Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Tissue Cell ; 42(2): 116-20, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20334886

ABSTRACT

The insertion site of the tendon to the skeletal element is hypovascular and is one of the most common sites of dysfunction in the musculoskeletal system. However, the resident cells have been poorly defined due to a lack of a specific marker for tenocytes. We previously reported that Tenomodulin (Tnmd) and Chondromodulin-1 (Chm1) are homologous angiogenesis inhibitors and predominantly expressed in the avascular region of tendons and cartilage, respectively. In this study, we analyzed the expression of Tnmd, Chm1, alpha 1 chain of the type I collagen (Col1a1) and alpha 1 chain of the type II collagen (Col2a1) at the insertion site of the Achilles, patellar, or rotator cuff tendons of 1-week-old rabbits by in situ hybridization analysis. Tnmd was co-expressed with Col1a1 in tenocytes of these tendons, while Chm1 and Col2a1 were detected in chondrocytes of the hyaline cartilage. Interestingly, the cell population between Tnmd/Col1a1 positive tenocytes and Chm1/Col2a1 positive chondrocytes expressed Col1a1 but none of the other markers (Tnmd, Chm1, and Col2a1). Red blood cells were exclusively present at the interface between the tendon substance and cartilage in the insertion site of the Achilles tendon. Lack of Tnmd and Chm1 in this newly characterized cell population may allow the transitional zone between the poorly vascularized tendon and cartilage to establish the unique vascular pattern for blood supply.


Subject(s)
Connective Tissue Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Tendons/metabolism , Animals , Animals, Newborn , Cartilage/cytology , Cartilage/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Cloning, Molecular , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Collagen Type II/genetics , Connective Tissue Cells/cytology , Erythrocytes/cytology , In Situ Hybridization , Phenotype , RNA, Messenger/analysis , RNA, Messenger/genetics , Rabbits , Tendons/blood supply , Tendons/cytology , Weight-Bearing/physiology
3.
Osteoarthritis Cartilage ; 9 Suppl A: S102-8, 2001.
Article in English | MEDLINE | ID: mdl-11680673

ABSTRACT

OBJECTIVE: Full-thickness defects that penetrate articular cartilage are filled by fibrous, or fibrocartilaginous tissue and, to a very limited extent, also by hyaline cartilage. In rabbits, small full-thickness defects (to < or =3 mm in diameter) are capable of regenerating surfacing hyaline cartilage. However, chondrogenic differentiation does not occur in larger defects (> or =5 mm in diameter). We studied the involvement of fibroblast growth factor-2 (FGF-2) in the cartilaginous repair response in full-thickness defects of articular cartilage in vivo, and attempted to facilitate cartilaginous repair of the defects by the local administration of FGF-2. DESIGN: The right knee joint of male adolescent Japanese white rabbits was entered through a medial parapatellan approach, and the patella was dislocated laterally to expose the articular surface of the femoral trochlea. Full-thickness defects were created in the weight-bearing area of the femoral trochlea with a hand-drill (the 5-mm diameter defects in 80 rabbits and the 3-mm diameter defects in 40 rabbits). The animals were fitted with an osmotic pump connected to silastic medical grade tubing, and a length of the tubing about 5 mm long was introduced into the articular knee cavity. The 5-mm-diameter defects received FGF-2 (50 pg/h) or sterile saline via an osmotic pump for the initial 2 weeks. Five animals each were sacrificed after 1, 2, 4, 8, or 24 weeks after creation of defects. The 3-mm diameter defects received a neutralizing monoclonal antibody against FGF-2 (50 ng/h) or pre-immune mouse IgG (50 ng/h) for the initial 2 weeks. Five animals each were sacrificed after 2, 3, or 4 weeks after creation of defects. The distal portion of each femur was removed, fixed, decalcified, and embedded in paraffin for the subsequent histological analysis. Sections were cut in the transverse plane, and histologically examined. RESULTS: The administration of FGF-2 (50 pg/h) resulted in successful regeneration of articular cartilage and the subchondral bone within 8 weeks after creation of 5-mm diameter defects. In these defects, undifferentiated mesenchymal cells initiated chondrogenic differentiation coupled with replacement by subchondral bone, resulting in the resurfacing of the defects by hyaline cartilage and the recovery of subchondral bone up to the original bone-articular cartilage junction. In contrast, the administration of a neutralizing monoclonal antibody against FGF-2 clearly interfered with the action of endogenous FGF-2 in 3-mm diameter defects, which were filled with fibrous tissue. None of the antibody-treated defects were covered with cartilage. We then assessed the proliferative capacity of the undifferentiated mesenchymal cells in the defects by immunostaining the proliferating cell nuclear antigen (PCNA) at 1 week after creation of defects. The capacity of reparative tissue to form cartilage was well correlated with the occurrence in the defects of a cell population that was PCNA-positive, undifferentiated, and capable of self-renewal. CONCLUSIONS: The local administration of FGF-2 resulted in the successful resurfacing of large (5 mm in diameter) defects by hyaline cartilage. Prechondrogenic mesenchymal cells were the likely targets of FGF-2, which probably promoted the formation of cartilage by stimulating a selective expansion of chondroprogenitor cells. Thus, activation of FGF-2 signalling is critically important for the induction of cartilaginous repair response in full-thickness articular cartilage.


