Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Front Cell Dev Biol ; 12: 1360041, 2024.
Article in English | MEDLINE | ID: mdl-38895158

ABSTRACT

Fibrocartilaginous entheses consist of tendons, unmineralized and mineralized fibrocartilage, and subchondral bone, each exhibiting varying stiffness. Here we examined the functional role of sclerostin, expressed in mature mineralized fibrochondrocytes. Following rapid mineralization of unmineralized fibrocartilage and concurrent replacement of epiphyseal hyaline cartilage by bone, unmineralized fibrocartilage reexpanded after a decline in alkaline phosphatase activity at the mineralization front. Sclerostin was co-expressed with osteocalcin at the base of mineralized fibrocartilage adjacent to subchondral bone. In Scx-deficient mice with less mechanical loading due to defects of the Achilles tendon, sclerostin+ fibrochondrocyte count significantly decreased in the defective enthesis where chondrocyte maturation was markedly impaired in both fibrocartilage and hyaline cartilage. Loss of the Sost gene, encoding sclerostin, elevated mineral density in mineralized zones of fibrocartilaginous entheses. Atomic force microscopy analysis revealed increased fibrocartilage stiffness. These lines of evidence suggest that sclerostin in mature mineralized fibrochondrocytes acts as a modulator for mechanical tissue integrity of fibrocartilaginous entheses.

2.
PLoS One ; 18(2): e0280634, 2023.
Article in English | MEDLINE | ID: mdl-36795722

ABSTRACT

Chondromodulin (Cnmd) is a glycoprotein known to stimulate chondrocyte growth. We examined in this study the expression and functional role of Cnmd during distraction osteogenesis that is modulated by mechanical forces. The right tibiae of the mice were separated by osteotomy and subjected to slow progressive distraction using an external fixator. In situ hybridization and immunohistochemical analyses of the lengthened segment revealed that Cnmd mRNA and its protein in wild-type mice were localized in the cartilage callus, which was initially generated in the lag phase and was lengthened gradually during the distraction phase. In Cnmd null (Cnmd-/-) mice, less cartilage callus was observed, and the distraction gap was filled by fibrous tissues. Additionally, radiological and histological investigations demonstrated delayed bone consolidation and remodeling of the lengthened segment in Cnmd-/- mice. Eventually, Cnmd deficiency caused a one-week delay in the peak expression of VEGF, MMP2, and MMP9 genes and the subsequent angiogenesis and osteoclastogenesis. We conclude that Cnmd is necessary for cartilage callus distraction.


Subject(s)
Bony Callus , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Osteogenesis, Distraction , Animals , Mice , Cartilage , External Fixators , Osteogenesis/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics
3.
Bone ; 149: 115969, 2021 08.
Article in English | MEDLINE | ID: mdl-33892176

ABSTRACT

During tooth movement in orthodontic treatment, bone formation and resorption occur on the tension and compression sides of the alveolar bone, respectively. Although the bone formation activity increases in the periodontal ligament (PDL) on the tension side, the PDL itself is not ossified and maintains its homeostasis, indicating that there are negative regulators of bone formation in the PDL. Our previous report suggested that scleraxis (Scx) has an inhibitory effect on ossification of the PDL on the tension side through the suppression of calcified extracellular matrix formation. However, the molecular biological mechanisms of Scx-modulated inhibition of ossification in the tensioned PDL are not fully understood. The aim of the present study is to clarify the inhibitory role of Scx in osteoblast differentiation of PDL cells and its underlying mechanism. Our in vivo experiment using a mouse experimental tooth movement model showed that Scx expression was increased during early response of the PDL to tensile force. Scx knockdown upregulated expression of alkaline phosphatase, an early osteoblast differentiation marker, in the tensile force-loaded PDL cells in vitro. Transforming growth factor (TGF)-ß1-Smad3 signaling in the PDL was activated by tensile force and inhibitors of TGF-ß receptor and Smad3 suppressed the tensile force-induced Scx expression in PDL cells. Tensile force induced ephrin A2 (Efna2) expression in the PDL and Efna2 knockdown upregulated alkaline phosphatase expression in PDL cells under tensile force loading. Scx knockdown eliminated the tensile force-induced Efna2 expression in PDL cells. These findings suggest that the TGF-ß1-Scx-Efna2 axis is a novel molecular mechanism that negatively regulates the tensile force-induced osteoblast differentiation of PDL cells.


