Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
J Bone Joint Surg Am ; 87(5): 1059-68, 2005 May.
Article in English | MEDLINE | ID: mdl-15866970

ABSTRACT

BACKGROUND: Demineralized bone matrix and recombinant human bone morphogenetic protein-2 or 7 (BMP-2 or BMP-7)-containing collagenous matrix have been shown to induce new bone formation in orthotopic and heterotopic sites. We examined the ability of subcutaneous implants of collagen combined with adenoviral vector containing the BMP-2 gene (AdBMP-2) to induce bone formation in rats. We also evaluated whether targeting the AdBMP-2 vector through an alternative receptor pathway, fibroblast growth factor (FGF), would increase the vector's potency. METHODS: In a time-course study, rat subcutaneous sites were implanted with (1) AdBMP-2 in rat-bone-derived collagen or (2) rat-bone-derived collagen alone. Samples were collected three, seven, fourteen, or thirty-five days after treatment. In a dose-response study, bone induction by AdBMP-2 in collagen (AdBMP-2/collagen) or by AdBMP-2 and FGF2 Fab' anti-adenovirus knob protein antibody in collagen (FGF2-AdBMP-2/collagen) was tested at fourteen days. Viral vector doses of 1 x 10(9) PN (viral particle number), 3 x 10(9) PN, 1 x10(10) PN, 3 x 10(10) PN, or 1 x 10(11) PN per implant were used. Equal amounts of collagen (25 mg) were used to formulate all implants. Explanted tissues were evaluated histologically to determine bone formation, specific activity of alkaline phosphatase, and calcium content. RESULTS: AdBMP-2/collagen implants induced robust bone formation. New bone was formed by the fourteenth day after implantation. In contrast, little or no bone was induced by the implant containing collagen alone. FGF2-AdBMP-2/collagen implants stimulated significantly more bone formation (p < 0.05) than did AdBMP-2/collagen implants, regardless of the dose of viral particles. CONCLUSIONS: Local delivery of AdBMP-2 in a collagen matrix rapidly induces bone formation, and targeting the virus through FGF receptors enhances the osteogenic potential of AdBMP-2.


Subject(s)
Bone Morphogenetic Proteins/administration & dosage , Implants, Experimental , Osteogenesis/drug effects , Recombinant Proteins/administration & dosage , Transforming Growth Factor beta/administration & dosage , Adenoviridae/genetics , Alkaline Phosphatase/metabolism , Animals , Bone Matrix , Bone Morphogenetic Protein 2 , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblast Growth Factor 2/physiology , Genetic Vectors , Rats , Rats, Long-Evans
2.
J Bone Joint Surg Am ; 83-A Suppl 1(Pt 1): S70-8, 2001.
Article in English | MEDLINE | ID: mdl-11263669

ABSTRACT

BACKGROUND: The vasculature is an important component in the musculoskeletal system, and vascularization is a key event in the development of normal cartilage and bone formation. Blood vessels deliver nutrients, oxygen, and precursor cells to maintain the structural and functional integrity of joints and soft and hard tissues. Therefore, agents that help to inhibit proliferation and retain the phenotype of vascular smooth muscle cells (SMCs) are of critical importance. In this study, we examined the capacity of bone morphogenetic protein-7 (BMP-7) to inhibit the proliferation of SMCs and maintain their phenotype. METHODS: A thymidine-incorporation assay was used to monitor the proliferative activity of SMCs on stimulation with platelet-derived growth factor (PDGF) and transforming growth factor-beta (TGF-beta), agents known to be stimulatory for these cells. Reverse transcriptase-polymerase chain reaction (RT-PCR), Northern blot analysis, and enzyme-linked immunosorbent assay (ELISA) were used to monitor the modulation of various genes and gene products. Immunolocalization of SMC specific markers was also performed. RESULTS: BMP-7 inhibited both serum-stimulated and growth factor-induced (PDGF-BB and TGF-beta1) SMC growth, as measured by 3H-thymidine uptake and cell number, in primary human aortic smooth muscle (HASM) cell cultures. The addition of BMP-7 stimulated the expression of developmentally regulated as well as SMC-specific markers, namely, Id-1 and Id-2, alpha-actin, and SMC-specific heavy-chain myosin, as examined by semiquantitative and quantitative RT-PCR and by Northern blot analysis. Additionally, BMP-7 exhibited anti-inflammatory activity by downregulating intercellular adhesion molecule-1 (ICAM-1) expression. The collagen type III/I ratio that becomes lower with the transdifferentiation of SMCs into myofibroblasts is maintained in BMP-7-treated cultures compared with untreated controls. Studies on the mechanism of action indicate that BMP-7 treatment induces cyclin-dependent kinase-2 inhibitor, p21, which was inhibited during PDGF-BB-induced proliferation of SMCs. Finally, BMP-7 upregulates the expression of the inhibitory Smads, Smad6 and Smad7, which are known to inhibit TGF-beta superfamily signaling. CONCLUSIONS: These results suggest that BMP-7 maintains the expression of the vascular SMC phenotype. Thus, BMP-7 may prevent vascular proliferative disorders and potentially could act as a palliative agent following damage to the vasculature. CLINICAL RELEVANCE: In musculoskeletal disorders in which the vasculature plays an important role, BMP-7 may be of benefit as an anti-inflammatory and anti-proliferative agent for vascular endothelium and help maintain vascular integrity.