Subject(s)
Cartilage, Articular/physiology , Cell Differentiation/physiology , Chondrocytes/cytology , Regeneration/physiology , Stem Cells/cytology , Animals , Antibodies, Monoclonal/physiology , Fibroblast Growth Factor 2/physiology , Hindlimb , Male , Mesoderm/cytology , Osteogenesis/physiology , Proliferating Cell Nuclear Antigen/physiology , Rabbits
4.
Osteoarthritis Cartilage ; 9 Suppl A: S91-101, 2001.
Article in English | MEDLINE | ID: mdl-11680695

ABSTRACT

OBJECTIVE: Cartilage is a typical avasclar tissue that exhibits powerful resistance to angiogenesis or vascular invasion. We previously identified a cartilage-specific 25 kDa glycosylated protein, chondromodulin-I (ChM-I), as anti-angiogenic factor. Taking advantage of ectopic bone formation and xenograft tumour model by human chondrosarcoma cell line OUMS-27, we examined how ChM-I is involved in switching of angiogenesis in cartilage. DESIGN: Gene expression pattern of ChM-I was examined in 4-week-old mice and mouse embryos by northern blot analysis and in situ hybridization. To evaluate the effect of ChM-I on ectopic bone formation, guanidine extracts of demineralized bone matrix were mixed with the ChM-I-bound heparin-Sepharose beads and were implanted onto the fasciae of back muscle of 6-week old nude mice. To analyse the effect of ChM-I on tumour angiogenesis, the level of ChM-I mRNA in cartilaginous tumours was assessed by competitive PCR, and compared with that of articular cartilage. Then, human chondrosarcoma OUMS-27 cells were inoculated into the back of nude mice to form a tumour about 45 mm3 in size. Recombinant ChM-I protein was administrated into OUMS-27 xenograft tumours for the initial 5 days to study its effect against tumour-angiogenesis. RESULTS: ChM-I gene was specifically expressed in cartilage of 4-week-old mice. Eye and thymus were also identified as minor expression sites. However, during endochondral bone development, cartilage changes its character from anti-angiogenic into angiogenic prior to the replacement of calcified cartilage by bone. In embryos, ChM-I mRNA was expressed in proliferative and upper hypertrophic cartilage zones in the developing cartilaginous bone rudiments, but completely abolished in lower hypertrophic and calcified cartilage zones. Purified ChM-I protein apparently inhibited vascular invasion into cartilage induced by the implantation of demineralized bone matrix in nude mice, leading to the inhibition of replacement of cartilage. The level of ChM-I transcripts in the lower-grade chondrosarcomas was substantially reduced to several hundreds or less in the lower-grade chondrosarcomas, compared with that of articular cartilage or other benign cartilage tumours. The local administration of recombinant human ChM-I almost completely blocked tumour angiogenesis and growth in the human chondrosarcoma xenografts in mice. CONCLUSIONS: ChM-I is involved in the anti-angiogenic property of cartilage and its absence creates a permissive microenvironment for vascular invasion into cartilage under physiological and pathological conditions.