Subject(s)
Ephrin-A2 , Transforming Growth Factor beta1 , Cell Differentiation , Cells, Cultured , Ligaments , Osteoblasts , Osteogenesis , Periodontal Ligament , Tooth Movement Techniques
4.
PLoS One ; 15(12): e0242286, 2020.
Article in English | MEDLINE | ID: mdl-33259516

ABSTRACT

A multipotent cell population co-expressing a basic-helix-loop-helix transcription factor scleraxis (Scx) and SRY-box 9 (Sox9) has been shown to contribute to the establishment of entheses (tendon attachment sites) during mouse embryonic development. The present study aimed to investigate the involvement of Scx+/Sox9+ cells in the postnatal formation of fibrocartilaginous entheses and in the healing process after injury, using ScxGFP transgenic mice. We demonstrate that Scx+/Sox9+ cells are localized in layers at the insertion site during the postnatal formation of fibrocartilaginous entheses of supraspinatus tendon until postnatal 3 weeks. Further, these cells were rarely seen at postnatal 6 weeks, when mature fibrocartilaginous entheses were formed. Furthermore, we investigated the involvement of Scx+/Sox9+ cells in the healing process after supraspinatus tendon enthesis injury, comparing the responses of 20- and 3-week-old mice. In the healing process of 20-week-old mice with disorganized fibrovascular tissue in response to injury, a small number of Scx+/Sox9+ cells transiently appeared from 1 week after injury, but they were rarely seen at 4 weeks after injury. Meanwhile, in 3-week-old mice, a thin layer of fibrocartilaginous tissue with calcification was formed at healing enthesis at 4 weeks after injury. From 1 to 2 weeks after injury, more Scx+/Sox9+ cells, widely distributed at the injured site, were seen compared with the 20-week-old mice. At 4 weeks after injury, these cells were located near the surface of the recreated fibrocartilaginous layer. This spatiotemporal localization pattern of Scx+/Sox9+ cells at the injured enthesis in our 3-week-old mouse model was similar to that in postnatal fibrocartilaginous enthesis formation. These findings indicate that Scx+/Sox9+ cells may have a role as entheseal progenitor-like cells during postnatal maturation of fibrocartilaginous entheses and healing after injury in a manner similar to that seen in embryonic development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , SOX9 Transcription Factor/genetics , Tendon Injuries/therapy , Wound Healing/genetics , Animals , Cell Lineage/genetics , Disease Models, Animal , Fibrocartilage/growth & development , Fibrocartilage/injuries , Fibrocartilage/metabolism , Humans , Mice , Mice, Transgenic , Musculoskeletal System/pathology , Postnatal Care , Rotator Cuff/growth & development , Rotator Cuff/pathology , Stem Cells/metabolism , Tendon Injuries/genetics , Tendon Injuries/pathology , Tendons/growth & development , Tendons/metabolism , Tendons/pathology
5.
Sci Rep ; 9(1): 4605, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872687

ABSTRACT

A previously identified enhancer 10 kb upstream of the Aggrecan (Acan) gene (UE) can drive cartilage specific reporter expression in vivo. Here, we report that the paralogous transcription factors PAX1 and PAX9 differentially drive UE, depending on the presence or absence of SOX9-driven transactivation. In the developing vertebral column, PAX1/9 expression was inversely correlated with Acan expression. Moreover, PAX1/9 was co-expressed with SOX9/5/6 in the intervertebral mesenchyme and the inner annulus fibrosus (AF), and with SOX9 in the outer AF. Significant Acan upregulation was observed during chondrification of Pax1-silenced AF cells, while, Acan was significantly downregulated by persistent expression of Pax1 in cartilage. Deletion of UE using CRISPR/Cas9 resulted in ~30% and ~40% reduction of Acan expression in cartilage and the AF, respectively. In the UE, PAX1/9 acts as weak transactivators through a PAX1/9-binding site partially overlapped with a SOX9-binding site. In the presence of SOX9, which otherwise drives robust Acan expression along with SOX5/6, PAX1/9 competes with SOX9 for occupancy of the binding site, resulting in reduced transactivation of Acan. Coimmunoprecipitation revealed the physical interaction of Pax1 with SOX9. Thus, transactivation of the UE is differentially regulated by concerted action of PAX1/9, SOX9, and SOX5/6 in a context-dependent manner.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Paired Box Transcription Factors/metabolism , SOX9 Transcription Factor/metabolism , Transcriptional Activation , Animals , Base Sequence , Biomarkers , Fluorescent Antibody Technique , Gene Silencing , Mice , Mice, Transgenic , Paired Box Transcription Factors/chemistry , Paired Box Transcription Factors/genetics , Phenotype
6.
Sci Rep ; 8(1): 3155, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29453333

