
  • At the molecular level, the effects of BMPs in brown preadipocytes are mediated, at least in part, by increases in expression of the PPARγ coactivator-1 alpha (PGC-1α), which is linked to mitochondrial function, adipocyte cell fate decision and adaptive thermogenesis. To evaluate the effect of BMP expression on these parameters, adenoviral constructs for the expression of BMP-7 or LacZ (as a control) 5×108 plaque-forming units per gram body weight via tail veins were injected into 4-week and 12-week old C57BL/6 mice via the tail vein. These mice were sacrificed 15 days after adenoviral injection. Various adipose depots were collected and weighed.
  • Although BMP-7 expression had no significant effect on total body weight in either age group over this short period of observation (FIG. 2A), BMP-7 treatment did result in a significant increase in brown, but not white, fat mass in 4-week old mice (FIGS. 2B and 2C). In the 12-week old mice, there was also a trend toward increased BAT mass, but this did not quite reach statistical significance. Expression of UCP-1, on the other hand, was significantly increased in brown fat isolated from 12-w old BMP-7-treated animals (FIG. 2D).
  • As shown in FIGS. 2A-2C, adenoviral-mediated expression of BMP-7 in C57BL/6 mice results in a significant increase in brown, but not white, fat mass relative to total body weight and energy expenditure. These data demonstrate an important role of BMPs in enhancing brown adipocyte differentiation, and suggest it may be serve as a potential molecular switch between brown and white adipose tissues, providing a potential therapeutic approach for treatment of obesity.
  • Furthermore, adenoviral-mediated expression of BMP-7 in vivo leads to an increase in energy expenditure. As a consequence of the increase of BAT mass in 4-week old mice and the elevated UCP-1 expression in 12-week old animals by BMP-7 treatment, the BMP-7 adenovirally-treated mice showed a significant increase in energy expenditure in both light and dark cycles (FIGS. 2E and 2F).
  • The increase of BAT mass in 4-week old mice was not due to an increase in the size of the brown fat cells, but to an increase in cell number (data not shown), consistent with the hypothesis that BMP-7 can act as a growth and differentiation factor on the brown fat precursor cells. This could involve recruitment of pluripotent mesenchymal stem cells and/or promoting differentiation of existing committed preadipocytes present in the SVF, as suggested by the experiments described above. In addition, the increase of UCP-1 expression in BAT in 12-week old animals suggests that BMPs can directly increase thermogenic function of mature brown adipocytes in adult mammals.
  • Example 3 Implantation of Committed C3H10T1/2 Cells into Athymic Mice Results in Development of Both BAT and WAT
  • To evaluate the effects of implanting pluripotent mesenchymal stem cells into a living mammal, C3H10T1/2 cells were treated with 3.3 nM recombinant BMP-7 for 3 days, and then injected into athymic mice. 1.5×107 BMP-7 treated cells were injected subcutaneously into the sternum/thoracic regions of 5-w old BALB/c athymic mice; this region was chosen because it is generally free of any fat deposits. Mice were sacrificed 6 weeks after implantation.
  • Dissection and histological examination demonstrated that only cells treated with BMP-7 developed into tissues. As shown in FIGS. 3A-3D, fat deposits developed at the implant site with morphological characteristics of both brown fat (3C, compare with 3A, brown fat in wild type) and white fat (3D, compare with white fat shown in 3B). These results demonstrate that BMP-7-treated C3H10T1/2 implants develop into tissues containing both brown and white adipocytes.