Subject(s)
Bone Morphogenetic Proteins/physiology , Muscle, Smooth, Vascular/physiology , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 7 , Cells, Cultured , Down-Regulation/physiology , Enzyme-Linked Immunosorbent Assay , Humans , Intercellular Adhesion Molecule-1/physiology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/physiology
3.
J Bone Miner Res ; 15(9): 1798-809, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10976999

ABSTRACT

To investigate the long-term efficacy of irradiated recombinant human osteogenic protein 1 (hOP-1) in bone regeneration and morphogenesis, hOP-1 was combined with a bovine collagenous matrix carrier (0, 0.1, 0.5, and 2.5 mg hOP-1/g of matrix), sterilized with 2.5 Mrads of y-irradiation, and implanted in 80 calvarial defects in 20 adult baboons (Papio ursinus). The relative efficacy of partially purified bone-derived baboon bone morphogenetic proteins (BMPs), known to contain several osteogenic proteins, was compared with the recombinant hOP-1 device in an additional four baboons. Histology and histomorphometry on serial undecalcified sections prepared from the specimens harvested on day 90 and day 365 showed that gamma-irradiated hOP-1 devices induced regeneration of the calvarial defects by day 90, although with reduced bone area compared with a previous published series of calvarial defects treated with nonirradiated hOP-1 devices. One year after application of the irradiated hOP-1 devices, bone and osteoid volumes and generated bone tissue areas were comparable with nonirradiated hOP-1 specimens. Moreover, 365 days after healing regenerates induced by 0.5 mg and 2.5 mg of irradiated hOP-1 devices showed greater amounts of bone and osteoid volumes when compared with those induced by nonirradiated hOP-1 devices. On day 90, defects treated with 0.1 mg and 0.5 mg of bone-derived baboon BMPs, combined with irradiated matrix, showed significantly less bone compared with defects receiving irradiated devices containing 0.1 mg and 0.5 mg hOP-1; 2.5 mg of partially purified BMPs induced bone and osteoid volumes comparable with the 0.1-mg and 0.5-mg hOP-1 devices. Control specimens of y-irradiated collagenous matrix without hOP-1 displayed a nearly 2-fold reduction in osteoconductive bone repair when compared with nonirradiated controls. These findings suggest that the reduction in bone volume and bone tissue area on day 90 may be caused by a reduced performance of the irradiated collagenous matrix substratum rather than to a reduction in the biological activity of the irradiated recombinant osteogenic protein. This is supported by the results of in vitro and in vivo studies performed to determine the structural integrity of the recovered gamma-irradiated hOP-1 before application in the baboon. Recoveries by high-performance liquid chromatography (HPLC) and sodium dodecyl sulfate/ polyacrylamide gel electrophoresis (SDS/PAGE)/immunoblot analyses indicated that doses of 2.5-3 Mrads of gamma-irradiation did not significantly affect the structural integrity of the recovered hOP-1. Biological activity of the recovered hOP-1 was confirmed in vitro by showing induction of alkaline phosphatase activity in rat osteosarcoma cells (ROS) and in vivo by de novo endochondral bone formation in the subcutaneous space of the rat. These findings in the adult primate indicate that a single application of gamma-irradiated hOP-1 combined with the irradiated xenogeneic bovine collagenous matrix carrier is effective in regenerating and maintaining the architecture of the induced bone at doses of 0.5 mg/g and 2.5 mg/g of carrier matrix.


Subject(s)
Bone Development/drug effects , Bone Matrix/transplantation , Bone Morphogenetic Proteins/pharmacology , Bone Regeneration/drug effects , Collagen/metabolism , Papio/physiology , Skull/drug effects , Transforming Growth Factor beta , Alkaline Phosphatase/metabolism , Animals , Bone Matrix/metabolism , Bone Matrix/radiation effects , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/administration & dosage , Bone Morphogenetic Proteins/radiation effects , Cattle , Collagen/radiation effects , Embryonic Induction/drug effects , Gamma Rays , Histocytochemistry , Humans , Immunoblotting , Models, Animal , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Recombinant Proteins/radiation effects , Skull/abnormalities , Skull/physiology , Time Factors , Transplantation, Heterologous
4.
Am J Physiol Renal Physiol ; 279(1): F130-43, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10894795

ABSTRACT

Unilateral ureteral obstruction (UUO) is a model of renal injury characterized by progressive tubulointerstitial fibrosis and renal damage, while relatively sparing the glomerulus and not producing hypertension or abnormalities in lipid metabolism. Tubulointerstitial fibrosis is a major component of several kidney diseases associated with the progression to end-stage renal failure. Here we report that when a critical renal developmental morphogen, osteogenic protein-1 (OP-1; 100 or 300 microg/kg body wt), is administered at the time of UUO and every other day thereafter, interstitial inflammation and fibrogenesis are prevented, leading to preservation of renal function during the first 5 days after obstruction. Compared with angiotensin-converting enzyme inhibition with enalapril treatment, OP-1 was more effective in preventing tubulointerstitial fibrosis and in preserving renal function. The mechanism of OP-1- induced renal protection was associated with prevention of tubular atrophy, an effect not shared with enalapril, and was related to preservation of tubular epithelial integrity. OP-1 blocked the stimulation of epithelial cell apoptosis produced by UUO, which promoted maintenance of tubular epithelial integrity. OP-1 preserved renal blood flow (RBF) during UUO, but enalapril also stimulated RBF. Thus OP-1 treatment inhibited tubular epithelial disruption stimulated by the renal injury of UUO, preventing tubular atrophy and diminishing the activation of tubulointerstitial inflammation and fibrosis and preserving renal function.