ABSTRACT

Tenomodulin (Tnmd) is a type II transmembrane glycoprotein predominantly expressed in tendons and ligaments. We found that scleraxis (Scx), a member of the Twist-family of basic helix-loop-helix transcription factors, is a transcriptional activator of Tnmd expression in tenocytes. During embryonic development, Scx expression preceded that of Tnmd. Tnmd expression was nearly absent in tendons and ligaments of Scx-deficient mice generated by transcription activator-like effector nucleases-mediated gene disruption. Tnmd mRNA levels were dramatically decreased during serial passages of rat tenocytes. Scx silencing by small interfering RNA significantly suppressed endogenous Tnmd mRNA levels in tenocytes. Mouse Tnmd contains five E-box sites in the ~1-kb 5'-flanking region. A 174-base pair genomic fragment containing a TATA box drives transcription in tenocytes. Enhancer activity was increased in the upstream region (-1030 to -295) of Tnmd in tenocytes, but not in NIH3T3 and C3H10T1/2 cells. Preferential binding of both Scx and Twist1 as a heterodimer with E12 or E47 to CAGATG or CATCTG and transactivation of the 5'-flanking region were confirmed by electrophoresis mobility shift and dual luciferase assays, respectively. Scx directly transactivates Tnmd via these E-boxes to positively regulate tenocyte differentiation and maturation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Ligaments/cytology , Membrane Proteins/genetics , Tenocytes/metabolism , Transcriptional Activation , Amino Acid Sequence , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/chemistry , Biomarkers/metabolism , Mice , Protein Multimerization , Protein Structure, Quaternary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Initiation Site
7.
Sci Rep ; 7: 45010, 2017 03 22.
Article in English | MEDLINE | ID: mdl-28327634

ABSTRACT

Scleraxis (Scx) is a basic helix-loop-helix transcription factor that is expressed persistently in tendons/ligaments, but transiently in entheseal cartilage. In this study, we generated a novel ScxCre knock-in (KI) allele, by in-frame replacement of most of Scx exon 1 with Cre recombinase (Cre), to drive Cre expression using Scx promoter and to inactivate the endogenous Scx. Reflecting the intensity and duration of endogenous expression, Cre-mediated excision occurs in tendinous and ligamentous tissues persistently expressing Scx. Expression of tenomodulin, a marker of mature tenocytes and ligamentocytes, was almost absent in tendons and ligaments of ScxCre/Cre KI mice lacking Scx to indicate defective maturation. In homozygotes, the transiently Scx-expressing entheseal regions such as the rib cage, patella cartilage, and calcaneus were small and defective and cartilaginous tuberosity was missing. Decreased Sox9 expression and phosphorylation of Smad1/5 and Smad3 were also observed in the developing entheseal cartilage, patella, and deltoid tuberosity of ScxCre/Cre KI mice. These results highlighted the functional importance of both transient and persistent expression domains of Scx for proper integration of the musculoskeletal components.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Musculoskeletal Development/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/pathology , Cartilage/abnormalities , Cartilage/metabolism , Gene Expression Regulation, Developmental , Gene Order , Gene Targeting , Genes, Reporter , Genetic Loci , Homologous Recombination , Ligaments/abnormalities , Ligaments/growth & development , Ligaments/metabolism , Mice , Mice, Knockout , Phosphorylation , SOX9 Transcription Factor/genetics , Smad Proteins/genetics , Smad Proteins/metabolism , Tendons/abnormalities , Tendons/growth & development , Tendons/metabolism
8.
PLoS Genet ; 12(1): e1005802, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26820155