  • To evaluate the effects of the BMP-7 treated pluripotent mesenchymal stem cell implants on metabolism, blood glucose and insulin levels were evaluated after recipient mice were fed glucose or insulin. Blood glucose concentrations were determined using Glucometer Elite XL (Bayer, Tarrytown, N.Y.). Insulin concentrations were determined using Insulin ELISA kit (Crystal Chem. Inc., Chicago, Ill.). The results, shown in FIGS. 4A and 4B, demonstrate that the recipient mice were more sensitive to insulin than were controls (FIG. 4A, p=0.00052), though blood insulin levels remained essentially the same (FIG. 4B). There was also no change in serum leptin or adiponectin levels. Thus, the amount of brown fat that developed from the implants was sufficient to affect insulin sensitivity in the recipient mice.
  • Example 4 The Effect of Different Bone Morphogenetic Proteins (BMPs) on Induction of Mitochondrial Biogenesis
  • Differentiation of BAT is accompanied by mitochondrial biogenesis, to the extent that the resultant abundant mitochondria and cytochromes cause the brown color of this tissue (Nedergaard et al, in Brown Adipose Tissue, Trayhurn and Nicholls, Eds. (Edward Arnold, Baltimore, 1986)). The coactivator PGC-1α plays a central role in integrating the transcriptional cascade regulating brown adipogenesis and mitochondrial function (J. Lin et al., Cell Metab 1:361-370 (2005); Puigserver et al., Cell 92:829-839 (1998); Wu et al., Cell 98:115-124 (1999); Puigserver and Spiegelman, Endocr. Rev. 24:78-90 (2003); Kelly and Scarpulla, Genes Dev. 18:357-368 (2004)). PGC-1α stimulates expression of nuclear respiratory factor (NRF)-1 and NRF-2, and coactivates the transcriptional function of these factors on expression of mitochondrial transcription factor A (Tfam), which is a direct regulator of mitochondrial replication and transcription (Wu et al., 1999, supra).
  • In the brown preadipocyte cell line, 3 days of treatment with either BMP-6 or BMP-7 in the presence of insulin and T3 was sufficient to enhance expression of PGC-1α and PGC-1β by 2- to 6-fold, respectively (FIG. 5A), accompanied by an approximately 2-fold increase in expression of NRF-1, Tfam and cytochrome C (Cyto C). PGC-1α is also known to enhance the transcriptional activity of PPARγ and thyroid hormone receptor on the UCP-1 promoter in brown adipocytes (Puigserver et al., Cell 92:829-839 (1998)). Thus, the powerful induction of UCP-1 protein expression by BMP-7 in brown adipocytes was likely to be mediated by PGC-1α. In contrast, under the same conditions, in white preadipocytes (3T3-L1 cells), BMP-6 and BMP-7 caused a 50% reduction in PGC-1α and NRF-2 gene expression and had no effect on expression of PGC-1β, NRF-1, Tfam and Cyto C. After 8 days of treatment, expression of genes involved in mitochondrial biogenesis in brown preadipocytes was further increased by BMP-6 and BMP-7 (FIG. 5B). At this time point, BMP-6 also induced some increase in expression of PGC-1α, PGC-1β, Tfam and Cyto C in 3T3-L1 white preadipocytes, but with no effect on UCP-1.
  • The early induction of expression of PGC-1α, PGC-1β, NRF-1, Tfam and Cyto C in brown preadipocytes by BMP-6 and BMP-7 suggests a direct effect of these BMPs on regulation of mitochondrial biogenesis and function. This effect of BMPs was further confirmed by electron microscopy of cells treated with insulin and T3 in the absence or presence of BMP-7 for 9 days. In the brown preadipocytes, BMP-7 markedly increased both number and size of mitochondria (FIG. 5C). Mitochondrial number appeared to be slightly increased in 3T3-L1 cells after 9 days of BMP-7 treatment, however, the effect in these cells was much more modest than was observed in the brown fat precursors (FIG. 5D). A similar modest increase in mitochondrial content has previously been observed in 3T3-L1 cells using conventional differentiation protocols (Wilson-Fritch et al., Mol Cell Biol 23:1085-1094 (2003), Wilson-Fritch et al., J Clin Invest 114:1281-1289 (2004)).
  • Thus, BMP-6 and BMP-7 have major effects to stimulate differentiation, mitochondrial biogenesis, and UCP-1 expression in brown preadipocytes, but only have minimal effect in 3T3-L1 white preadipocytes; and have no effect on induction of UCP-1 expression in the latter cells.
  • Other Embodiments
  • It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims (29)