Subject(s)
Bone Morphogenetic Proteins/therapeutic use , Fibrosis/complications , Fibrosis/drug therapy , Kidney/pathology , Transforming Growth Factor beta , Ureteral Obstruction/complications , Actins/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Atrophy/drug therapy , Atrophy/pathology , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/administration & dosage , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/pharmacology , Cell Size/drug effects , Collagen/metabolism , Enalapril/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis/metabolism , Fibrosis/pathology , Immunohistochemistry , Inflammation/complications , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Kidney/drug effects , Kidney/metabolism , Kidney/physiopathology , Macrophages/drug effects , Macrophages/immunology , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects
5.
Cancer Res ; 60(11): 2840-4, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10850425

ABSTRACT

Bone morphogenetic proteins (BMPs) are potential regulators of prostate cancer cell growth and metastasis that signal through an interaction with BMP membrane receptors (BMPRs) type I and type II. In the present study, Western blot and immunohistochemical analysis of BMPRs were carried out in benign and malignant human prostate tissues to explain the loss of BMP response in human prostate cancer cells. The results demonstrated that the benign prostate specimens expressed high levels of all three BMPRs. In normal prostate, BMPRs were localized predominantly to epithelial cells. Among prostate cancer specimens, well-differentiated cancers were positive for the expression of BMPR-II, BMPR-IA, and BMPR-IB, for the most part. In contrast, only 1 of 10 poorly differentiated prostate cancer cases was positive for each of the three BMPRs (P < 0.005 for all three receptors). Taken together, these results indicate that human prostate cancer cells frequently exhibit loss of expression of BMPRs and suggest that loss of BMPRs may play an important role during the progression of prostate cancer.


Subject(s)
Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Receptors, Growth Factor/biosynthesis , Blotting, Western , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Protein Receptors, Type II , Disease Progression , Epithelial Cells/metabolism , Humans , Immunohistochemistry , Male , Prostate/metabolism , Prostatic Neoplasms/pathology , Transforming Growth Factors/metabolism
6.
J Cell Physiol ; 184(1): 37-45, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10825232

ABSTRACT

Vascular proliferative disorders are characterized by migration and proliferation of vascular smooth muscle cells (SMCs), loss of expression of SMC phenotype, and enhanced extracellular matrix synthesis (e.g., type I collagen). We report here that bone morphogenetic protein-7 (BMP-7), a member of the transforming growth factor-beta (TGF-beta) superfamily, is capable of inhibiting both serum-stimulated and growth factor-induced (platelet-derived growth factor [PDGF-BB] and TGF-beta1) cell growth as measured by (3)H-thymidine uptake into DNA synthesis and cell number in primary human aortic smooth muscle (HASM) cell cultures. Concomitantly, addition of BMP-7 stimulates the expression of SMC-specific markers, namely alpha-actin and heavy chain myosin as examined by RT-PCR and Northern blot analyses. The collagen type III/I ratio that becomes lower with the transdifferentiation of SMCs into myofibroblasts is also maintained in BMP-7-treated cultures as compared to untreated controls. Studies on the mechanism of action indicate that BMP-7 treatment inhibits cyclin-dependent kinase 2 (cdk-2) that was stimulated during PDGF-BB-induced proliferation of SMCs and upregulates the expression of the inhibitory Smad, Smad6, which was shown to inhibit TGF-beta superfamily signaling. These results collectively suggest that BMP-7 maintains the expression of vascular SMC phenotype and may prevent vascular proliferative disorders, thus potentially acting as a palliative after damage to the vascular integrity.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Gene Expression Regulation/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Actins/genetics , Aorta , Apoptosis/drug effects , Becaplermin , Bone Morphogenetic Protein 7 , Bone Morphogenetic Protein Receptors, Type II , Cell Division/drug effects , Cells, Cultured , Humans , Muscle, Smooth, Vascular/drug effects , Myosin Heavy Chains/genetics , Platelet-Derived Growth Factor/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-sis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology
7.
J Biol Chem ; 275(9): 6075-9, 2000 Mar 03.
Article in English | MEDLINE | ID: mdl-10692396