ABSTRACT

Previously, we identified an adolescent idiopathic scoliosis susceptibility locus near human ladybird homeobox 1 (LBX1) and FLJ41350 by a genome-wide association study. Here, we characterized the associated non-coding variant and investigated the function of these genes. A chromosome conformation capture assay revealed that the genome region with the most significantly associated single nucleotide polymorphism (rs11190870) physically interacted with the promoter region of LBX1-FLJ41350. The promoter in the direction of LBX1, combined with a 590-bp region including rs11190870, had higher transcriptional activity with the risk allele than that with the non-risk allele in HEK 293T cells. The ubiquitous overexpression of human LBX1 or either of the zebrafish lbx genes (lbx1a, lbx1b, and lbx2), but not FLJ41350, in zebrafish embryos caused body curvature followed by death prior to vertebral column formation. Such body axis deformation was not observed in transcription activator-like effector nucleases mediated knockout zebrafish of lbx1b or lbx2. Mosaic expression of lbx1b driven by the GATA2 minimal promoter and the lbx1b enhancer in zebrafish significantly alleviated the embryonic lethal phenotype to allow observation of the later onset of the spinal curvature with or without vertebral malformation. Deformation of the embryonic body axis by lbx1b overexpression was associated with defects in convergent extension, which is a component of the main axis-elongation machinery in gastrulating embryos. In embryos overexpressing lbx1b, wnt5b, a ligand of the non-canonical Wnt/planar cell polarity (PCP) pathway, was significantly downregulated. Injection of mRNA for wnt5b or RhoA, a key downstream effector of Wnt/PCP signaling, rescued the defective convergent extension phenotype and attenuated the lbx1b-induced curvature of the body axis. Thus, our study presents a novel pathological feature of LBX1 and its zebrafish homologs in body axis deformation at various stages of embryonic and subsequent growth in zebrafish.


Subject(s)
Homeodomain Proteins/genetics , Morphogenesis/genetics , Scoliosis/genetics , Transcription Factors/genetics , Wnt Proteins/genetics , Zebrafish Proteins/genetics , Adolescent , Animals , Cell Polarity/genetics , Gene Expression Regulation, Developmental , HEK293 Cells , Homeodomain Proteins/biosynthesis , Humans , Polymorphism, Single Nucleotide , Scoliosis/pathology , Transcription Factors/biosynthesis , Wnt Proteins/biosynthesis , Wnt Signaling Pathway/genetics , Wnt-5a Protein , Zebrafish , Zebrafish Proteins/biosynthesis
9.
Jpn Dent Sci Rev ; 52(4): 84-92, 2016 Nov.
Article in English | MEDLINE | ID: mdl-28408960

ABSTRACT

Tendons and ligaments are dense fibrous bands of connective tissue that integrate musculoskeletal components in vertebrates. Tendons connect skeletal muscles to the bone and function as mechanical force transmitters, whereas ligaments bind adjacent bones together to stabilize joints and restrict unwanted joint movement. Fibroblasts residing in tendons and ligaments are called tenocytes and ligamentocytes, respectively. Tenomodulin (Tnmd) is a type II transmembrane glycoprotein that is expressed at high levels in tenocytes and ligamentocytes, and is also present in periodontal ligament cells and tendon stem/progenitor cells. Tnmd is related to chondromodulin-1 (Chm1), a cartilage-derived angiogenesis inhibitor, and both Tnmd and Chm1 are expressed in the CD31- avascular mesenchyme. The conserved C-terminal hydrophobic domain of these proteins, which is characterized by the eight Cys residues to form four disulfide bonds, may have an anti-angiogenic function. This review highlights the molecular characterization and function of Tnmd, a specific marker of tendons and ligaments.

10.
Development ; 142(4): 787-96, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25670797

ABSTRACT

The periodontal ligament (PDL) is a mechanosensitive noncalcified fibrous tissue connecting the cementum of the tooth and the alveolar bone. Here, we report that scleraxis (Scx) and osterix (Osx) antagonistically regulate tensile force-responsive PDL fibrogenesis and osteogenesis. In the developing PDL, Scx was induced during tooth eruption and co-expressed with Osx. Scx was highly expressed in elongated fibroblastic cells aligned along collagen fibers, whereas Osx was highly expressed in the perialveolar/apical osteogenic cells. In an experimental model of tooth movement, Scx and Osx expression was significantly upregulated in parallel with the activation of bone morphogenetic protein (BMP) signaling on the tension side, in which bone formation compensates for the widened PDL space away from the bone under tensile force by tooth movement. Scx was strongly expressed in Scx(+)/Osx(+) and Scx(+)/Osx(-) fibroblastic cells of the PDL that does not calcify; however, Scx(-)/Osx(+) osteogenic cells were dominant in the perialveolar osteogenic region. Upon BMP6-driven osteoinduction, osteocalcin, a marker for bone formation was downregulated and upregulated by Scx overexpression and knockdown of endogenous Scx in PDL cells, respectively. In addition, mineralization by osteoinduction was significantly inhibited by Scx overexpression in PDL cells without affecting Osx upregulation, suggesting that Scx counteracts the osteogenic activity regulated by Osx in the PDL. Thus, Scx(+)/Osx(-), Scx(+)/Osx(+) and Scx(-)/Osx(+) cell populations participate in the regulation of tensile force-induced remodeling of periodontal tissues in a position-specific manner.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Periodontal Ligament/metabolism , Tensile Strength/physiology , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA Interference , Rats , Rats, Wistar , Sp7 Transcription Factor , Transcription Factors/genetics
11.
PLoS One ; 9(4): e94239, 2014.
Article in English | MEDLINE | ID: mdl-24710035