1. A population of pluripotent mesenchymal stem cells, wherein the cells:
(i) are genetically engineered to express one or both of a BMP-6 or -7 polypeptide;
(ii) have been treated with one or both of a bone morphogenetic protein 6 (BMP 6) polypeptide or nucleic acid, or a bone morphogenetic protein 7 (BMP 7) polypeptide or nucleic acid; or
(iii) have an increased level of one or both of bone morphogenetic protein 6 (BMP 6) polypeptide or nucleic acid, or bone morphogenetic protein 7 (BMP 7) polypeptide or nucleic acid.
2. (canceled)
3. (canceled)
4. The population of BMP-activated pluripotent mesenchymal stem cells of claim 1, wherein the cells have been contacted with another compound selected from the group consisting of BMP-1, BMP-3, peroxisome proliferator-activated receptor gamma (PPARγ), Retinoid X receptor, alpha (RxRα), insulin, T3, a thiazolidinedione (TZD), vitamin A, retinoic acid, insulin, glucocorticoid or agonist thereof; Wingless-type (Wnt), Insulin-like Growth Factor-1 (TGF-1), Epidermal growth factor (EGF), Fibroblast growth factor (FGF), Transforming growth factor (TGF)-α, TGF-β, Tumor necrosis factor alpha (TNFα), Macrophage colony stimulating factor (MCSF), Vascular endothelial growth factor (VEGF) and/or Platelet-derived growth factor (PDGF).
5. The population of BMP-activated pluripotent mesenchymal stem cells of claim 1, wherein the cells are secondary cells.
6. The population of BMP-activated pluripotent mesenchymal stem cells of claim 1, wherein the cells are primary cells.
7. A therapeutic composition comprising the population of BMP-activated pluripotent mesenchymal stem cells of claim 1, and a pharmaceutically acceptable carrier.
8. A method of promoting brown adipogenesis in a subject, the method comprising administering to the subject a population of BMP-activated pluripotent mesenchymal stem cells of claim 1.
9. A method of preparing a population of BMP-activated pluripotent mesenchymal stem cells, the method comprising contacting a population of pluripotent mesenchymal stem cells with one or both of bone morphogenetic protein 6 (BMP-6) polypeptide or nucleic acid or bone morphogenetic protein 7 (BMP-7) polypeptide or nucleic acid.
10. The method of claim 9, further comprising obtaining a sample comprising a population of pluripotent mesenchymal stem cells from a subject.
11. The method of claim 10, further comprising enriching the sample to obtain a purified population of pluripotent mesenchymal stem cells.
12. The method of claim 11, wherein the purified population of pluripotent mesenchymal stem cells comprises at least 60% pluripotent mesenchymal stem cells.
13. A delivery system configured to allow the introduction of cells into a subject, wherein the delivery system comprises a reservoir containing a population of BMP-activated pluripotent mesenchymal stem cells of claim 1.
14. The delivery system of claim 13, further comprising a needle in fluid communication with the reservoir.
15. The delivery system of claim 13, wherein the population of BMP-activated pluripotent mesenchymal stem cells is in a pharmaceutically acceptable carrier.
16. A method of promoting brown adipogenesis in a subject, the method comprising administering to the subject a population of BMP-activated pluripotent mesenchymal stem cells that have been treated with one or both of a bone morphogenetic protein (BMP)-6 or BMP-7 polypeptide or nucleic acid, wherein said population of BMP-activated pluripotent mesenchymal stem cells, or their progeny, undergo brown adipogenesis.
17. The method of claim 16, wherein the cells are administered to decrease fat stores or weight in the subject.
18. The method of claim 16, wherein the cells are administered to enhance insulin sensitivity in the subject.
19. The methods of claim 16, wherein the BMP-activated population of pluripotent mesenchymal stem cells comprises a number of cells sufficient to promote brown adipogenesis in the subject.
20. The method of claim 16, wherein the BMP-activated pluripotent mesenchymal stem cells have been treated with a BMP-7 polypeptide.
21. The method of claim 16, wherein the BMP-activated pluripotent mesenchymal stem cells have been treated with a BMP-7 nucleic acid.
22. The method of claim 16, wherein the BMP-activated pluripotent mesenchymal stem cells have been treated with a BMP-6 polypeptide.
23. The method of claim 16, wherein the BMP-activated pluripotent mesenchymal stem cells have been treated with a BMP-6 nucleic acid.
24. The method of claim 16, wherein the cells have been contacted with another compound selected from the group consisting of BMP-1, BMP-3, peroxisome proliferator-activated receptor gamma (PPARγ), Retinoid X receptor, alpha (RxRα), insulin, T3, a thiazolidinedione (TZD), vitamin A, retinoic acid, insulin, glucocorticoid or agonist thereof, Wingless-type (Wnt), Insulin-like Growth Factor-1 (IGF-1), Epidermal growth factor (EGF), Fibroblast growth factor (FGF), Transforming growth factor (TGF)-α, TGF-β, Tumor necrosis factor alpha (TNFα), Macrophage colony stimulating factor (MCSF), Vascular endothelial growth factor (VEGF) and/or Platelet-derived growth factor (PDGF).
25. The method of claim 16, wherein the pluripotent mesenchymal stem cells are secondary cells.
26. The method of claim 16, wherein the pluripotent mesenchymal stem cells are primary cells.
27. The method of claim 26, wherein the primary cells are from the subject.
28. The method of claim 16, wherein the subject is an obese human subject.
29. The method of claim 16, wherein the subject is a food animal.
US11/914,425 2005-06-01 2006-06-01 Methods and compositions for inducing brown adipogenesis Abandoned US20100150885A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/914,425 US20100150885A1 (en) 2005-06-01 2006-06-01 Methods and compositions for inducing brown adipogenesis