Subject(s)
Cartilage/physiology , Growth Substances/physiology , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Neovascularization, Physiologic/physiology , Osteogenesis/physiology , Animals , Blotting, Northern/methods , Bone Transplantation , Cricetinae , Gene Expression , In Situ Hybridization/methods , Mice , Mice, Nude , Polymerase Chain Reaction/methods , Transplantation, Heterologous , Tumor Cells, Cultured/transplantation
5.
Mech Dev ; 105(1-2): 157-62, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11429291

ABSTRACT

Chondromodulin-I (ChM-I) is suggested in higher vertebrate systems to function as a key regulatory protein for cartilage development. To further understand the process of chondrogenesis and the function of ChM-I, we have cloned the zebrafish cDNA for chondromodulin-1 (chm1) and have mapped the chm1 gene locus. The expression profile of chm1 was determined during zebrafish embryonic development and compared to that of type II collagen (col2a1). Maternal chm1 transcripts were detected before midblastula transition and zygotic expression of chm1 was first observed in the notochord at the 10-somite stage. At later developmental stages, chm1 expression was detected in areas surrounding the otic vesicles, in the developing craniofacial cartilage elements, and in the chondrogenic region of the pectoral fins.


Subject(s)
Cartilage/embryology , Growth Substances/biosynthesis , Growth Substances/genetics , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Notochord/metabolism , Amino Acid Sequence , Animals , Cattle , DNA, Complementary/metabolism , Humans , In Situ Hybridization , Molecular Sequence Data , Phylogeny , Radiation Hybrid Mapping , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Time Factors , Tissue Distribution , Zebrafish , Zygote/metabolism
6.
Invest Ophthalmol Vis Sci ; 42(6): 1193-200, 2001 May.
Article in English | MEDLINE | ID: mdl-11328727

ABSTRACT

PURPOSE: To determine the role in the eye of chondromodulin (ChM)-I, which has been identified in cartilage as an angiogenic inhibitor, the expression and localization and a possible function of ChM-I were investigated. METHODS: Expression and localization of ChM-I in rat eyes were examined by RNase protection assay and in situ hybridization and by immunostaining, using an antibody against a synthetic peptide. The effect of recombinant ChM-I on tube morphogenesis of retinal endothelial cells was examined in culture. RESULTS: The rat ChM-I gene was determined to encode the open reading frame of 334 amino acid residues, and ChM-I mRNA was exclusively expressed in cartilage, eye, and cerebellum in rats. ChM-I mRNA expression was evident in the iris-ciliary body, retina, and scleral compartments, but not in other compartments of the eye. In situ hybridization revealed mRNA expression in the ganglion cells, inner nuclear layer cells, and pigment epithelium in the retina and in the nonpigment epithelium of the ciliary body. Immunoreactive ChM-I was present in these cells and also in the vitreous body. Western blot analysis detected an approximately 25-kDa band of ChM-I presumed as a secretory form in the aqueous humor and vitreous body and an approximately 37-kDa band as a precursor form in the retina. Recombinant human ChM-I inhibited tube morphogenesis of human retinal endothelial cells in vitro. CONCLUSIONS: These observations indicate a potential role for ChM-I in inhibition of angiogenesis in the rat eye.


Subject(s)
Eye Proteins/genetics , Eye/metabolism , Growth Substances/genetics , Intercellular Signaling Peptides and Proteins , Membrane Proteins , RNA, Messenger/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Cell Differentiation/drug effects , Ciliary Body/metabolism , Endothelium, Vascular/drug effects , Epithelial Cells/metabolism , Eye Proteins/metabolism , Eye Proteins/pharmacology , Growth Substances/metabolism , Growth Substances/pharmacology , In Situ Hybridization , Microscopy, Fluorescence , Molecular Sequence Data , Nuclease Protection Assays , Pigment Epithelium of Eye/metabolism , Rats , Rats, Inbred WKY , Retina/metabolism , Retinal Ganglion Cells/metabolism , Sclera/metabolism
7.
Am J Pathol ; 158(4): 1465-72, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11290564