ABSTRACT

Chondromodulin-I (ChM-I) is a 20-25 kDa anti-angiogenic glycoprotein in cartilage matrix. In the present study, we identified a novel 14-kDa species of ChM-I by immunoblotting, and purified it by immunoprecipitation with a newly raised monoclonal antibody against ChM-I. The N-terminal amino acid sequencing indicated that it was an N-terminal truncated form of ChM-I generated by the proteolytic cleavage at Asp37-Asp38. This 14-kDa ChM-I was shown by the modified Boyden chamber assay to have very little inhibitory activity on the VEGF-A-induced migration of vascular endothelial cells in contrast to the intact 20-25 kDa form of ChM-I (ID50 = 8 nM). Immunohistochemistry suggested that 20-25 kDa ChM-I was exclusively localized in the avascular zones, i.e. the resting, proliferating, and prehypertrophic zones, of the cartilaginous molds of developing long bone, whereas the 14-kDa form of ChM-I was found in hypertrophic and calcified zones. Immunoblotting demonstrated that mature growth-plate chondrocytes isolated from rat costal cartilage actively secrete ChM-I almost exclusively as the intact 20-25 kDa form into the medium in primary culture. Taken together, our results suggest that intact 20-25 kDa ChM-I is stored as a component of extracellular matrix in the avascular cartilage zones, but it is inactivated by a single N-terminal proteolytic cleavage in the hypertrophic zone of growth-plate cartilage.


Subject(s)
Bone Development , Calcification, Physiologic , Cartilage/metabolism , Growth Plate/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Proteolysis , Amino Acid Sequence , Animals , Chondrocytes/metabolism , Growth Plate/cytology , Growth Plate/pathology , Growth Plate/physiology , Humans , Hypertrophy , Male , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Transport , Rats
12.
Exp Cell Res ; 319(20): 3128-39, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24080012

ABSTRACT

Paired box gene 1 (Pax1) indirectly promotes the early stages of chondrogenic differentiation through induction and transactivation of Nk3 homeobox 2 (Nkx3.2), a transcriptional repressor. Later in chondrogenic differentiation, Nkx3.2 blocks chondrocyte hypertrophy by repressing Runt-related transcription factor 2 (Runx2). Here we report the inhibitory action of Pax1 on chondrocyte maturation, independently of Nkx3.2. Upon cartilage formation, Pax1 expression in the ventral sclerotome was gradually decreased except for the perichondrial region of the vertebral bodies and the intervertebral region, both of which express SRY-box containing gene 9 (Sox9). Forced expression of Pax1 in the chick forelimb resulted in the formation of shortened skeletal elements with a significant reduction of proteoglycans (PGs) accumulation in cartilage as well as a lack of the cortical bone formation and vascular invasion into the primary ossification center. Pax1-misexpressing chondrocytes exhibited aberrant cell morphology with a marked downregulation of Aggrecan (Agc1). Pax1-misexpressing cultured chondrocytes failed to accumulate cartilaginous PGs and became fibroblastic, in association with downregulation of the expression of Sox9, Nkx3.2, Indian hedgehog (Ihh), type II collagen (Col2a1), Chondromodulin-1 (Chm1), and Agc1. Accumulation of cartilaginous PGs in chondrocytes was also reduced by forced expression of Pax1 and Sox9. Thus, chondrocyte maturation driven by Sox9 is antagonized by Pax1 that is downregulated during chondrogenic differentiation.


Subject(s)
Cell Differentiation , Chondrocytes/cytology , Chondrocytes/metabolism , Paired Box Transcription Factors/metabolism , Animals , Cells, Cultured , Chickens , Down-Regulation
13.
Nat Genet ; 45(6): 676-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23666238

ABSTRACT

Adolescent idiopathic scoliosis (AIS) is the most common pediatric skeletal disease. We previously reported a locus on chromosome 10q24.31 associated with AIS susceptibility in Japanese using a genome-wide association study (GWAS) consisting of 1,033 cases and 1,473 controls. To identify additional AIS-associated loci, we expanded the study by adding X-chromosome SNPs in the GWAS and increasing the size of the replication cohorts. Through a stepwise association study including 1,819 cases and 25,939 controls, we identified a new susceptibility locus on chromosome 6q24.1 in Japanese (P = 2.25 × 10(-10); odds ratio (OR) = 1.28). The most significantly associated SNP, rs6570507, was in GPR126 (encoding G protein-coupled receptor 126). Its association was replicated in Han Chinese and European-ancestry populations (combined P = 1.27 × 10(-14); OR = 1.27). GPR126 was highly expressed in cartilage, and the knockdown of gpr126 in zebrafish caused delayed ossification of the developing spine. Our results should provide insights into the etiology and pathogenesis of AIS.