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US68654205P 2005-06-01 2005-06-01
PCT/US2006/021120 WO2006130690A2 (en) 2005-06-01 2006-06-01 Methods and compositions for inducing brown adipogenesis
US11/914,425 US20100150885A1 (en) 2005-06-01 2006-06-01 Methods and compositions for inducing brown adipogenesis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/021120 A-371-Of-International WO2006130690A2 (en) 2005-06-01 2006-06-01 Methods and compositions for inducing brown adipogenesis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/219,157 Continuation US9346869B2 (en) 2005-06-01 2014-03-19 Methods and compositions for inducing brown adipogenesis

Publications (1)

Publication Number Publication Date
US20100150885A1 true US20100150885A1 (en) 2010-06-17

Family

ID=37482265

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/914,425 Abandoned US20100150885A1 (en) 2005-06-01 2006-06-01 Methods and compositions for inducing brown adipogenesis
US14/219,157 Active US9346869B2 (en) 2005-06-01 2014-03-19 Methods and compositions for inducing brown adipogenesis

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/219,157 Active US9346869B2 (en) 2005-06-01 2014-03-19 Methods and compositions for inducing brown adipogenesis

Country Status (2)

Country Link
US (2) US20100150885A1 (en)
WO (1) WO2006130690A2 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110038923A1 (en) * 2006-11-01 2011-02-17 Gary Weisinger Adipocyte-specific constructs and methods for inhibiting platelet-type 12 lipoxygenase expression
US20110064797A1 (en) * 2009-09-17 2011-03-17 The Ohio State University Capsule of thermogenic cells for treating a metabolic disease
US20110104133A1 (en) * 2008-05-06 2011-05-05 Joslin Diabetes Center, Inc. Methods and compositions for inducing brown adipogenesis
US20130209418A1 (en) * 2012-02-15 2013-08-15 Johnson & Johnson Regenerative Therapeutics, Llc Methods and composition related to brown adipose-like cells
US20140017789A1 (en) * 2011-06-29 2014-01-16 Francisco Javier Silva Brown Fat Cell Compositions and Methods
US20150216935A1 (en) * 2012-08-09 2015-08-06 Brown University Autologous Cell-Based Therapy for Treating Obesity
US9132169B2 (en) 2003-10-17 2015-09-15 Joslin Diabetes Center, Inc. Methods and compositions for modulating adipocyte function
US20150258148A1 (en) * 2012-06-06 2015-09-17 University Of Central Florida Research Foundation, Inc. Compositions, Methods and Systems for Cellular Differentiation from Stem Cells
EP2825638A4 (en) * 2012-03-12 2015-11-25 Univ Singapore Generation of brown adipose tissue (bat) from mesenchymal cells
WO2016007742A1 (en) * 2014-07-09 2016-01-14 Riester Scott M Treating rotator cuff conditions
WO2016016572A1 (en) * 2014-07-29 2016-02-04 Universite Pierre Et Marie Curie (Paris 6) Method for in vitro production of adipocyte progenitors and adipocytes
US9809798B2 (en) 2011-03-11 2017-11-07 National University Of Singapore Pericyte progenitors from peripheral blood
US10167449B2 (en) 2013-04-19 2019-01-01 Biorestorative Therapies, Inc. Human brown adipose derived stem cells and uses
US10596192B2 (en) 2015-07-16 2020-03-24 Mayo Foundation For Medical Education And Research Treating rotator cuff conditions
JP2021048866A (en) * 2014-02-24 2021-04-01 エナジーシス ファーマシューティカルズ, インコーポレイテッド Methods and compositions for inducing differentiation of human brown adipocyte progenitors

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008107422A1 (en) * 2007-03-02 2008-09-12 National University Of Ireland, Galway Osteopontin for the prediction and treatment of cardiovascular diseases
US20150004144A1 (en) * 2011-12-02 2015-01-01 The General Hospital Corporation Differentiation into brown adipocytes