ABSTRACT

Chondromodulin-I (ChM-I) is a novel cartilage-specific matrix protein. In the growth plates of the long bones, ChM-I was shown to be expressed in mature to upper hypertrophic chondrocytes, and to be deposited in the cartilage matrix. As ChM-I strongly inhibits angiogenesis, cartilage is avascular. Also, ChM-I has bifunctional activity against chondrocyte proliferation. On the other hand, pleomorphic adenomas of the salivary glands frequently have chondroid elements. To elucidate the relationship between chondroid formation and hypovascularity in salivary pleomorphic adenomas, we immunohistochemically examined the expression and localization of ChM-I in 35 cases of this tumor. ChM-I was immunolocalized to the lacunae in the chondroid elements of pleomorphic adenomas (100%). Type II collagen and aggrecan were immunolocalized throughout the matrix around lacuna cells of the chondroid element (100%, 91.7%), and ChM-I was infrequently immunolocalized to the spindle-shaped myoepithelial cells in the myxoid element (37.5%). Fibroblast growth factor-2 was strongly immunolocalized to the lacuna cells in the chondroid element (100%), among the neoplastic myoepithelial cells in the myxoid elements (96.9%), and on the basement membranes around the solid nests of neoplastic myoepithelial cells (71.4%). Although CD34 is a marker of endothelial cells, CD34 was expressed in the endothelial cells in only a few areas around the epithelial elements and in the fibrous element of pleomorphic adenomas. No signals for CD34 were observed in chondroid elements in pleomorphic adenomas (P < 0.001), but a few signals were seen in the myxoid elements (P < 0.05). These findings suggested that lacuna cells and neoplastic myoepithelial cells expressed ChM-I, and that this molecule may play an important role in hypovascularity and chondroid differentiation in pleomorphic adenoma. In conclusion, pleomorphic adenoma expressed ChM-I, which is involved in hypovascularity and chondroid formation in this type of tumor.


Subject(s)
Adenoma, Pleomorphic/metabolism , Adenoma, Pleomorphic/pathology , Chondrocytes/pathology , Extracellular Matrix Proteins , Growth Substances/metabolism , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Salivary Gland Neoplasms/metabolism , Salivary Gland Neoplasms/pathology , Aggrecans , Antigens, CD34/metabolism , Cartilage/metabolism , Collagen/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Immunohistochemistry , Lectins, C-Type , Proteoglycans/metabolism
8.
Biochem Biophys Res Commun ; 280(5): 1323-7, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11162673

ABSTRACT

Murine expressed sequence tags (EST) showing homology with chondromodulin-I (ChM-I) were identified. Cloning of the full-length cDNA revealed a novel protein (317 amino acid residues) having a domain homologous to ChM-I, and we termed it tenodmoulin (TeM). The predicted amino acid sequence revealed 33% overall identity with mouse ChM-I precursor. Overall structural features were conserved well in TeM, including a single transmembrane domain at the N-terminal region and the putative antiangiogenic domain with eight cysteine residues. However, TeM lacked a hormone-processing signal present in the ChM-I precursor, suggesting that it may function as a type II transmembrane protein on cell surface. TeM transcript (1.4 kb in size) was detected in skeletal muscle by Northern blot analysis. In situ hybridization analysis revealed that the expression of TeM mRNA was not associated with muscle fibers, but was tightly associated with epimysium and tendon, both of which are classified as dense connective tissue having little vascularity.


Subject(s)
Growth Substances/genetics , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Membrane Proteins , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cloning, Molecular , DNA, Complementary/chemistry , DNA, Complementary/genetics , Female , Gene Expression , Humans , In Situ Hybridization , Mice , Molecular Sequence Data , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution
9.
J Biol Chem ; 276(14): 11031-40, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11134040

ABSTRACT

To determine the role of fibroblast growth factor (FGF).FGF receptor (FGFR) signaling in chondrogenesis, we analyzed the gene expression of alternatively spliced FGFRs during chondrogenic differentiation of ATDC5 cells in vitro. Two isoforms of FGFR3 were expressed in these cells. One was the complete form of FGFR3 (FGFR3) already reported, and the other was a novel one that lacks the acid box domain (FGFR3DeltaAB). The gene of FGFR3DeltaAB was expressed in undifferentiated ATDC5 cells. In contrast, the transcripts of FGFR3 were not detectable in undifferentiated cells but increased during cellular condensation, which is an obligatory step for chondrogenic differentiation. FGFR1 and FGFR2 expression was higher than that of FGFR3 in undifferentiated cells. The gene expression of cell cycle inhibitor p21 was induced during cell condensation and correlated best with the expression of FGFR3 among the FGFR isoforms expressed. The differential expression of FGFR3 isoforms during chondrogenesis suggests that these isoforms may play different roles in the regulation of growth and differentiation in chondrocytes. To define the mitogenic response of FGFR3DeltaAB and FGFR3 to FGFs, their cDNAs were stably transfected into mouse BaF3 pro-B cells. FGFR3 preferentially mediates the mitogenic response to FGF1 and poor response to FGF2. In contrast, FGFR3DeltaAB mediated a higher mitogenic response to FGF2 as well as to FGF1. In addition, FGFR3DeltaAB responds to FGF1 at lower concentrations of heparin than FGFR3 does. These results suggest that the acid box plays an important role in the regulation of FGFR3 to mediate biological activities in response to FGFs.