Subject(s)
Polymorphism, Single Nucleotide , Receptors, G-Protein-Coupled/genetics , Adolescent , Animals , Case-Control Studies , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Humans , Linkage Disequilibrium , Scoliosis
14.
Development ; 140(11): 2280-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23615282

ABSTRACT

SRY-box containing gene 9 (Sox9) and scleraxis (Scx) regulate cartilage and tendon formation, respectively. Here we report that murine Scx(+)/Sox9(+) progenitors differentiate into chondrocytes and tenocytes/ligamentocytes to form the junction between cartilage and tendon/ligament. Sox9 lineage tracing in the Scx(+) domain revealed that Scx(+) progenitors can be subdivided into two distinct populations with regard to their Sox9 expression history: Scx(+)/Sox9(+) and Scx(+)/Sox9(-) progenitors. Tenocytes are derived from Scx(+)/Sox9(+) and Scx(+)/Sox9(-) progenitors. The closer the tendon is to the cartilaginous primordium, the more tenocytes arise from Scx(+)/Sox9(+) progenitors. Ligamentocytes as well as the annulus fibrosus cells of the intervertebral discs are descendants of Scx(+)/Sox9(+) progenitors. Conditional inactivation of Sox9 in Scx(+)/Sox9(+) cells causes defective formation in the attachment sites of tendons/ligaments into the cartilage, and in the annulus fibrosus of the intervertebral discs. Thus, the Scx(+)/Sox9(+) progenitor pool is a unique multipotent cell population that gives rise to tenocytes, ligamentocytes and chondrocytes for the establishment of the chondro-tendinous/ligamentous junction.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cartilage/growth & development , Gene Expression Regulation, Developmental , Ligaments/growth & development , SOX9 Transcription Factor/genetics , Stem Cells/cytology , Tendons/growth & development , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone and Bones/physiology , Mesoderm/growth & development , Mice , Mice, Transgenic , SOX9 Transcription Factor/metabolism
15.
Genesis ; 51(4): 275-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23349075

ABSTRACT

Scleraxis (Scx) is a basic helix-loop-helix transcription factor that is a marker for the tendon/ligament cell lineage. The ∼11 kb genomic region from the mouse Scx gene locus faithfully recapitulates the endogenous Scx expression pattern in ScxGFP transgenic (Tg) mice. We have established two Tg mouse lines expressing Cre-recombinase (Cre) using this regulatory region (ScxCre-L and ScxCre-H). The specificity and efficiency of Cre recombination in these Tg lines are evaluated by crossing with Rosa-CAG-LSL-tdTomato (Ai14) or ROSA26R (R26R) reporter mice. The recombination in ScxCre-H;Ai14 mice is efficiently achieved in the endogenous Scx expression domains including the branchial arches, the syndetome, and the lateral plate mesoderm. Further analysis of ScxCre-H;Ai14;ScxGFP embryos reveal that expression of the ScxGFP transgene largely overlaps with Cre activity detected by tdTomato at embryonic day 12.5 (E12.5). In ScxCre-L;R26R or ScxCre-H;R26R neonates, Cre activity is detected in tendons, ligaments, intervertebral discs, joints, and cartilage around the chondro-tendinous/ligamentous junction, the prospective enthesis. The present results suggest that ScxCre Tg lines are useful for targeting the gene specifically in the Scx-expressing domains.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Integrases/genetics , Mice, Transgenic/genetics , Animals , Founder Effect , Gene Expression , Green Fluorescent Proteins/genetics , Mice , Musculoskeletal System/metabolism
16.
Exp Cell Res ; 318(13): 1492-507, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22510437