ABSTRACT

Smad6 is an inhibitory Smad that is induced by bone morphogenetic proteins (BMPs) and interferes with BMP signaling. We have isolated the mouse Smad6 promoter and identified the regions responsible for transcriptional activation by BMPs. The proximal BMP-responsive element (PBE) in the Smad6 promoter is important for the transcriptional activation by BMPs and contains a 28-base pair GC-rich sequence including four overlapping copies of the GCCGnCGC-like motif, which is a binding site for Drosophila Mad and Medea. We generated a luciferase reporter construct (3GC2-Lux) containing three repeats of the GC-rich sequence derived from the PBE. BMPs and BMP receptors induced transcriptional activation of 3GC2-Lux in various cell types, and this activation was enhanced by cotransfection of BMP-responsive Smads, i.e. Smad1 or Smad5. Moreover, direct DNA binding of BMP-responsive Smads and common-partner Smad4 to the GC-rich sequence of PBE was observed. These results indicate that the expression of Smad6 is regulated by the effects of BMP-activated Smad1/5 on the Smad6 promoter.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Phosphoproteins/metabolism , Promoter Regions, Genetic , Receptors, Growth Factor , Trans-Activators/genetics , Trans-Activators/metabolism , Animals , Binding Sites/genetics , Bone Morphogenetic Protein Receptors , Bone Morphogenetic Proteins/metabolism , Cell Line , Cloning, Molecular , Gene Expression Regulation , Genes, Reporter , Mice , Receptors, Cell Surface/metabolism , Regulatory Sequences, Nucleic Acid , Signal Transduction , Smad Proteins , Smad1 Protein , Smad5 Protein , Smad6 Protein , Transcriptional Activation , Transfection
8.
Mol Biol Cell ; 10(11): 3801-13, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10564272

ABSTRACT

The biological effects of type I serine/threonine kinase receptors and Smad proteins were examined using an adenovirus-based vector system. Constitutively active forms of bone morphogenetic protein (BMP) type I receptors (BMPR-IA and BMPR-IB; BMPR-I group) and those of activin receptor-like kinase (ALK)-1 and ALK-2 (ALK-1 group) induced alkaline phosphatase activity in C2C12 cells. Receptor-regulated Smads (R-Smads) that act in the BMP pathways, such as Smad1 and Smad5, also induced the alkaline phosphatase activity in C2C12 cells. BMP-6 dramatically enhanced alkaline phosphatase activity induced by Smad1 or Smad5, probably because of the nuclear translocation of R-Smads triggered by the ligand. Inhibitory Smads, i.e., Smad6 and Smad7, repressed the alkaline phosphatase activity induced by BMP-6 or the type I receptors. Chondrogenic differentiation of ATDC5 cells was induced by the receptors of the BMPR-I group but not by those of the ALK-1 group. However, kinase-inactive forms of the receptors of the ALK-1 and BMPR-I groups blocked chondrogenic differentiation. Although R-Smads failed to induce cartilage nodule formation, inhibitory Smads blocked it. Osteoblast differentiation induced by BMPs is thus mediated mainly via the Smad-signaling pathway, whereas chondrogenic differentiation may be transmitted by Smad-dependent and independent pathways.


Subject(s)
DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Growth Factor/metabolism , Trans-Activators/metabolism , Activin Receptors , Adenoviridae/genetics , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type I , Cell Differentiation , Cell Line , Chondrocytes , Genetic Vectors , Histocytochemistry , Mice , Osteoblasts , Phosphoproteins/metabolism , Phosphorylation , Smad Proteins , Smad1 Protein , Smad5 Protein , Smad6 Protein , Smad7 Protein
9.
J Cell Sci ; 112 ( Pt 20): 3519-27, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10504300

ABSTRACT

Bone morphogenetic protein (BMP)-6 is a member of the transforming growth factor (TGF)-(&bgr;) superfamily, and is most similar to BMP-5, osteogenic protein (OP)-1/BMP-7, and OP-2/BMP-8. In the present study, we characterized the endogenous BMP-6 signaling pathway during osteoblast differentiation. BMP-6 strongly induced alkaline phosphatase (ALP) activity in cells of osteoblast lineage, including C2C12 cells, MC3T3-E1 cells, and ROB-C26 cells. The profile of binding of BMP-6 to type I and type II receptors was similar to that of OP-1/BMP-7 in C2C12 cells and MC3T3-E1 cells; BMP-6 strongly bound to activin receptor-like kinase (ALK)-2 (also termed ActR-I), together with type II receptors, i.e. BMP type II receptor (BMPR-II) and activin type II receptor (ActR-II). In addition, BMP-6 weakly bound to BMPR-IA (ALK-3), to which BMP-2 also bound. In contrast, binding of BMP-6 to BMPR-IB (ALK-6), and less efficiently to ALK-2 and BMPR-IA, together with BMPR-II was detected in ROB-C26 cells. Intracellular signalling was further studied using C2C12 and MC3T3-E1 cells. Among the receptor-regulated Smads activated by BMP receptors, BMP-6 strongly induced phosphorylation and nuclear accumulation of Smad5, and less efficiently those of Smad1. However, Smad8 was constitutively phosphorylated, and no further phosphorylation or nuclear accumulation of Smad8 by BMP-6 was observed. These findings indicate that in the process of differentiation to osteoblasts, BMP-6 binds to ALK-2 as well as other type I receptors, and transduces signals mainly through Smad5 and possibly through Smad1.