Citations (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4997828A (en) * 1989-02-14 1991-03-05 The Rockefeller University Method of weight control by low level administration of cobalt protoporphyrin or cobalt mesoporphyrin
US5141905A (en) * 1986-07-01 1992-08-25 Rosen Vicki A Dna sequences encoding bmp-7 proteins
US5187076A (en) * 1986-07-01 1993-02-16 Genetics Institute, Inc. DNA sequences encoding BMP-6 proteins
US5318898A (en) * 1991-04-02 1994-06-07 Genetics Institute, Inc. Production of recombinant bone-inducing proteins
US5366875A (en) * 1986-07-01 1994-11-22 Genetics Institute, Inc. Methods for producing BMP-7 proteins
US5385887A (en) * 1993-09-10 1995-01-31 Genetics Institute, Inc. Formulations for delivery of osteogenic proteins
US5707112A (en) * 1996-05-03 1998-01-13 Magna Lomason Corporation Linear recliner with memory seatback dump mechanism
US5837539A (en) * 1990-11-16 1998-11-17 Osiris Therapeutics, Inc. Monoclonal antibodies for human mesenchymal stem cells
US6048964A (en) * 1995-12-12 2000-04-11 Stryker Corporation Compositions and therapeutic methods using morphogenic proteins and stimulatory factors
US20010051344A1 (en) * 1994-06-17 2001-12-13 Shalon Tidhar Dari Methods for constructing subarrays and uses thereof
US20020001825A1 (en) * 2000-03-31 2002-01-03 Nobuyuki Itoh Fibroblast growth factor-like molecules and uses thereof
US20020015771A1 (en) * 1999-04-28 2002-02-07 Michihiro Sugano Agent for increasing brown fat, comprising conjugated linoleic acid as active ingredient
US20020082413A1 (en) * 1997-05-30 2002-06-27 Dana-Farber Cancer Institute PGC-1, a novel brown fat PPARgamma coactivator
US20020090391A1 (en) * 1995-02-22 2002-07-11 Peter Geistlich Resorbable extracellular matrix for reconstruction of cartilage
US20020168765A1 (en) * 2001-03-23 2002-11-14 Prockop Darwin J. Early stage multipotential stem cells in colonies of bone marrow stromal cells
US20020169122A1 (en) * 2001-02-23 2002-11-14 Wyeth Chondrogenic potential of human bone marrow-derived CD105+ cells by BMP
US6593112B1 (en) * 1995-06-05 2003-07-15 Human Genome Sciences, Inc. Polynucleotides encoding fibroblast growth factor 15
US20030162706A1 (en) * 2002-02-08 2003-08-28 The Procter & Gamble Company Angiogenesis modulating proteins
US20030220238A1 (en) * 2001-01-22 2003-11-27 Adams Sean H. BFIT compositions and methods of use
US20040106142A1 (en) * 2002-11-12 2004-06-03 Becton, Dickinson And Company Diagnosis of sepsis or SIRS using biomarker profiles
US20040241145A1 (en) * 2001-08-22 2004-12-02 Jun-Ichi Hata Method of bone regeneration
US20050187154A1 (en) * 2003-10-17 2005-08-25 Joslin Diabetes Center, Inc. Methods and compositions for modulating adipocyte function
US20050261223A1 (en) * 2004-03-05 2005-11-24 Czech Michael P RIP140 regulation of glucose transport
US20050272649A1 (en) * 2002-08-28 2005-12-08 Hruska Keith A Conjoint administration of morphogens and ACE inhibitors in treatment of chronic renal failure
US20060246495A1 (en) * 2005-04-15 2006-11-02 Garrett James A Diagnosis of sepsis
US20070015701A1 (en) * 2005-06-01 2007-01-18 Samuel Zalipsky Macromolecular conjugates of bone morphogenetic protein-7
US7355049B2 (en) * 2003-06-24 2008-04-08 Hoffmann-La Roche Inc. Biaryloxymethylarenecarboxylic acids as glycogen synthase activator
US20080107755A1 (en) * 2006-04-24 2008-05-08 Alltech, Inc. Methods and compositions for altering cell function
US20080269150A1 (en) * 2005-11-14 2008-10-30 Laurent Bernard Fischer Intra-vascular kidney gene therapy with plasmid encoding BMP-7
US7459527B2 (en) * 2004-03-31 2008-12-02 Xencor, Inc. BMP-7 variants with improved properties
US20090220973A1 (en) * 2006-04-03 2009-09-03 Joslin Diabetes Center, Inc. Obesity and body fat distribution
US7825098B2 (en) * 2005-04-04 2010-11-02 Joslin Diabetes Center, Inc. Methods and compositions for modulating Necdin function
US20100291170A1 (en) * 1996-05-06 2010-11-18 Stryker Corporation Novel therapies for chronic renal failure

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5399677A (en) 1993-12-07 1995-03-21 Genetics Institute, Inc. Mutants of bone morphogenetic proteins
CA2283461A1 (en) * 1997-03-14 1998-09-17 Uab Research Foundation Adenoviral vectors with modified tropism
US6548739B2 (en) 1999-06-18 2003-04-15 City Of Hope Method for activating peroxisome proliferator activated receptor-γ
WO2002012887A2 (en) 2000-08-08 2002-02-14 Millennium Pharmaceuticals, Inc. Methods and compositions for the diagnosis and treatment of brown adipose cell disorders
EP1334365B1 (en) 2000-11-06 2007-02-21 Thrasos, Inc. Screening methods for bone morphogenetic mimetics
AU2002352148A1 (en) 2001-06-29 2003-03-03 Abbott Gmnh & Co. Kg Bis-aryl thiazole derivatives
AU2002331898A1 (en) 2001-09-24 2003-04-07 Board Of Supervisors Of Louisiana State Universityand Agricultural And Mechanical College Induction of brown adipocytes by transcription factor nfe2l2
AU2003228356A1 (en) 2002-03-22 2003-10-13 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for preventing obesity and obesity related disorders
US20050136429A1 (en) 2003-07-03 2005-06-23 Massachusetts Institute Of Technology SIRT1 modulation of adipogenesis and adipose function
US9631176B2 (en) 2003-11-04 2017-04-25 Biomaster, Inc. Method for preparing stem cells from fat tissue
ES2710099T3 (en) 2004-09-24 2019-04-23 Mesoblast Inc Progeny of multipotential expanded mesenchymal precursor cells (MEMP) and their uses
WO2007087053A2 (en) 2005-12-22 2007-08-02 Centocor, Inc Bmp-7 variant compositions, methods and uses
JP2009524425A (en) 2006-01-27 2009-07-02 プロスティミクス カンパニー リミテッド Method for mass production of growth factors using adipose-derived stem cells
EP1848162B1 (en) 2006-04-19 2017-03-08 Nokia Solutions and Networks GmbH & Co. KG Method to reduce the transmission latency in GSM/EDGE delay-sensitive applications