ABSTRACT

Sox9 is a high-mobility group box-containing transcription factor that functions as a key regulator of chondrogenesis. We here report that Sox9 mediates the direct conversion of tenocytes to chondrocytes through an intermediate state in which both differentiation programs are active. Sox9 is abundantly expressed in cartilage but is undetectable in limb tendons that express Scleraxis (Scx) and Tenomodulin (Tnmd), tendon-specific early and late molecular markers, respectively. Upon forced expression of Sox9 in the chick forelimb, ectopic cartilage formation is preferentially observed in fibrous tissues including the tendons, ligaments, perichondrium/periosteum, dermis, and muscle connective tissues. Tnmd expression in tenocytes isolated from leg tendons was markedly upregulated by forced expression of basic helix-loop-helix (b-HLH) activators including Scx, Paraxis, Twist1 and Twist2. In contrast, the overexpression of Sox9 in monolayer tenocytes resulted in the downregulation of Tnmd and Scx expressions during passaging in culture, and the induction of cartilage molecular markers such as type II collagen (Col2a1) and Chondromodulin-I (ChM-I). This Sox9-driven switching from a tenocytic to a chondrocytic gene expression profile was associated with a dramatic change from a spindle to a polygonal cellular morphology. The extracellular accumulation of cartilage-characteristic proteoglycans was also observed. These data suggest that tenocytes have a strong potential for conversion into chondrocytes through the activities of Sox9 both in vitro and in vivo.


Subject(s)
Avian Proteins/metabolism , Cell Transdifferentiation/physiology , Chondrocytes/cytology , Chondrocytes/metabolism , SOX9 Transcription Factor/metabolism , Tendons/cytology , Tendons/metabolism , Animals , Avian Proteins/genetics , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Transdifferentiation/genetics , Cells, Cultured , Chick Embryo , Chondrogenesis/genetics , Chondrogenesis/physiology , Choristoma/genetics , Choristoma/metabolism , Choristoma/pathology , DNA Primers/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Myoblasts/cytology , Myoblasts/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , SOX9 Transcription Factor/genetics
17.
J Bone Miner Metab ; 28(6): 659-71, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20458606

ABSTRACT

Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are bioactive lysophospholipids that affect various cellular processes through G protein-coupled receptors. In our current study, we found by in situ hybridization that E11.5 mouse embryos strongly expressed the LPA receptor subtype LPA(1) in cartilaginous bone primordia and the surrounding mesenchymal cells. However, despite their wide-ranging actions, the roles of lysophospholipids in chondrogenesis remain poorly understood. The mouse clonal cell line ATDC5 undergoes a sequential differentiation of chondroprogenitor cells in vitro. Undifferentiated and differentiated ATDC5 cells express LPA(1) and other lysophospholipid receptors including S1P receptor S1P(1) and S1P(2). Taking advantage of this cell model, we studied the effects of LPA on the activities of chondroprogenitor cells. LPA markedly stimulates both DNA synthesis and the migration of ATDC5 chondroprogenitor cells in culture, whereas S1P suppresses the migration of these cells. Treatment with Ki16425, an LPA(1)- and LPA(3)-specific receptor antagonist, suppressed the fetal bovine serum-stimulated migration of ATDC5 cells by almost 80%. These results indicate that LPA plays an important role in the activation of chondroprogenitor cells.


Subject(s)
Chondrocytes/metabolism , Chondrogenesis , Lysophospholipids/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Animals , Bone and Bones/cytology , Bone and Bones/embryology , Bone and Bones/metabolism , Cell Line , Cell Migration Inhibition/drug effects , Cell Movement/drug effects , Cell Proliferation , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrogenesis/drug effects , DNA/biosynthesis , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , In Situ Hybridization , Isoxazoles/pharmacology , MAP Kinase Signaling System , Mice , Mice, Inbred ICR , Propionates/pharmacology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors
18.
Dev Biol ; 332(2): 196-211, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19464280

ABSTRACT

During endochondral bone formation, vascular invasion initiates the replacement of avascular cartilage by bone. We demonstrate herein that the cartilage-specific overexpression of VEGF-A(164) in mice results in the hypervascularization of soft connective tissues away from cartilage. Unexpectedly, perichondrial tissue remained avascular in addition to cartilage. Hypervascularization of tissues similarly occurred when various VEGF-A isoforms were overexpressed in the chick forelimb, but also in this case perichondrial tissue and cartilage were completely devoid of vasculature. However, following bony collar formation, anti-angiogenic properties in perichondrial tissue were lost and perichondrial angiogenesis was accelerated by VEGF-A(146), VEGF-A(166), or VEGF-A(190). Once the perichondrium was vascularized, osteoclast precursors were recruited from the circulation and the induction of MMP9 and MMP13 can be observed in parallel with the activation of TGF-beta signaling. Neither perichondrial angiogenesis nor the subsequent cartilage vascularization was found to be accelerated by the non-heparin-binding VEGF-A(122) or by the VEGF-A(166)DeltaE(162)-R(166) mutant lacking a neuropilin-binding motif. Hence, perichondrial angiogenesis is a prerequisite for subsequent cartilage vascularization and is differentially regulated by VEGF-A isoforms.