Subject(s)
Bone Morphogenetic Proteins/physiology , Osteoblasts/cytology , 3T3 Cells , Activin Receptors, Type I , Activin Receptors, Type II , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein 6 , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Protein Receptors, Type II , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation , Cell Division , Cell Line , Kinetics , Mice , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Receptors, Growth Factor/physiology , Recombinant Proteins/metabolism , Signal Transduction/physiology , Transfection
10.
J Bone Miner Res ; 14(7): 1145-52, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10404014

ABSTRACT

Members of the transforming growth factor-beta (TGF-beta) family transduce signals from the cell membrane to the nucleus via specific type I and type II receptors and Smad proteins. Smad1 and Smad5 mediate intracellular signaling of bone morphogenetic protein (BMP), whereas Smad2 and Smad3 transduce TGF-beta signaling. Smad4 is a common mediator required for both pathways. Smad6 and Smad7 inhibit signaling by members of the TGF-beta superfamily. Here, we examined the expression of Smad1 to Smad7 proteins during endochondral ossification of epiphyseal plate of growing rats using immunohistochemical techniques. The expression of Smad proteins was correlated with the expression of TGF-beta1 and its receptors, and BMP-2/4 and BMP receptors. The results show that TGF-beta1 and BMP-2/4 were actively expressed in chondrocytes that are undergoing proliferation and maturation, which overlaps with expression of their corresponding type I and type II receptors. The Smads, however, exhibited a distinct expression pattern, respectively. For example, Smad1 and Smad5 were highly expressed in proliferating chondrocytes and in those chondrocytes that are undergoing maturation. The TGF-beta/activin-restricted Smads were also expressed in a nearly complementary fashion; Smad2 was strongly expressed in proliferating chondrocytes, whereas Smad3 was strongly observed in maturing chondrocytes. Smad4 was broadly expressed in all zones of epiphyseal plate. Inhibitory Smads, Smad6 and Smad7, were strongly expressed in the zone of cartilage that contained mature chondrocytes. Our findings show a colocalization of the pathway-restricted and inhibitory Smads with activating ligands or ligands whose action they antagonize and their receptors in various zones of epiphyseal growth plate, suggesting that TGF-beta superfamily Smad signaling pathways plays a morphogenic role during endochondral bone formation.


Subject(s)
Activin Receptors, Type I , DNA-Binding Proteins/metabolism , Growth Plate/metabolism , Osteogenesis/physiology , Second Messenger Systems/physiology , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Protein Receptors, Type II , Bone Morphogenetic Proteins/metabolism , Cell Nucleus/metabolism , Chondrocytes/metabolism , Growth Plate/physiology , Immunohistochemistry , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Growth Factor/metabolism , Receptors, Transforming Growth Factor beta/metabolism
11.
J Clin Invest ; 102(1): 202-14, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9649574

ABSTRACT

We have shown that osteogenic protein-1 (OP-1) (bone morphogenetic protein-7) is responsible for the induction of nephrogenic mesenchyme during embryonic kidney development. Gene knock-out studies showed that OP-1 null mutant mice die of renal failure within the first day of postnatal life. In the present study, we evaluated the effect of recombinant human OP-1 for the treatment of acute renal failure after 60 min bilateral renal artery occlusion in rats. Bioavailability studies in normal rats indicate that approximately 1.4 microg OP-1/ml is available in the circulation 1 min after intravenous administration of 250 microg/kg, which then declines steadily with a half life of 30 min. About 0.5% of the administered OP-1 dose/g tissue is targeted for OP-1 receptors in the kidney. We show that OP-1 preserves kidney function, as determined by reduced blood urea nitrogen and serum creatinine, and increased survival rate when administered 10 min before or 1 or 16 h after ischemia, and then at 24-h intervals up to 72 h after reperfusion. Histochemical and molecular analyses demonstrate that OP-1: (a) minimizes infarction and cell necrosis, and decreases the number of plugged tubules; (b) suppresses inflammation by downregulating the expression of intercellular adhesive molecule, and prevents the accumulation and activity of neutrophils; (c) maintains the expression of the vascular smooth muscle cell phenotype in pericellular capillaries; and (d) reduces programmed cell death during the recovery. Collectively, these data suggest that OP-1 prevents the loss of kidney function associated with ischemic injury and may provide a basis for the treatment of acute renal failure.


Subject(s)
Acute Kidney Injury/drug therapy , Bone Morphogenetic Proteins/pharmacology , Ischemia/drug therapy , Kidney/blood supply , Transforming Growth Factor beta , Animals , Apoptosis , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Growth Substances/genetics , Humans , Intercellular Adhesion Molecule-1/analysis , Kidney/drug effects , Male , RNA, Messenger/analysis , Rats , Rats, Wistar , Recombinant Proteins/therapeutic use
12.
J Biol Chem ; 273(27): 17079-85, 1998 Jul 03.
Article in English | MEDLINE | ID: mdl-9642273

ABSTRACT

Bone morphogenetic proteins (BMPs)/osteogenic proteins (OPs), members of the transforming growth factor-beta superfamily, have a wide variety of effects on many cell types including osteoblasts and chondroblasts, and play critical roles in embryonic development. BMPs transduce their effects through binding to two different types of serine/threonine kinase receptors, type I and type II. Signaling by these receptors is mediated by the recently identified Smad proteins. Despite the rapid progress in understanding of the signaling mechanism downstream of BMP receptors, the target genes of BMPs are poorly understood in mammals. Here we identified a novel gene, termed BMP/OP-responsive gene (BORG), in C2C12 mouse myoblast cell line which trans-differentiates into osteoblastic cells in response to BMPs. Expression of BORG was dramatically induced in C2C12 cells by the treatment with BMP-2 or OP-1 within 2 h and peaked at 12-24 h, whereas transforming growth factor-beta had a minimal effect. BMP-dependent expression of BORG was also detected in other cell types which are known to respond to BMPs, suggesting that BORG is a common target gene of BMPs. Cloning and sequence analysis of BORG cDNA and the genomic clones revealed that, unexpectedly, the transcript of BORG lacks any extensive open reading frames and contains a cluster of multiple interspersed repetitive sequences in its middle part. The unusual structural features suggested that BORG may function as a noncoding RNA, although it is spliced and polyadenylated as authentic protein-coding mRNAs. Together with the observation that transfection of antisense oligonucleotides of BORG partially inhibited BMP-induced differentiation in C2C12 cells, it is possible that a new class of RNA molecules may have certain roles in the differentiation process induced by BMPs.