Patent Citations (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5141905A (en) * 1986-07-01 1992-08-25 Rosen Vicki A Dna sequences encoding bmp-7 proteins
US5187076A (en) * 1986-07-01 1993-02-16 Genetics Institute, Inc. DNA sequences encoding BMP-6 proteins
US5366875A (en) * 1986-07-01 1994-11-22 Genetics Institute, Inc. Methods for producing BMP-7 proteins
US4997828A (en) * 1989-02-14 1991-03-05 The Rockefeller University Method of weight control by low level administration of cobalt protoporphyrin or cobalt mesoporphyrin
US5837539A (en) * 1990-11-16 1998-11-17 Osiris Therapeutics, Inc. Monoclonal antibodies for human mesenchymal stem cells
US5318898A (en) * 1991-04-02 1994-06-07 Genetics Institute, Inc. Production of recombinant bone-inducing proteins
US5516654A (en) * 1991-04-02 1996-05-14 Genetics Institute, Inc. Production of recombinant bone-inducing proteins
US5385887A (en) * 1993-09-10 1995-01-31 Genetics Institute, Inc. Formulations for delivery of osteogenic proteins
US20010051344A1 (en) * 1994-06-17 2001-12-13 Shalon Tidhar Dari Methods for constructing subarrays and uses thereof
US20020090391A1 (en) * 1995-02-22 2002-07-11 Peter Geistlich Resorbable extracellular matrix for reconstruction of cartilage
US6593112B1 (en) * 1995-06-05 2003-07-15 Human Genome Sciences, Inc. Polynucleotides encoding fibroblast growth factor 15
US6048964A (en) * 1995-12-12 2000-04-11 Stryker Corporation Compositions and therapeutic methods using morphogenic proteins and stimulatory factors
US5707112A (en) * 1996-05-03 1998-01-13 Magna Lomason Corporation Linear recliner with memory seatback dump mechanism
US20100291170A1 (en) * 1996-05-06 2010-11-18 Stryker Corporation Novel therapies for chronic renal failure
US20030229204A1 (en) * 1997-05-30 2003-12-11 Spiegelman Bruce M. PCG-1, a novel brown fat PPARgamma coactivator
US20030073819A1 (en) * 1997-05-30 2003-04-17 Dana-Farber Cancer Institute PGC-1, a novel brown fat PPARgamma coactivator
US20020082413A1 (en) * 1997-05-30 2002-06-27 Dana-Farber Cancer Institute PGC-1, a novel brown fat PPARgamma coactivator
US20020015771A1 (en) * 1999-04-28 2002-02-07 Michihiro Sugano Agent for increasing brown fat, comprising conjugated linoleic acid as active ingredient
US20020001825A1 (en) * 2000-03-31 2002-01-03 Nobuyuki Itoh Fibroblast growth factor-like molecules and uses thereof
US20030220238A1 (en) * 2001-01-22 2003-11-27 Adams Sean H. BFIT compositions and methods of use
US20020169122A1 (en) * 2001-02-23 2002-11-14 Wyeth Chondrogenic potential of human bone marrow-derived CD105+ cells by BMP
US20020168765A1 (en) * 2001-03-23 2002-11-14 Prockop Darwin J. Early stage multipotential stem cells in colonies of bone marrow stromal cells
US20040241145A1 (en) * 2001-08-22 2004-12-02 Jun-Ichi Hata Method of bone regeneration
US20030162706A1 (en) * 2002-02-08 2003-08-28 The Procter & Gamble Company Angiogenesis modulating proteins
US20050272649A1 (en) * 2002-08-28 2005-12-08 Hruska Keith A Conjoint administration of morphogens and ACE inhibitors in treatment of chronic renal failure
US20040106142A1 (en) * 2002-11-12 2004-06-03 Becton, Dickinson And Company Diagnosis of sepsis or SIRS using biomarker profiles
US7355049B2 (en) * 2003-06-24 2008-04-08 Hoffmann-La Roche Inc. Biaryloxymethylarenecarboxylic acids as glycogen synthase activator
US20050187154A1 (en) * 2003-10-17 2005-08-25 Joslin Diabetes Center, Inc. Methods and compositions for modulating adipocyte function
US7576052B2 (en) * 2003-10-17 2009-08-18 Joslin Diabetes Center, Inc. Methods and compositions for modulating adipocyte function
US20110117049A1 (en) * 2003-10-17 2011-05-19 Joslin Diabetes Center, Inc. Methods and Compositions for Modulating Adipocyte Function
US20100098638A1 (en) * 2004-03-05 2010-04-22 University Of Massachusetts RIP140 Regulation of Diabetes
US20050261223A1 (en) * 2004-03-05 2005-11-24 Czech Michael P RIP140 regulation of glucose transport
US7459527B2 (en) * 2004-03-31 2008-12-02 Xencor, Inc. BMP-7 variants with improved properties
US7825098B2 (en) * 2005-04-04 2010-11-02 Joslin Diabetes Center, Inc. Methods and compositions for modulating Necdin function
US20060246495A1 (en) * 2005-04-15 2006-11-02 Garrett James A Diagnosis of sepsis
US20070015701A1 (en) * 2005-06-01 2007-01-18 Samuel Zalipsky Macromolecular conjugates of bone morphogenetic protein-7
US20080269150A1 (en) * 2005-11-14 2008-10-30 Laurent Bernard Fischer Intra-vascular kidney gene therapy with plasmid encoding BMP-7
US20090220973A1 (en) * 2006-04-03 2009-09-03 Joslin Diabetes Center, Inc. Obesity and body fat distribution
US20080107755A1 (en) * 2006-04-24 2008-05-08 Alltech, Inc. Methods and compositions for altering cell function

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9132169B2 (en) 2003-10-17 2015-09-15 Joslin Diabetes Center, Inc. Methods and compositions for modulating adipocyte function
US9663790B2 (en) 2006-11-01 2017-05-30 The Medical Research, Infrastructure, And Health Services Fund Of The Tel Aviv Medical Center Adipocyte-specific constructs and methods for inhibiting platelet-type 12 lipoxygenase expression
US9279127B2 (en) * 2006-11-01 2016-03-08 The Medical Research Fund At The Tel-Aviv Sourasky Medical Center Adipocyte-specific constructs and methods for inhibiting platelet-type 12 lipoxygenase expression
US20110038923A1 (en) * 2006-11-01 2011-02-17 Gary Weisinger Adipocyte-specific constructs and methods for inhibiting platelet-type 12 lipoxygenase expression
US20110104133A1 (en) * 2008-05-06 2011-05-05 Joslin Diabetes Center, Inc. Methods and compositions for inducing brown adipogenesis
US20110064797A1 (en) * 2009-09-17 2011-03-17 The Ohio State University Capsule of thermogenic cells for treating a metabolic disease
US8858990B2 (en) * 2009-09-17 2014-10-14 The Ohio State University Capsule of thermogenic cells for treating a metabolic disease
US9809798B2 (en) 2011-03-11 2017-11-07 National University Of Singapore Pericyte progenitors from peripheral blood
US9133438B2 (en) 2011-06-29 2015-09-15 Biorestorative Therapies, Inc. Brown fat cell compositions and methods
US20140023622A1 (en) * 2011-06-29 2014-01-23 Francisco Javier Silva Brown Fat Cell Compositions and Methods
US11851682B2 (en) 2011-06-29 2023-12-26 Biorestorative Therapies, Inc. Brown fat cell compositions and methods
US11066646B2 (en) 2011-06-29 2021-07-20 Biorestorative Therapies, Inc. Brown fat cell compositions and methods
US20140017789A1 (en) * 2011-06-29 2014-01-16 Francisco Javier Silva Brown Fat Cell Compositions and Methods
US10597638B2 (en) * 2011-06-29 2020-03-24 Biorestorative Therapies, Inc. Brown fat cell compositions and methods
US20130209418A1 (en) * 2012-02-15 2013-08-15 Johnson & Johnson Regenerative Therapeutics, Llc Methods and composition related to brown adipose-like cells
EP2825638A4 (en) * 2012-03-12 2015-11-25 Univ Singapore Generation of brown adipose tissue (bat) from mesenchymal cells
US20150258148A1 (en) * 2012-06-06 2015-09-17 University Of Central Florida Research Foundation, Inc. Compositions, Methods and Systems for Cellular Differentiation from Stem Cells
US20150216935A1 (en) * 2012-08-09 2015-08-06 Brown University Autologous Cell-Based Therapy for Treating Obesity
US10167449B2 (en) 2013-04-19 2019-01-01 Biorestorative Therapies, Inc. Human brown adipose derived stem cells and uses
US10941383B2 (en) 2013-04-19 2021-03-09 Biorestorative Therapies, Inc. Human brown adipose derived stem cells and uses
US11667892B2 (en) 2013-04-19 2023-06-06 Biorestorative Therapies, Inc. Human brown adipose derived stem cells and uses
JP2021048866A (en) * 2014-02-24 2021-04-01 エナジーシス ファーマシューティカルズ, インコーポレイテッド Methods and compositions for inducing differentiation of human brown adipocyte progenitors
JP7256165B2 (en) 2014-02-24 2023-04-11 エナジーシス ファーマシューティカルズ, インコーポレイテッド Methods and compositions for inducing differentiation of human brown adipocyte precursors
US10610570B2 (en) 2014-07-09 2020-04-07 Mayo Foundation For Medical Education And Research Treating rotator cuff conditions
WO2016007742A1 (en) * 2014-07-09 2016-01-14 Riester Scott M Treating rotator cuff conditions
JP2017522889A (en) * 2014-07-29 2017-08-17 ユニヴェルシテ・ピエール・エ・マリ・キュリ・(パリ・6) Methods for the production of preadipocytes and adipocytes in vitro
WO2016016572A1 (en) * 2014-07-29 2016-02-04 Universite Pierre Et Marie Curie (Paris 6) Method for in vitro production of adipocyte progenitors and adipocytes
US10596192B2 (en) 2015-07-16 2020-03-24 Mayo Foundation For Medical Education And Research Treating rotator cuff conditions
US11376280B2 (en) 2015-07-16 2022-07-05 Mayo Foundation For Medical Education And Research Treating rotator cuff conditions

Also Published As

Publication number Publication date
WO2006130690A3 (en) 2009-04-16
US9346869B2 (en) 2016-05-24
WO2006130690A2 (en) 2006-12-07
US20140296325A1 (en) 2014-10-02

Similar Documents

Publication Publication Date Title
US9346869B2 (en) Methods and compositions for inducing brown adipogenesis
EP2283119B1 (en) Methods and compositions for inducing brown adipogenesis
Zhao et al. Transforming growth factor β1 induces osteogenic differentiation of murine bone marrow stromal cells
Song et al. Transfection of mesenchymal stem cells with the FGF-2 gene improves their survival under hypoxic conditions
US9109045B2 (en) Mass producing growth factor using adipose derived adult stem cells
Schäffler et al. Concise review: adipose tissue-derived stromal cells—basic and clinical implications for novel cell-based therapies
Sato et al. Mechanical tension‐stress induces expression of bone morphogenetic protein (BMP)‐2 and BMP‐4, but not BMP‐6, BMP‐7, and GDF‐5 mRNA, during distraction osteogenesis
Lee et al. Enhancement of bone healing based on ex vivo gene therapy using human muscle-derived cells expressing bone morphogenetic protein 2
Barsby et al. Transforming growth factor beta3 promotes tendon differentiation of equine embryo-derived stem cells
Weller et al. Development and growth of mouse embryonic kidney in organ culture and modulation of development by soluble growth factor
Pakvasa et al. Neural EGF-like protein 1 (NELL-1): Signaling crosstalk in mesenchymal stem cells and applications in regenerative medicine
Cordonnier et al. Consistent osteoblastic differentiation of human mesenchymal stem cells with bone morphogenetic protein 4 and low serum
Kuhn et al. Fibroblast growth factor-2 and bone morphogenetic protein-2 have a synergistic stimulatory effect on bone formation in cell cultures from elderly mouse and human bone
Wang et al. Osteocalcin expressing cells from tendon sheaths in mice contribute to tendon repair by activating Hedgehog signaling
JP2002507407A (en) Heart-derived stem cells
WO2003103611A2 (en) Methods and compositions for the repair and/or regeneration of damaged myocardium
DK1490074T3 (en) Mixed-cell gene therapy
JPH08503198A (en) OP-3 induced morphogenesis
JP6349353B2 (en) Priming cell therapy
US20130078718A1 (en) Conversion of vascular endothelial cells into multipotent stem-like cells
Yang et al. A modified aggregate culture for chondrogenesis of human adipose-derived stem cells genetically modified with growth and differentiation factor 5
JP2001514026A (en) Engineered cells expressing bone morphogenetic proteins
Ahmed et al. Direct conversion of mouse embryonic fibroblast to osteoblast cells using hLMP-3 with Yamanaka factors
JP2000509377A (en) Methods for inducing bone formation
JP2001507354A (en) Treatment of mammalian myocardium with topical morphogens or morphogenically treated myogenic precursor cells

Legal Events

Date Code Title Description
AS Assignment

Owner name: JOSLIN DIABETES CENTER, INC.,MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TSENG, YU-HUA;KAHN, C. RONALD;SIGNING DATES FROM 20071205 TO 20071206;REEL/FRAME:020425/0331

STCB Information on status: application discontinuation

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