Subject(s)
Chondrocytes/metabolism , Receptors, Fibroblast Growth Factor/biosynthesis , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Cell Line , Chondrocytes/cytology , DNA, Complementary/analysis , DNA, Complementary/genetics , Gene Expression Regulation , Humans , Molecular Sequence Data , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction
11.
Cell Struct Funct ; 25(3): 195-204, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10984103

ABSTRACT

Clonal cell line ATDC5 enables the monitoring of the early- and late-phase chondrogenic differentiation in a single culture. Undifferentiated ATDC5 cells differentiate into type II collagen expressing chondrocytes through a cellular condensation stage (early-phase differentiation) and then to type X collagen-expressing hypertrophic chondrocytes (late-phase differentiation). Progression of cellular differentiation was accelerated by the activation of bone morphogenetic protein (BMP) signaling. ATDC5 cells expressed transcripts for at least four members of the BMP family. The BMP-4 transcripts were expressed in all stages of differentiation, as were transcripts for BMP type IA receptor (ALK-3) and BMP type II receptor. In contrast, transcripts for Growth/ Differentiation factor-S (GDF-5) were induced during a cellular condensation, and those for BMP-6 were induced during the formation of cartilage nodules, and declined as the differentiated ATDC5 cells became hypertrophic, and BMP-7 transcripts were only detected after cells became calcified. Exogenously added BMP-4 indeed promoted the early-phase differentiation. Late-phase differentiation of cells was also stimulated by BMP-4 and BMP-6. Thus, the cumulative increase in BMP signaling promoted the sequential transitions of differentiation steps of cells. These results indicate that the coordinated expressions of endogenous BMPs are involved in the progression of chondrogenic differentiation in ATDC5 cells.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Chondrocytes/cytology , Chondrogenesis/physiology , Animals , Blotting, Northern , Bone Morphogenetic Proteins/genetics , Cell Differentiation/genetics , Cell Line , Chondrocytes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Mice , Osteogenesis/physiology , RNA/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
12.
Pediatr Nephrol ; 14(7): 602-5, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10912526

ABSTRACT

Cartilage is unique among mesenchymal tissues in that it is resistant to vascular invasion due to an intrinsic angiogenesis inhibitor. Chondromodulin-I (ChM-I), a 25-kilodalton glycoprotein purified from bovine epiphyseal cartilage on the basis of growth-promoting activity for chondrocytes, was recently identified as an angiogenesis inhibitor. Human ChM-I cDNA revealed that the mature protein consists of 120 amino acids and is coded as the C-terminal part of a larger transmembrane precursor. Expression of ChM-I cDNA in CHO cells indicated that mature ChM-I molecules were secreted from the cells after post-translational modifications and cleavage from the precursor protein at the predicted processing site. ChM-I stimulated growth and colony formation of cultured chondrocytes, but inhibited angiogenesis in vitro and in vivo. In situ hybridization and immunohistochemistry revealed that ChM-I is specifically expressed in the avascular zone of cartilage in developing bone, but not present in the late hypertrophic and calcified zones that allow vascular invasion. ChM-I actually inhibited vascular invasion into cartilage that was ectopically induced by demineralized bone matrix in nude mice, leading to the suppression of replacement of cartilage by bone in vivo. These results suggest that ChM-I participates in the angiogenic switching of cartilage, and that the withdrawal of its expression allows capillary in-growth, which triggers the replacement of cartilage by bone during endochondral bone development.


Subject(s)
Cartilage/physiology , Growth Substances/physiology , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Amino Acid Sequence/genetics , Animals , Chondrogenesis/physiology , Cloning, Molecular , Gene Expression , Growth Substances/genetics , Humans , Molecular Sequence Data
13.
Bone ; 27(1): 69-74, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10865211

ABSTRACT

In vivo expression of the type III sodium-dependent phosphate transporter (NaPiT) Glvr-1 during endochondral ossification, suggests a functional role for inorganic phosphate (Pi) transport in cartilage calcification. For further analysis of this relationship, an in vitro model of endochondral ossification is required. In this context, we investigated the characteristics of Pi transport in the new chondrogenic cell line ATDC5 in relation to extracellular matrix (ECM) formation and mineralization. Pi uptake in ATDC-5 cells and in isolated matrix vesicles (MVs) is mediated by an Na-dependent Pi transporter with a pH dependency characteristic of a type III Pi carrier (lower activity at alkaline pH). Northern blot analysis indicated that ATDC-5 cells express Glvr-1 transcripts during the various stages of their maturation with a maximal level during the proliferating stage. In isolated MVs, Pi transport activity was maximal at day 21, concomitant with the beginning of type X collagen messenger RNA expression. These events preceded the initiation of matrix mineralization, which was apparent at day 25, and then gradually increased until day 47. This temporal relationship between maximal Pi transport activity in MVs and the expression of a marker of mineralizing chondrocytes is compatible with the possible involvement of Pi transport in the ECM calcification observed in ATDC-5 cell cultures. In conclusion, these observations suggest that ATCD-5 cells in culture represent a promising model for the analysis of a functional role of Pi transport in the initial events of endochondral ossification.


Subject(s)
Calcification, Physiologic , Cartilage/physiology , Cell Culture Techniques , Phosphates/metabolism , Receptors, Virus/physiology , Biological Transport , Cartilage/cytology , Humans
14.
J Bone Miner Res ; 15(3): 421-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10750556

ABSTRACT

Chondromodulin-1 (ChM-1) is a cartilage-specific glycoprotein that stimulates the growth of chondrocytes and inhibits the tube formation of endothelial cells. To clarify the tissue-specific expression and the role of ChM-1 in pathophysiological conditions, we analyzed the structure of the human ChM-1 gene and its promoter. On the screening of a human genomic cosmid library using the human ChM-1 complimentary DNA (cDNA) as a probe, two clones were obtained that contained ChM-1 cDNA. The restriction enzyme map and nucleotide sequence revealed the human ChM-1 gene consisting of seven exons and exon-intron boundaries. The human ChM-1 gene was assigned to chromosome 13q14-21 by fluorescence in situ hybridization (FISH) using the clone as a probe. A primer extension analysis using total RNA extracted from human cartilage revealed a major transcription start site with the sequence CGCT+1GG. The region approximately 3-kilobase (kb) nucleotides upstream of the translation start site was then sequenced and analyzed in terms of promoter activity. We found that a region 446 base pairs (bp) upstream of the start site had promoter activity in COS7, HeLa, and ATDC5 cells. In structure the promoter is a TATA-less type without a GC-rich region. The transcription factors Sox9, Og12, and Cart-1 did not affect the promoter activity. The transcription factor Ying-Yang1 suppressed the promoter activity but GABP protein did not change the promoter activity. The construct containing -446/+87 fused to the SV40 enhancer and green fluorescent protein (GFP) exhibited expression of GFP corresponding to the differentiation of ATDC5 cells to mature chondrocytes. These results suggest that the element -446/+87 confers the cartilage-specific expression of this gene by some factor(s) other than Sox9, Og12, and Cart-1.


Subject(s)
Cartilage/metabolism , Growth Substances/genetics , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Promoter Regions, Genetic , Animals , Base Sequence , COS Cells , Cartilage/cytology , Cell Line , Chlorocebus aethiops , Chromosomes, Human, Pair 13/genetics , Cosmids , DNA, Complementary/genetics , Exons/genetics , Gene Expression Regulation , Gene Library , Genes, Reporter , Green Fluorescent Proteins , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Introns/genetics , Luciferases/biosynthesis , Luciferases/genetics , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Molecular Sequence Data , Organ Specificity , Polymorphism, Restriction Fragment Length , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transcription Factors/physiology , Transcription, Genetic , Transfection
15.
FEBS Lett ; 469(1): 83-7, 2000 Mar 03.
Article in English | MEDLINE | ID: mdl-10708761

ABSTRACT

Mouse EC cell line ATDC5 undergoes differentiation to form cartilage nodules via the cellular condensation stage in the presence of insulin. ATDC5 cells expressed transcripts for bone morphogenetic protein-4 (BMP-4), and type IA and type II BMP receptors. Moreover, cells retained responsiveness to BMP-4, which induced the formation of chondrocytes in the culture. When transfected with a kinase domain-truncated type IA BMP receptor construct, cells failed to undergo differentiation beyond the condensation stage even in the presence of insulin. The soluble form of type IA BMP receptor also blocked the formation of chondrocytes in a dose dependent manner. These lines of evidence suggested that autocrine BMP-4 signaling is required for the conversion of chondrogenic precursor cells into chondrocytes.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cartilage/growth & development , Signal Transduction , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Protein Receptors, Type II , Cell Differentiation , Cell Line , Chondrocytes/drug effects , Humans , Insulin/pharmacology , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Transfection
16.
FEBS Lett ; 458(3): 436-40, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10570955

ABSTRACT

Chondromodulin-I (ChM-I) was previously identified as an angiogenesis inhibitor in cartilage. Here, we demonstrated that the level of ChM-I transcripts was substantially reduced to 100 or even less in the lower-grade chondrosarcomas, in articular cartilage or other benign cartilage tumors. We implanted human chondrosarcoma OUMS-27 cells into nude mice that reproducibly produced tumors with cartilaginous matrix. Tumor-induced angiogenesis was evident when the tumors were excised 30 days after implantation. However, the local administration of recombinant human ChM-I almost completely blocked vascular invasion and tumor growth in vivo. Moreover, ChM-I also inhibited the growth of HT-29 colon adenocarcinoma in vivo, implying its therapeutic potential for solid tumors.


Subject(s)
Bone Neoplasms/genetics , Chondrosarcoma/genetics , Growth Substances/pharmacology , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Neovascularization, Pathologic/pathology , Animals , Antineoplastic Agents/pharmacology , Bone Neoplasms/blood supply , Bone Neoplasms/pathology , Cartilage/pathology , Chondrosarcoma/blood supply , Chondrosarcoma/pathology , Gene Expression Regulation, Neoplastic , Growth Substances/analysis , HT29 Cells , Humans , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
17.
FEBS Lett ; 456(1): 165-70, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10452551

ABSTRACT

Chondromodulin-I (ChM-I) is an angiogenesis inhibitor isolated from fetal bovine cartilage. Here, we report the nucleotide sequence of chicken ChM-I cDNA. Chicken mature ChM-I had a significantly larger N-terminal hydrophilic domain than its mammalian counterparts. Chicken embryos expressed multiple transcripts (3.3, 2.0 and 1.7 kb in size) due to the alternative utilization of polyadenylation signals, whereas only the 1.7 kb transcripts were detected in mammals. Although confined to cartilage and eye at a later stage of development, whole-mount in situ hybridization revealed the expression of ChM-I mRNA in somites, heart, bronchial arches, roof plate, retina and limb buds. The expression pattern of the gene suggests a role for ChM-I in the morphogenesis during embryonic development.


Subject(s)
Gene Expression Regulation, Developmental , Growth Substances/genetics , Intercellular Signaling Peptides and Proteins , 3' Untranslated Regions , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cartilage/embryology , Chick Embryo , Cloning, Molecular , Eye/embryology , Growth Substances/metabolism , Heart/embryology , In Situ Hybridization , Limb Buds/metabolism , Molecular Sequence Data , Sequence Homology, Amino Acid , Transcription, Genetic
18.
Int J Dev Biol ; 43(1): 39-49, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10213081

ABSTRACT

During endochondral bone formation, vascular invasion into cartilage initiates the replacement of cartilage by bone. Chondromodulin-I, a 25 kDa glycoprotein purified from bovine epiphyseal cartilage, was recently identified as a novel endothelial cell growth inhibitor. Here we cloned the mouse chondromodulin-I cDNA from a mouse whole embryo cDNA library. Northern blot analysis revealed that the chondromodulin-I transcripts were expressed in association with the formation of cartilage expressing type II collagen from days 11 to 17 of gestation in mouse embryos, at which time cartilaginous bone rudiments were gradually replaced by bone. Chondromodulin-I mRNA was also detected in the thymus and eyes at a lower level. In situ hybridization revealed significant expression in all cartilaginous tissues in the embryos at days 13.5 and 16 of gestation. However, the expression was completely abolished in the hypertrophic cartilage zone prior to calcification. Upon chondrogenic differentiation of mouse ATDC5 cells in vitro, the expression of chondromodulin-I transcripts was induced concomitantly with the formation of type II collagen-expressing chondrocytes. The expression of the transcripts then declined as type X collagen-expressing hypertrophic chondrocytes appeared in the culture. Purified chondromodulin-I protein inhibited the vascular invasion into cartilage ectopically induced by demineralized bone matrix in nude mice, leading to the suppression of bone formation in vivo. These results suggest that chondromodulin-I is involved in the anti-angiogenic property of cartilage, and that the withdrawal of its expression allows the vascular invasion which triggers the replacement of cartilage by bone during endochondral bone development.


Subject(s)
Bone and Bones/embryology , Cartilage/blood supply , Growth Substances/genetics , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Transforming Growth Factor beta , Amino Acid Sequence , Animals , Blotting, Northern , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Cattle , Cell Line , Choristoma/metabolism , Cloning, Molecular , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/metabolism , In Situ Hybridization , Mice , Mice, Nude , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Time Factors , Tissue Distribution
19.
Eur J Biochem ; 260(3): 869-78, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10103018

ABSTRACT

Bovine chondromodulin-I (ChM-I) purified from fetal cartilage stimulated the matrix synthesis of chondrocytes, and inhibited the growth of vascular endothelial cells in vitro. The human counterpart of this bovine growth regulating factor has not been identified. We report here the cloning of human ChM-I precursor cDNA and its functional expression in Chinese hamster ovary (CHO) cells. We first identified a genomic DNA fragment which encoded the N-terminus of the ChM-I precursor, and then isolated human ChM-I cDNA from chondrosarcoma tissue by PCR. The deduced amino acid sequence revealed that mature human ChM-I consists of 120 amino acids. In total, 16 amino acid residues were substituted in the human sequence, compared to the bovine counterpart. Almost of all the substitutions were found in the N-terminal hydrophilic domain. In the C-terminal hydrophobic domain (from Phe42 to Val120), the amino acid sequence was identical except for Tyr90, indicating a functional significance of the domain. Northern blotting and in situ hybridization indicated a specific expression of ChM-I mRNA in cartilage. We also successfully determined the cartilage-specific localization of ChM-I protein, using a specific antibody against recombinant human ChM-I. Multiple transfection of the precursor cDNA into CHO cells enabled us to isolate the mature form of human ChM-I from the culture supernatant. Purified recombinant human ChM-I stimulated proteoglycan synthesis in cultured chondrocytes. In contrast, it inhibited the tube morphogenesis of cultured vascular endothelial cells in vitro and angiogenesis in chick chorioallantoic membrane in vivo.


Subject(s)
Growth Substances/genetics , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Amino Acid Sequence , Animals , Base Sequence , Bone and Bones/metabolism , CHO Cells , Chondrocytes/metabolism , Cloning, Molecular , Cricetinae , DNA, Complementary/analysis , Embryo, Mammalian/metabolism , Endothelium/metabolism , Growth Substances/biosynthesis , Humans , Molecular Sequence Data , Protein Biosynthesis , Proteins/genetics , RNA Precursors/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Sequence Homology, Amino Acid
20.
J Biochem ; 125(3): 436-42, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10050029

ABSTRACT

We previously determined the complete primary sequence of a heparin-binding growth-promoting factor, chondromodulin-II (ChM-II), which stimulated the growth of chondrocytes and osteoblasts in culture. Bovine ChM-II was a 16-kDa basic protein with 133 amino acid residues and exhibited a significant sequence similarity to the repeats of the chicken mim-1 gene product. Here we report the nucleotide sequences of bovine and mouse ChM-II cDNAs. The cDNAs each contained an open-reading frame corresponding to the ChM-II precursor with 151 amino acid residues. The N-terminus of the precursor included a secretory signal sequence of 18 amino acids prior to the mature ChM-II sequence. Unlike MIM-1, there was no repeat structure in the precursor protein, indicating that ChM-II was encoded as a gene product distinct from MIM-1. We then expressed recombinant bovine ChM-II protein which was purified to homogeneity. The recombinant protein stimulated the growth of rabbit growth plate chondrocytes, mouse MC3T3-E1 cells and rat UMR-106 osteoblastic cells in vitro.


Subject(s)
Chondrocytes/drug effects , Growth Substances/genetics , Intercellular Signaling Peptides and Proteins , Osteoblasts/drug effects , Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cattle , Cell Division/drug effects , Cell Line , Chondrocytes/cytology , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Growth Substances/pharmacology , Mice , Molecular Sequence Data , Osteoblasts/cytology , Protein Precursors/genetics , Proteins/pharmacology , Rabbits , Rats , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...