Subject(s)
Bone Morphogenetic Proteins/metabolism , RNA/metabolism , Animals , Base Sequence , Blotting, Northern , Cell Line , Cloning, Molecular , DNA, Complementary , Exons , Introns , Mice , Mice, Inbred C3H , Molecular Sequence Data , Oligonucleotides, Antisense/pharmacology , RNA/genetics
13.
Bone ; 22(6): 605-12, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9626398

ABSTRACT

Bone morphogenetic proteins (BMPs) and their receptors (BMPRs) are thought to play an important role in bone morphogenesis. The purpose of this study was to determine the locations of BMP-2/-4, osteogenic protein-1 (OP-1, also termed BMP-7), and BMP type II receptor (BMPR-II) during rat fracture healing by immunostaining, and thereby elucidate the possible roles of the BMPs and BMPR-II in intramembranous ossification and endochondral ossification. In the early stage of fracture repair, the expression of BMP-2/-4 and OP-1 was strongly induced in the thickened periosteum near the fracture ends, and coincided with an enhanced expression of BMPR-II. On day 7 after fracture, staining for BMP-2/-4 and OP-1 immunostaining was increased in various types of chondrocytes, and was strong in fibroblast-like spindle cells and proliferating chondrocytes in endochondral bone. On day 14 after fracture, staining with OP-1 antibody disappeared in proliferating and mature chondrocytes, while BMP-2/-4 staining continued in various types of chondrocytes until the late stage. In the newly formed trabecular bone, BMP-2/-4 and OP-1 were present at various levels. BMPR-II was actively expressed in both intramembranous ossification and endochondral ossification. Additionally, immunostaining for BMP-2/-4 and OP-1 was observed in multinucleated osteoclast-like cells on the newly formed trabecular bone, along with BMPR-II. In reference to our previous study of BMP type I receptors (BMPR-IA and BMPR-IB), BMPR-II was found to be co-localized with BMPR-IA and BMPR-IB. BMP-2/-4 and OP-1 antibodies exhibited distinct and overlapping immunostaining patterns during fracture repair. OP-1 may act predominantly in the initial phase of endochondral ossification, while BMP-2/-4 acts throughout this process. Thus, these findings suggested that BMPs acting through their BMP receptors may play major roles in modulating the sequential events leading to bone formation.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Fracture Healing/physiology , Protein Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Protein Receptors, Type II , Bone Morphogenetic Proteins/biosynthesis , Calcification, Physiologic/physiology , Cell Division , Chondrocytes/metabolism , Fibroblasts/metabolism , Immunohistochemistry , Osteoblasts/metabolism , Osteoclasts/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Rats , Rats, Wistar , Transforming Growth Factor beta/biosynthesis
14.
Biochem Biophys Res Commun ; 244(1): 26-9, 1998 Mar 06.
Article in English | MEDLINE | ID: mdl-9514869

ABSTRACT

Members of the transforming growth factor-beta (TGF-beta) superfamily transduce signals via Smad proteins. Smad2 and Smad3 mediate TGF-beta signaling, whereas Smad1 and Smad5 transduce bone morphogenetic protein (BMP) signals. Smad4 is a common mediator required for both pathways. Smad6 and Smad7 are recently identified members in the Smad family; they inhibit the signaling activity of the other Smad proteins. Here we show that expression of the Smad6 mRNA is dramatically induced by BMP-2 or osteogenic protein-1 (OP-1)/BMP-7 in various cells. BMP-2 induced expression of Smad7 in one cell type, although much less potently than that of Smad6. Smad6 message was induced by TGF-beta 1 in TGF-beta 1-responsive Mv1Lu cells, but the induction was transient in contrast to the induction by BMPs. These results indicate that Smad6 may form a feedback loop to regulate the signaling activity of BMPs.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , RNA, Messenger/biosynthesis , Trans-Activators , Animals , Bone Morphogenetic Protein 2 , CHO Cells , Cell Line , Cricetinae , DNA-Binding Proteins/drug effects , Gene Expression Regulation/drug effects , Humans , Ligands , Lung , Mice , Mice, Inbred C3H , Mink , RNA, Messenger/drug effects , Smad6 Protein , Transforming Growth Factor beta/pharmacology
16.
Mol Endocrinol ; 11(12): 1832-45, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9369451

ABSTRACT

Bone morphogenetic proteins induce chondrogenesis and osteogenesis in vivo. To investigate molecular mechanisms involved in chondrocyte induction, we examined the effect of osteogenic protein (OP)-1/bone morphogenetic protein-7 on the collagen X promoter. In rat calvaria-derived chondrogenic C5.18 cells, OP-1 up-regulates collagen X mRNA levels and its promoter activity in a cell type- specific manner. Deletion analysis localizes the OP-1 response region to 33 bp (-310/-278), which confers OP-1 responsiveness to both the minimal homologous and heterologous Rous sarcoma virus promoter. Transforming growth factor-beta2 or activin, which up-regulates the expression of a transforming growth factor-beta-inducible p3TP-Lux construct, has little effect on collagen X mRNA and on this 33-bp region. Mutational analysis shows that both an AP-1 like sequence (-294/-285, TGAATCATCA) and an A/T-rich myocyte enhancer factor (MEF)-2 like sequence (-310/-298, TTAAAAATAAAAA) in the 33-bp region are necessary for the OP-1 effect. Gel shift assays show interaction of distinct nuclear proteins from C5.18 cells with the AP-1-like and the MEF-2-like sequences. OP-1 rapidly induces nuclear protein interaction with the MEF-2-like sequence but not with the AP-1 like sequence. MEF-2-like binding activity induced by OP-1 is distinct from the MEF-2 family proteins present in C2C12 myoblasts, in which OP-1 does not induce collagen X mRNA or up-regulate its promoter activity. In conclusion, we identified a specific response region for OP-1 in the mouse collagen X promoter. Mutational and gel shift analyses suggest that OP-1 induces nuclear protein interaction with an A/T-rich MEF-2 like sequence, distinct from the MEF-2 present in myoblasts, and up-regulates collagen X promoter activity, which also requires an AP-1 like sequence.


Subject(s)
Bone Morphogenetic Proteins/physiology , Collagen/genetics , DNA-Binding Proteins/genetics , Promoter Regions, Genetic , Transcription Factor AP-1/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/physiology , Activins , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Cell Line , Collagen/drug effects , DNA-Binding Proteins/physiology , Fetus , Inhibins/pharmacology , MEF2 Transcription Factors , Mice , Myogenic Regulatory Factors , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Promoter Regions, Genetic/drug effects , Protein Binding/genetics , Rats , Skull/cytology , Transcription Factors/physiology , Transfection
17.
Endocrinology ; 138(10): 4240-7, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9322936

ABSTRACT

To begin delineating molecular mechanisms by which osteogenic protein-1 (OP-1) modulates its effect on the insulin-like growth factor (IGF) system in human skeletal cells, we evaluated time-course effects of OP-1 on the expression of IGFBP-3 messenger RNA (mRNA) in human SaOS-2 osteosarcoma cells and found that 100 ng/ml of OP-1 increased (maximum 10.7-fold at 24 h; P < 0.01) the level of IGFBP-3 mRNA in a time-dependent manner (from 3-36 h; treatment x time interaction, P < 0.001). The stimulatory effect of OP-1 on IGFBP-3 mRNA was not promoted by transcript stabilization; actually, OP-1 treatment selectively increased the decay of mRNA for IGFBP-3 (T1/2 = 5 h vs. 24 h for OP-1 and controls), but not for IGFBP-4 or beta-actin. Conversely, OP-1 acutely increased IGFBP-3 nuclear transcript abundance in total RNA samples ranging between 1-24 h of treatment. After 6 h of treatment, OP-1 produced an average 4-fold increase (P < 0.02; n = 4 experiments) in the level of IGFBP-3 nuclear transcripts vs. a 3-fold increase (P < 0.01; n = 2 experiments) in mRNA abundance. The OP-1 stimulated induction of IGFBP-3 nuclear transcript and mRNA expression was dependent on de novo protein synthesis. Transient transfection experiments were undertaken to isolate putative OP-1 stimulatory cis-elements within 1.8-kb of the IGFBP-3 5'-flanking region in SaOS-2 and TE-85 osteosarcoma cells. In these experiments, OP-1 did not stimulate IGFBP-3 proximal promoter activity in either cell line, thus suggesting that OP-1 reactive domains may be located either beyond the currently established 5'-flanking region, or within internal exon/intron regions of the IGFBP-3 gene. In conclusion, OP-1 treatment stimulates IGFBP-3 expression in human osteoblastic cells by a mechanism that largely promotes the production of IGFBP-3 nuclear transcripts, a process that requires de novo protein synthesis, and overrides an OP-1-induced targeted degradation of IGFBP-3 steady-state mRNA.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Bone Neoplasms/chemistry , DNA, Neoplasm/analysis , Insulin-Like Growth Factor Binding Protein 3/genetics , Osteosarcoma/chemistry , Transcription, Genetic , Transforming Growth Factor beta , Actins/analysis , Actins/genetics , Base Sequence , Blotting, Northern , Bone Morphogenetic Protein 7 , Bone Neoplasms/pathology , DNA Primers/analysis , DNA Primers/chemistry , DNA Primers/genetics , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Insulin-Like Growth Factor Binding Protein 3/analysis , Insulin-Like Growth Factor Binding Protein 4/analysis , Insulin-Like Growth Factor Binding Protein 4/genetics , Introns , Osteosarcoma/pathology , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Time Factors , Tumor Cells, Cultured
18.
J Bone Joint Surg Am ; 79(8): 1132-7, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9278071

ABSTRACT

UNLABELLED: Osteogenic protein-1 (also known as bone morphogenetic protein-7) is a member of the bone morphogenetic protein family. Bone morphogenetic proteins and related members of the TGF-beta (transforming growth factor-beta) superfamily are involved in the development and repair of bone. Recombinant bone morphogenetic proteins induce the formation of new cartilage and bone at heterotopic sites. We investigated the influence of recombinant osteogenic protein-1 (at doses of three, ten, thirty, or 100 nanograms per milliliter) on the synthesis and release of proteoglycans and the maintenance of a steady-state concentration of proteoglycans in explants of porcine articular cartilage that were maintained in chemically defined serum-free medium. We found a dose-dependent stimulation of proteoglycan synthesis and a concurrent decrease in the rate of release of proteoglycans from the explants. The size of the proteoglycan monomers and the composition of the glycosaminoglycan chains in the untreated articular cartilage were similar to those in the articular cartilage treated with osteogenic protein-1. The capacity of the newly synthesized proteoglycan monomers to form aggregates with exogenous hyaluronic acid was found to be similar to that of proteoglycans in bovine nasal cartilage. Our results demonstrated that osteogenic protein-1 stimulated the synthesis of proteoglycans and diminished the release of proteoglycans from explants of porcine articular cartilage. CLINICAL RELEVANCE: The maintenance and repair of articular cartilage is a formidable challenge in clinical orthopaedics. The stimulation of proteoglycan synthesis by osteogenic protein-1 (bone morphogenetic protein-7) in explants of cartilage maintained in chemically defined serum-free medium implies that recombinant osteogenic protein-1 may play a role in the maintenance of a steady-state concentration of proteoglycans in articular cartilage, a desirable prerequisite for optimum repair of cartilage. Osteogenic protein-1 can initiate the formation of cartilage from mesenchymal cells. Once new cartilage has formed at the site of repair, osteogenic protein-1 also may maintain the synthesis of proteoglycans.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Cartilage, Articular/metabolism , Proteoglycans/biosynthesis , Transforming Growth Factor beta/pharmacology , Animals , Bone Morphogenetic Protein 7 , Cartilage, Articular/drug effects , Glycosaminoglycans/analysis , Male , Recombinant Proteins , Swine
19.
Bone ; 21(1): 23-30, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9213004

ABSTRACT

To clarify the pathogenesis of ossification of the ligamentum flavum (OLF), we examined the expression and localization of bone morphogenetic proteins (BMPs) and their receptors (BMPRs) in the ligamentum flavum of the patients with OLF by immunohistochemical staining and compared them with staining patterns in control patients. The BMPRs appeared extensively in mature and immature chondrocytes around the calcified zone and in spindle-shaped cells and round cells in the remote part from ossified foci in examined tissue of OLF. The ligands for BMPRs, BMP-2/-4 and osteogenic protein-1 (OP-1)/BMP-7, colocalized in OLF patients. In the control cases, expression of BMPs and BMPRs was observed around the calcified zone at the insertion of the ligamentum flavum to the bone, and limited expression was found in the smaller range. Thus, the expression profile of BMPs and BMPRs in OLF patients was entirely different from the control patients, suggesting that BMPs may be involved in promoting endochondral ossification at ectopic ossification sites in OLF, and that ossification activity is continuous in these patients.


Subject(s)
Bone Morphogenetic Proteins/biosynthesis , Calcinosis/genetics , Ligamentum Flavum/metabolism , Receptors, Cell Surface/metabolism , Activin Receptors , Adult , Aged , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Protein Receptors , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Calcinosis/metabolism , Cartilage/cytology , Cartilage/metabolism , Cell Size/physiology , Female , Humans , Immunohistochemistry , Ligamentum Flavum/pathology , Ligamentum Flavum/physiology , Male , Middle Aged , Protein Serine-Threonine Kinases/metabolism , Radioligand Assay , Receptors, Growth Factor/metabolism , Transforming Growth Factor beta/metabolism
20.
Cell ; 90(2): 257-69, 1997 Jul 25.
Article in English | MEDLINE | ID: mdl-9244300

ABSTRACT

Ventral midline cells at different rostrocaudal levels of the central nervous system exhibit distinct properties but share the ability to pattern the dorsoventral axis of the neural tube. We show here that ventral midline cells acquire distinct identities in response to the different signaling activities of underlying mesoderm. Signals from prechordal mesoderm control the differentiation of rostral diencephalic ventral midline cells, whereas notochord induces floor plate cells caudally. Sonic hedgehog (SHH) is expressed throughout axial mesoderm and is required for the induction of both rostral diencephalic ventral midline cells and floor plate. However, prechordal mesoderm also expresses BMP7 whose function is required coordinately with SHH to induce rostral diencephalic ventral midline cells. BMP7 acts directly on neural cells, modifying their response to SHH so that they differentiate into rostral diencephalic ventral midline cells rather than floor plate cells. Our results suggest a model whereby axial mesoderm both induces the differentiation of overlying neural cells and controls the rostrocaudal character of the ventral midline of the neural tube.


Subject(s)
Bone Morphogenetic Proteins/physiology , Diencephalon/embryology , Proteins/genetics , Trans-Activators , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/analysis , Cells, Cultured , Chick Embryo , Diencephalon/chemistry , Diencephalon/cytology , Ectoderm/chemistry , Ectoderm/cytology , Ectoderm/physiology , Embryonic Induction/physiology , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins , Humans , Immunohistochemistry , In Situ Hybridization , Mesoderm/chemistry , Mesoderm/cytology , Mesoderm/physiology , RNA, Messenger/analysis , Rats , Spinal Cord/chemistry , Spinal Cord/cytology , Spinal Cord/embryology , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...