Subject(s)
Neovascularization, Physiologic , Osteogenesis , Protein Isoforms/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Amino Acid Sequence , Animals , Bone and Bones/drug effects , Bone and Bones/physiology , Cartilage/drug effects , Cartilage/physiology , Chick Embryo , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/physiology , Collagen Type I/genetics , Collagen Type I/metabolism , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/physiology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Humans , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Osteoblasts/cytology , Osteoblasts/physiology , Osteogenesis/drug effects , Osteogenesis/physiology , Peptides/genetics , Peptides/metabolism , Protein Isoforms/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/genetics
19.
Bone ; 43(6): 1047-56, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18793763

ABSTRACT

Chondromodulin-I (Chm-I) is a glycoprotein that stimulates the growth of chondrocytes and inhibits angiogenesis in vitro. Mice lacking the Chm1 gene show abnormal bone metabolism and pathological angiogenesis in cardiac valves in the mature stage although they develop normally without aberrations in endochondral bone formation during embryogenesis or in cartilage development during growth. These findings indicate that Chm-I is critical under conditions of stress such as bone repair through endochondral ossification of a fracture callus. We carried out the present study to examine the expression and role of Chm-I in bone repair using a stabilized tibial fracture model, and compared fracture healing in Chm1 knockout (Chm1(-/-)) mice with that in wild-type mice. Chm-I mRNA and protein localized in the external cartilaginous callus in the reparative phase of fracture healing. Radiological examination showed a delayed union in Chm1(-/-) mice although the fracture site was covered with both external and internal calluses. Chm1 null mutation reduced external cartilaginous callus formation as judged by marked decrease of type X collagen alpha 1 (Col10a1) expression and the total amount of cartilage matrix. Interestingly, the majority of chondrocytes in the periosteal callus failed to differentiate into mature chondrocytes in Chm1(-/-) mice, while the hypertrophic maturation of chondrocytes between the cortices was not affected. These results suggest that Chm-I is involved in hypertrophic maturation of periosteal chondrocytes. Although a direct effect of Chm-I on bones is still unclear, bony callus formation was increased while external cartilaginous callus decreased in Chm1(-/-) mice. We conclude that in the absence of Chm1, predominant primary bone healing occurs due to an indirect effect induced by reduction of cartilaginous callus rather than to a direct effect on osteogenic function, resulting in a delayed union.


Subject(s)
Bony Callus/physiopathology , Fracture Healing , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Animals , Bone Resorption , Immunohistochemistry , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
20.
Cell Tissue Res ; 332(1): 111-22, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18239943

ABSTRACT

Chondromodulin-I (ChM-I) and tenomodulin (TeM) are homologous angiogenesis inhibitors. We have analyzed the spatial relationships between capillary networks and the localization of these molecules during mouse and chick development. ChM-I and TeM proteins have been localized to the PECAM-1-negative avascular region: ChM-I is expressed in the avascular cartilage, whereas TeM is detectable in dense connective tissues, including tendons and ligaments. We have also examined the vasculature of chick embryos by injection with India ink and have performed in situ hybridization of the ChM-I and TeM genes. The onset of ChM-I expression is associated with chondrogenesis during mouse embryonic development. ChM-I expression is also detectable in precartilaginous or noncartilaginous avascular mesenchyme in chick embryos, including the somite, sclerotome, and heart. Hence, the expression domains of ChM-I and TeM during vertebrate development incorporate the typical avascular regions of the mesenchymal tissues.


Subject(s)
Embryo, Mammalian/metabolism , Embryo, Nonmammalian/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mesoderm/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cartilage/embryology , Cartilage/metabolism , Chick Embryo , Collagen Type II/genetics , Connective Tissue/embryology , Connective Tissue/metabolism , Gene Expression , Heart/embryology , High Mobility Group Proteins/genetics , Ligaments/embryology , Ligaments/metabolism , Membrane Proteins/metabolism , Mesoderm/embryology , Mice , Mice, Inbred ICR , Musculoskeletal System/embryology , Musculoskeletal System/metabolism , Myocardium/metabolism , Notochord/embryology , Notochord/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , SOX9 Transcription Factor , Spine/embryology , Spine/metabolism , Tendons/embryology , Tendons/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL