Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Bone ; 178: 116934, 2024 01.
Article in English | MEDLINE | ID: mdl-37839663

ABSTRACT

Intramembranous bone regeneration plays an important role in fixation of intramedullary implants used in joint replacement and dental implants used in tooth replacement. Despite widespread recognition of the importance of intramembranous bone regeneration in these clinical procedures, the underlying mechanisms have not been well explored. A previous study that examined transcriptomic profiles of regenerating bone from the marrow space showed that increased periostin gene expression preceded increases in several osteogenic genes. We therefore sought to determine the role of cells transiently expressing periostin in intramedullary intramembranous bone regeneration. We used a genetic mouse model that allows tamoxifen-inducible fluorescent labeling of periostin expressing cells. These mice underwent ablation of the bone marrow cavity through surgical disruption, a well-established intramembranous bone regeneration model. We found that in intact bones, fluorescently labeled cells were largely restricted to the periosteal surface of cortical bone and were absent in bone marrow. However, following surgical disruption of the bone marrow cavity, cells transiently expressing periostin were found within the regenerating tissue of the bone marrow compartment even though the cortical bone remained intact. The source of these cells is likely heterogenous, including cells occupying the periosteal surface as well as pericytes and endothelial cells within the marrow cavity. We also found that diphtheria toxin-mediated depletion of cells transiently expressing periostin at the time of surgery impaired intramembranous bone regeneration in mice. These data suggest a critical role of periostin expressing cells in intramedullary intramembranous bone regeneration and may lead to novel therapeutic interventions to accelerate or enhance implant fixation.


Subject(s)
Bone Regeneration , Endothelial Cells , Mice , Animals , Osteogenesis , Bone and Bones , Bone Marrow
2.
Adv Sci (Weinh) ; 10(36): e2302874, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37973554

ABSTRACT

Under diabetic conditions, blood glucose fluctuations and exacerbated immunopathological inflammatory environments pose significant challenges to periosteal regenerative repair strategies. Responsive immune regulation in damaged tissues is critical for the immune microenvironment, osteogenesis, and angiogenesis stabilization. Considering the high-glucose microenvironment of such acute injury sites, a functional glucose-responsive immunomodulation-assisted periosteal regeneration composite material-PLA(Polylactic Acid)/COLI(Collagen I)/Lipo(Liposome)-APY29 (PCLA)-is constructed. Aside from stimulating osteogenic differentiation, owing to the presence of surface self-assembled type I collagen in the scaffolds, PCLA can directly respond to focal area high-glucose microenvironments. The PCLA scaffolds trigger the release of APY29-loaded liposomes, shifting the macrophages toward the M2 phenotype, inhibiting the release of inflammatory cytokines, improving the bone immune microenvironment, and promoting osteogenic differentiation and angiogenesis. Bioinformatics analyses show that PCLA enhances bone repair by inhibiting the inflammatory signal pathway regulating the polarization direction and promoting osteogenic and angiogenic gene expression. In the calvarial periosteal defect model of diabetic rats, PCLA scaffolds induce M2 macrophage polarization and improve the inflammatory microenvironment, significantly accelerating periosteal repair. Overall, the PCLA scaffold material regulates immunity in fluctuating high-glucose inflammatory microenvironments, achieves relatively stable and favorable osteogenic microenvironments, and facilitates the effective design of functionalized biomaterials for bone regeneration therapy in patients with diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Osteogenesis , Rats , Humans , Animals , Periosteum , Tissue Scaffolds , Immunomodulation , Glucose
3.
Genes (Basel) ; 14(10)2023 10 14.
Article in English | MEDLINE | ID: mdl-37895286

ABSTRACT

Musculoskeletal diseases (MSDs) are characterized as injuries and illnesses that affect the musculoskeletal system. MSDs affect every population worldwide and are associated with substantial global burden. Variations in the makeup of the gut microbiota may be related to chronic MSDs. There is growing interest in exploring potential connections between chronic MSDs and variations in the composition of gut microbiota. The human microbiota is a complex community consisting of viruses, archaea, bacteria, and eukaryotes, both inside and outside of the human body. These microorganisms play crucial roles in influencing human physiology, impacting metabolic and immunological systems in health and disease. Different body areas host specific types of microorganisms, with facultative anaerobes dominating the gastrointestinal tract (able to thrive with or without oxygen), while strict aerobes prevail in the nasal cavity, respiratory tract, and skin surfaces (requiring oxygen for development). Together with the immune system, these bacteria have coevolved throughout time, forming complex biological relationships. Changes in the microbial ecology of the gut may have a big impact on health and can help illnesses develop. These changes are frequently impacted by lifestyle choices and underlying medical disorders. The potential for safety, expenses, and efficacy of microbiota-based medicines, even with occasional delivery, has attracted interest. They are, therefore, a desirable candidate for treating MSDs that are chronic and that may have variable progression patterns. As such, the following is a narrative review to address the role of the human microbiome as it relates to MSDs.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Musculoskeletal Diseases , Humans , Gastrointestinal Tract/microbiology , Bacteria , Oxygen
4.
Genes (Basel) ; 14(2)2023 02 04.
Article in English | MEDLINE | ID: mdl-36833336

ABSTRACT

Temporomandibular joint disorders (TMDs) are conditions that affect the muscles of mastication and joints that connect the mandible to the base of the skull. Although TMJ disorders are associated with symptoms, the causes are not well proven. Chemokines play an important role in the pathogenesis of TMJ disease by promoting chemotaxis inflammatory cells to destroy the joint synovium, cartilage, subchondral bone, and other structures. Therefore, enhancing our understanding of chemokines is critical for developing appropriate treatment of TMJ. In this review, we discuss chemokines including MCP-1, MIP-1α, MIP-3a, RANTES, IL-8, SDF-1, and fractalkine that are known to be involved in TMJ diseases. In addition, we present novel findings that CCL2 is involved in ß-catenin-mediated TMJ osteoarthritis (OA) and potential molecular targets for the development of effective therapies. The effects of common inflammatory factors, IL-1ß and TNF-α, on chemotaxis are also described. In conclusion, this review aims to provide a theoretical basis for future chemokine-targeted therapies for TMJ OA.


Subject(s)
Osteoarthritis , Temporomandibular Joint Disorders , Humans , Temporomandibular Joint Disorders/pathology , Osteoarthritis/pathology , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/pharmacology
5.
J Dent Sci ; 17(3): 1378-1386, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35784155

ABSTRACT

Background/purpose: The temporomandibular joint (TMJ) is a bi-arthrodial joint that is composed of the temporal bone glenoid fossa and the condylar head of the mandible both having fibrocartilaginous articular surfaces. Functional overloading of the TMJ is the main cause of TMJ osteoarthritis (TMJ OA) disease. The aim of this study was to establish immortalized TMJ fibrocartilage cell clones to provide enough cells to adequately investigate the molecular mechanisms studies of TMJ OA. Materials and methods: We have isolated temporomandibular condyle chondrocytes from adult Sprague Dawley rat. The cells were cultured and immortalized by treating with Y-27632, a well-characterized inhibitor of Rho-Associated Kinase (ROCK). Clones were characterized on the basis of cell morphology and analyses of marker gene expression through 45 passages. Results: Cells from the condylar fibrocartilage of the TMJ were successfully immortalized by ROCK inhibitor, retaining a consistent cuboidal cell morphology and the expression of several cell markers of polymorphic cell fate. In addition, they retained phenotype features similar to the primary parental TMJ fibrocartilage cells when the cells were challenged with different cytokines and growth factors. Conclusion: These studies establish a novel immortalized cell line through ROCK inhibitor Y-27632, that retains the polymorphic phenotype of primary cell lines from TMJ fibrocartilage chondrocyte cell through a high number of passages, serving as a valuable preclinical resource for mechanistic in vitro assessment of TMJ health, disease, and regeneration.

6.
Int J Oral Sci ; 12(1): 26, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32989215

ABSTRACT

Orofacial pain or tenderness is a primary symptom associated with temporomandibular joint (TMJ) disorders (TMDs). To understand the pathological mechanisms underlying TMDs, several mouse models have been developed, including mechanical stimulus-induced TMD and genetic mouse models. However, a lack of feasible approaches for assessing TMD-related nociceptive behaviours in the orofacial region of mice has hindered the in-depth study of TMD-associated mechanisms. This study aimed to explore modifications of three existing methods to analyse nociceptive behaviours using two TMD mouse models: (1) mechanical allodynia was tested using von Frey filaments in the mouse TMJ region by placing mice in specially designed chambers; (2) bite force was measured using the Economical Load and Force (ELF) system; and (3) spontaneous feeding behaviour tests, including eating duration and frequency, were analysed using the Laboratory Animal Behaviour Observation Registration and Analysis System (LABORAS). We successfully assessed changes in nociceptive behaviours in two TMD mouse models, a unilateral anterior crossbite (UAC)-induced TMD mouse model and a ß-catenin conditional activation mouse model. We found that the UAC model and ß-catenin conditional activation mouse model were significantly associated with signs of increased mechanical allodynia, lower bite force, and decreased spontaneous feeding behaviour, indicating manifestations of TMD. These behavioural changes were consistent with the cartilage degradation phenotype observed in these mouse models. Our studies have shown reliable methods to analyse nociceptive behaviours in mice and may indicate that these methods are valid to assess signs of TMD in mice.


Subject(s)
Malocclusion , Temporomandibular Joint Disorders , Animals , Disease Models, Animal , Facial Pain , Mice , Nociception , Temporomandibular Joint
7.
Sci Rep ; 10(1): 7790, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32385306

ABSTRACT

Snorc (Small NOvel Rich in Cartilage) has been identified as a chondrocyte-specific gene in the mouse. Yet little is known about the SNORC protein biochemical properties, and mechanistically how the gene is regulated transcriptionally in a tissue-specific manner. The goals of the present study were to shed light on those important aspects. The chondrocyte nature of Snorc expression was confirmed in mouse and rat tissues, in differentiated (day 7) ATDC5, and in RCS cells where it was constitutive. Topological mapping and biochemical analysis brought experimental evidences that SNORC is a type I protein carrying a chondroitin sulfate (CS) attached to serine 44. The anomalous migration of SNORC on SDS-PAGE was due to its primary polypeptide features, suggesting no additional post-translational modifications apart from the CS glycosaminoglycan. A highly conserved SOX9-binding enhancer located in intron 1 was necessary to drive transcription of Snorc in the mouse, rat, and human. The enhancer was active independently of orientation and whether located in a heterologous promoter or intron. Crispr-mediated inactivation of the enhancer in RCS cells caused reduction of Snorc. Transgenic mice carrying the intronic multimerized enhancer drove high expression of a ßGeo reporter in chondrocytes, but not in the hypertrophic zone. Altogether these data confirmed the chondrocyte-specific nature of Snorc and revealed dependency on the intronic enhancer binding of SOX9 for transcription.


Subject(s)
Chondrocytes/metabolism , Gene Expression Regulation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , SOX9 Transcription Factor/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cells, Cultured , Chondroitin Sulfates/metabolism , Humans , Membrane Proteins/chemistry , Mice , Mice, Transgenic , Open Reading Frames , Protein Binding , Protein Domains , Proteoglycans/chemistry , Rats , Transcription Initiation Site , Transcription, Genetic
8.
Ann Rheum Dis ; 79(5): 635-645, 2020 05.
Article in English | MEDLINE | ID: mdl-32156705

ABSTRACT

OBJECTIVES: In this study, we aim to determine the effect of metformin on osteoarthritis (OA) development and progression. METHODS: Destabilisation of the medial meniscus (DMM) surgery was performed in 10-week-old wild type and AMP-activated protein kinase (AMPK)α1 knockout (KO) mice. Metformin (4 mg/day in drinking water) was given, commencing either 2 weeks before or 2 weeks after DMM surgery. Mice were sacrificed 6 and 12 weeks after DMM surgery. OA phenotype was analysed by micro-computerised tomography (µCT), histology and pain-related behaviour tests. AMPKα1 (catalytic alpha subunit of AMPK) expression was examined by immunohistochemistry and immunofluorescence analyses. The OA phenotype was also determined by µCT and MRI in non-human primates. RESULTS: Metformin upregulated phosphorylated and total AMPK expression in articular cartilage tissue. Mild and more severe cartilage degeneration was observed at 6 and 12 weeks after DMM surgery, evidenced by markedly increased Osteoarthritis Research Society International scores, as well as reduced cartilage areas. The administration of metformin, commencing either before or after DMM surgery, caused significant reduction in cartilage degradation. Prominent synovial hyperplasia and osteophyte formation were observed at both 6 and 12 weeks after DMM surgery; these were significantly inhibited by treatment with metformin either before or after DMM surgery. The protective effects of metformin on OA development were not observed in AMPKα1 KO mice, suggesting that the chondroprotective effect of metformin is mediated by AMPK signalling. In addition, we demonstrated that treatment with metformin could also protect from OA progression in a partial medial meniscectomy animal model in non-human primates. CONCLUSIONS: The present study suggests that metformin, administered shortly after joint injury, can limit OA development and progression in injury-induced OA animal models.


Subject(s)
AMP-Activated Protein Kinases/genetics , Cartilage, Articular/drug effects , Metformin/pharmacology , Osteoarthritis/drug therapy , Up-Regulation/genetics , Animals , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/pathology , Disease Models, Animal , Disease Progression , Gene Expression Regulation , Humans , Hypoglycemic Agents/pharmacology , Menisci, Tibial/pathology , Menisci, Tibial/surgery , Mice , Mice, Knockout , Mice, Obese , Osteoarthritis/pathology , Random Allocation , Sensitivity and Specificity , Signal Transduction/genetics
9.
Sci Rep ; 7(1): 2371, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28539595

ABSTRACT

Runx2 may play an important role in development of osteoarthritis (OA). However, the specific role of Runx2 in articular chondrocyte function and in OA development in adult mice has not been fully defined. In this study, we performed the destabilization of the medial meniscus (DMM) surgery at 12-week-old mice to induce OA in adult Runx2 Agc1CreER mice, in which Runx2 was specifically deleted in Aggrecan-expressing chondrocytes by administering tamoxifen at 8-weeks of age. Knee joint samples were collected 8- and 12-weeks post-surgery and analyzed through histology, histomorphometry and micro-computed tomography (µCT). Our results showed that severe OA-like defects were observed after DMM surgery in Cre-negative control mice, including articular cartilage degradation and subchondral sclerosis, while the defects were significantly ameliorated in Runx2 Agc1CreER KO mice. Immunohistochemical (IHC) results showed significantly reduced expression of MMP13 in Runx2 Agc1CreER KO mice compared to that in Cre-negative control mice. Results of quantitative reverse-transcription PCR (qRT-PCR) demonstrated that expression of the genes encoding for matrix degradation enzymes was significantly decreased in Runx2 Agc1CreER KO mice. Thus, our findings suggest that inhibition of Runx2 in chondrocytes could at least partially rescue DMM-induced OA-like defects in adult mice.


Subject(s)
Chondrocytes/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Menisci, Tibial/surgery , Osteoarthritis/metabolism , Aggrecans/genetics , Aggrecans/metabolism , Animals , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Disease Progression , Gene Expression , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Menisci, Tibial/physiopathology , Mice, Knockout , Mice, Transgenic , Osteoarthritis/genetics , Osteoarthritis/physiopathology , X-Ray Microtomography
10.
J Biol Chem ; 292(2): 528-538, 2017 Jan 13.
Article in English | MEDLINE | ID: mdl-27881681

ABSTRACT

Type II collagen α1 is specific for cartilaginous tissues, and mutations in its gene are associated with skeletal diseases. Its expression has been shown to be dependent on SOX9, a master transcription factor required for chondrogenesis that binds to an enhancer region in intron 1. However, ChIP sequencing revealed that SOX9 does not strongly bind to intron 1, but rather it binds to intron 6 and a site 30 kb upstream of the transcription start site. Here, we aimed to determine the role of the novel SOX9-binding site in intron 6. We prepared reporter constructs that contain a Col2a1 promoter, intron 1 with or without intron 6, and the luciferase gene. Although the reporter constructs were not activated by SOX9 alone, the construct that contained both introns 1 and 6 was activated 5-10-fold by the SOX9/SOX5 or the SOX9/SOX6 combination in transient-transfection assays in 293T cells. This enhancement was also observed in rat chondrosarcoma cells that stably expressed the construct. CRISPR/Cas9-induced deletion of intron 6 in RCS cells revealed that a 10-bp region of intron 6 is necessary both for Col2a1 expression and SOX9 binding. Furthermore, SOX9, but not SOX5, binds to this region as demonstrated in an electrophoretic mobility shift assay, although both SOX9 and SOX5 bind to a larger 325-bp fragment of intron 6 containing this small sequence. These findings suggest a novel mechanism of action of SOX5/6; namely, the SOX9/5/6 combination enhances Col2a1 transcription through a novel enhancer in intron 6 together with the enhancer in intron 1.


Subject(s)
Collagen Type II/biosynthesis , Enhancer Elements, Genetic/physiology , Gene Expression Regulation/physiology , Introns/physiology , SOX9 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Collagen Type II/genetics , Humans , Mice , Rats , SOX9 Transcription Factor/genetics , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism , Transcription, Genetic/physiology
11.
Sci Rep ; 6: 29916, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27436052

ABSTRACT

Several lines of evidence indicate that connective tissue growth factor (CTGF/CCN2) stimulates chondrocyte proliferation and maturation. Given the fact that SOX9 is essential for several steps of the chondrocyte differentiation pathway, we asked whether Ctgf (Ccn2) is the direct target gene of SOX9. We found that Ctgf mRNA was down-regulated in primary sternal chondrocytes from Sox9(flox/flox) mice infected with Ad-CMV-Cre. We performed ChIP-on-chip assay using anti-SOX9 antibody, covering the Ctgf gene from 15 kb upstream of its 5'-end to 10 kb downstream of its 3'-end to determine SOX9 interaction site. One high-affinity interaction site was identified in the Ctgf proximal promoter by ChIP-on-chip assay. An important SOX9 regulatory element was found to be located in -70/-64 region of the Ctgf promoter. We found the same site for SOX9 binding to the Ctgf promoter in nucleus pulposus (NP) cells. The loss of Sox9 in growth plate chondrocytes in knee joint and in NP cells in intervertebral disc led to the decrease in CTGF expression. We suggest that Ctgf is the direct target gene of SOX9 in chondrocytes and NP cells. Our study establishes a strong link between two regulatory molecules that have a major role in cartilaginous tissues.


Subject(s)
Chondrocytes/cytology , Chondrocytes/metabolism , Connective Tissue Growth Factor/genetics , Growth Plate/cytology , Nucleus Pulposus/cytology , SOX9 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites , Cells, Cultured , Connective Tissue Growth Factor/metabolism , Extremities , Gene Deletion , Humans , Mice, Knockout , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sternum/cytology
12.
Mol Cell Biol ; 36(17): 2282-99, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27325675

ABSTRACT

Sphingomyelin phosphodiesterase 3 (SMPD3), a lipid-metabolizing enzyme present in bone and cartilage, has been identified to be a key regulator of skeletal development. A homozygous loss-of-function mutation called fragilitas ossium (fro) in the Smpd3 gene causes poor bone and cartilage mineralization resulting in severe congenital skeletal deformities. Here we show that Smpd3 expression in ATDC5 chondrogenic cells is downregulated by parathyroid hormone-related peptide through transcription factor SOX9. Furthermore, we show that transgenic expression of Smpd3 in the chondrocytes of fro/fro mice corrects the cartilage but not the bone abnormalities. Additionally, we report the generation of Smpd3(flox/flox) mice for the tissue-specific inactivation of Smpd3 using the Cre-loxP system. We found that the skeletal phenotype in Smpd3(flox/flox); Osx-Cre mice, in which the Smpd3 gene is ablated in both late-stage chondrocytes and osteoblasts, closely mimics the skeletal phenotype in fro/fro mice. On the other hand, Smpd3(flox/flox); Col2a1-Cre mice, in which the Smpd3 gene is knocked out in chondrocytes only, recapitulate the fro/fro mouse cartilage phenotype. This work demonstrates that Smpd3 expression in both chondrocytes and osteoblasts is required for normal endochondral bone development.


Subject(s)
Chondrocytes/cytology , Osteoblasts/cytology , Osteogenesis , SOX9 Transcription Factor/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrocytes/metabolism , Down-Regulation , Mice , Osteoblasts/metabolism , Sphingomyelin Phosphodiesterase/genetics
13.
Spine (Phila Pa 1976) ; 41(5): E255-61, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26693672

ABSTRACT

STUDY DESIGN: Establishment of immortalized cell lines derived from rat intervertebral disc cells by Rho-associated kinase (ROCK) inhibitor, Y-27632. OBJECTIVE: To determine whether rat nucleus pulposus (NP) and annulus fibrosus (AF) cells could be immortalized, retain their phenotype, and used as cell lines for in vitro cell biology. SUMMARY OF BACKGROUND DATA: Intervertebral disc degeneration is a major factor for most low-back pain. However, the mechanism of the disease is not well understood by the limitation to obtain sufficient amounts of primary disc cells. Therefore, the establishment of disc cell lines will help in vitro molecular signaling studies to understand the mechanism of degenerative disc disease. METHODS: Cells were isolated from the NP and AF tissues of lumbar discs of adult Sprague Dawley rat. Tissues were digested and cultured with DMEM/Ham's F-12 (1:1) and 20% FBS and antibiotics. From day 3, cells were grown in the presence of 10  µM Y-27632, a well-characterized inhibitor of the ROCK, and subcultured by trypsinization, passaging them 1:3 onto 100  mm culture dishes. Morphologic and genetic analyses were performed on the different passaged cells. RESULTS: ROCK inhibitor successfully immortalized rat NP and AF cells. They passaged for over 50 generations with sustained expression levels of several NP and AF cell markers. In addition, they retained phenotypic features similar to the primary parental NP and AF cells when the cells were challenged with different cytokines and growth factors. CONCLUSION: We established immortalized rat NP and AF cell lines using a method of treating cells with ROCK inhibitor Y-27632 and demonstrated that these immortalized cells retain the properties of primary cells and could serve as useful tools for in vitro signaling studies or drug screening studies to develop novel therapeutic strategies. LEVEL OF EVIDENCE: N/A.


Subject(s)
Amides/pharmacology , Annulus Fibrosus/cytology , Annulus Fibrosus/drug effects , Nucleus Pulposus/cytology , Nucleus Pulposus/drug effects , Pyridines/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Animals , Annulus Fibrosus/metabolism , Cell Line, Transformed , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Intervertebral Disc/cytology , Intervertebral Disc/drug effects , Intervertebral Disc/metabolism , Nucleus Pulposus/metabolism , Rats , Rats, Sprague-Dawley , rho-Associated Kinases/metabolism
15.
Gene ; 555(2): 80-7, 2015 Jan 25.
Article in English | MEDLINE | ID: mdl-25311550

ABSTRACT

Because miR-146a expression in articular chondrocytes is associated with osteoarthritis (OA), we assessed whether miR-146a is linked to cartilage degeneration in the spine. Monolayer cultures of nucleus pulposus (NP) cells from the intervertebral discs (IVD) of bovine tails were transfected with a miR-146a mimic. To provoke inflammatory responses and catabolic extracellular matrix (ECM) degradation, cells were co-treated with interleukin-1 (IL-1). Transfection of miR-146a decreases IL-1 induced mRNA levels of inflammatory genes and catabolic proteases in NP cells based on quantitative real-time reverse transcriptase PCR (qRT-PCR) analysis. Similarly, miR146a suppresses IL-1 induced protein levels of matrix metalloproteinases and aggrecanases as revealed by immunoblotting. Disc segments from wild type (WT) and miR-146a knockout (KO) mice were cultured ex vivo in the presence or absence of IL-1 for 3days. Histological and immuno-histochemical (IHC) analyses of disc organ cultures revealed that IL-1 mediates changes in proteoglycan (PG) content and in-situ levels of catabolic proteins (MMP-13 and ADAMTS-5) in the nucleus pulposus of the disc. However, these IL-1 effects are more pronounced in miR-146a KO discs compared to WT discs. For example, absence of miR-146a increases the percentage of MMP-13 and ADAMTS-5 positive cells after treatment with IL-1. Thus, miR-146a appears to protect against IL-1 induced IVD degeneration and inflammation. Stimulation of endogenous miR-146a expression or exogenous delivery of miRNA-146a are viable therapeutic strategies that may decelerate disc degeneration and regain a normal homeostatic balance in extracellular matrix production and turn-over.


Subject(s)
Gene Expression Regulation , Inflammation/metabolism , Interleukin-1/pharmacology , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc/metabolism , MicroRNAs/metabolism , ADAM Proteins/metabolism , ADAMTS5 Protein , Animals , Cattle , Cells, Cultured , Extracellular Matrix/metabolism , Homeostasis , Immunohistochemistry , In Vitro Techniques , Matrix Metalloproteinase 13/metabolism , Mice , Mice, Knockout , Proteoglycans/metabolism , Transfection
16.
PLoS One ; 9(9): e107577, 2014.
Article in English | MEDLINE | ID: mdl-25229425

ABSTRACT

The transcription factor SOX9 plays an essential role in determining the fate of several cell types and is a master factor in regulation of chondrocyte development. Our aim was to determine which genes in the genome of chondrocytes are either directly or indirectly controlled by SOX9. We used RNA-Seq to identify genes whose expression levels were affected by SOX9 and used SOX9 ChIP-Seq to identify those genes that harbor SOX9-interaction sites. For RNA-Seq, the RNA expression profile of primary Sox9flox/flox mouse chondrocytes infected with Ad-CMV-Cre was compared with that of the same cells infected with a control adenovirus. Analysis of RNA-Seq data indicated that, when the levels of Sox9 mRNA were decreased more than 8-fold by infection with Ad-CMV-Cre, 196 genes showed a decrease in expression of at least 4-fold. These included many cartilage extracellular matrix (ECM) genes and a number of genes for ECM modification enzymes (transferases), membrane receptors, transporters, and others. In ChIP-Seq, 75% of the SOX9-interaction sites had a canonical inverted repeat motif within 100 bp of the top of the peak. SOX9-interaction sites were found in 55% of the genes whose expression was decreased more than 8-fold in SOX9-depleted cells and in somewhat fewer of the genes whose expression was reduced more than 4-fold, suggesting that these are direct targets of SOX9. The combination of RNA-Seq and ChIP-Seq has provided a fuller understanding of the SOX9-controlled genetic program of chondrocytes.


Subject(s)
Chondrocytes/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , SOX9 Transcription Factor/metabolism , Animals , Binding Sites , Chromatin Immunoprecipitation , Gene Expression , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Mice , Mice, Knockout , Nucleotide Motifs , Position-Specific Scoring Matrices , Protein Binding , Protein Transport , SOX9 Transcription Factor/genetics
17.
Nucleic Acids Res ; 38(18): 6018-28, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20484372

ABSTRACT

Mutations in SOX9, a gene essential for chondrocyte differentiation cause the human disease campomelic dysplasia (CD). To understand how SOX9 activates transcription, we characterized the DNA binding and cell-free transcription ability of wild-type SOX9 and a dimerization domain SOX9 mutant. Whereas formation of monomeric mutant SOX9-DNA complex increased linearly with increasing SOX9 concentrations, formation of a wild-type SOX9-DNA dimeric complex increased more slowly suggesting a more sigmoidal-type progression. Stability of SOX9-DNA complexes, however, was unaffected by the dimerization mutation. Both wild-type and mutant SOX9 activated transcription of a naked Col2a1 DNA template. However, after nucleosomal assembly, only wild-type and not the mutant was able to remodel chromatin and activate transcription of this template. Using a cell line, in which the Col2a1 vector was stably integrated, no differences were seen in the interactions of wild-type and mutant SOX9 with the chromatin of the Col2a1 vector using ChIP. However, the mutant was unable to activate transcription in agreement with in vitro results. We hypothesize that the SOX9 dimerization domain is necessary to remodel the Col2a1 chromatin in order to allow transcription to take place. These results further clarify the mechanism that accounts for CD in patients harboring SOX9 dimerization domain mutations.


Subject(s)
Chondrocytes/metabolism , Chromatin Assembly and Disassembly , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcriptional Activation , Animals , Cell Line , Chromatin/metabolism , Collagen Type II/genetics , DNA/metabolism , Dimerization , Enhancer Elements, Genetic , Humans , Mutation , Protein Structure, Tertiary , Recombinant Proteins/metabolism , SOX9 Transcription Factor/chemistry
18.
PLoS One ; 5(4): e10113, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20404928

ABSTRACT

BACKGROUND: Our previous work has provided strong evidence that the transcription factor SOX9 is completely needed for chondrogenic differentiation and cartilage formation acting as a "master switch" in this differentiation. Heterozygous mutations in SOX9 cause campomelic dysplasia, a severe skeletal dysmorphology syndrome in humans characterized by a generalized hypoplasia of endochondral bones. To obtain insights into the logic used by SOX9 to control a network of target genes in chondrocytes, we performed a ChIP-on-chip experiment using SOX9 antibodies. METHODOLOGY/PRINCIPAL FINDINGS: The ChIP DNA was hybridized to a microarray, which covered 80 genes, many of which are involved in chondrocyte differentiation. Hybridization peaks were detected in a series of cartilage extracellular matrix (ECM) genes including Col2a1, Col11a2, Aggrecan and Cdrap as well as in genes for specific transcription factors and signaling molecules. Our results also showed SOX9 interaction sites in genes that code for proteins that enhance the transcriptional activity of SOX9. Interestingly, a strong SOX9 signal was also observed in genes such as Col1a1 and Osx, whose expression is strongly down regulated in chondrocytes but is high in osteoblasts. In the Col2a1 gene, in addition to an interaction site on a previously identified enhancer in intron 1, another strong interaction site was seen in intron 6. This site is free of nucleosomes specifically in chondrocytes suggesting an important role of this site on Col2a1 transcription regulation by SOX9. CONCLUSIONS/SIGNIFICANCE: Our results provide a broad understanding of the strategies used by a "master" transcription factor of differentiation in control of the genetic program of chondrocytes.


Subject(s)
Cell Differentiation/genetics , Chondrocytes/cytology , Collagen Type II/genetics , Genome , SOX9 Transcription Factor/genetics , Animals , Binding Sites , Cartilage/cytology , Cell Line , Extracellular Matrix Proteins/genetics , Gene Expression Profiling , Gene Regulatory Networks , Oligonucleotide Array Sequence Analysis , Rats , SOX9 Transcription Factor/metabolism
19.
Development ; 136(8): 1263-72, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19261698

ABSTRACT

Limb development constitutes a central model for the study of tissue and organ patterning; yet, the mechanisms that regulate the patterning of limb vasculature have been left understudied. Vascular patterning in the forming limb is tightly regulated in order to ensure sufficient gas exchange and nutrient supply to the developing organ. Once skeletogenesis is initiated, limb vasculature undergoes two seemingly opposing processes: vessel regression from regions that undergo mesenchymal condensation; and vessel morphogenesis. During the latter, vessels that surround the condensations undergo an extensive rearrangement, forming a stereotypical enriched network that is segregated from the skeleton. In this study, we provide evidence for the centrality of the condensing mesenchyme of the forming skeleton in regulating limb vascular patterning. Both Vegf loss- and gain-of-function experiments in limb bud mesenchyme firmly established VEGF as the signal by which the condensing mesenchyme regulates the vasculature. Normal vasculature observed in limbs where VEGF receptors Flt1, Flk1, Nrp1 and Nrp2 were blocked in limb bud mesenchyme suggested that VEGF, which is secreted by the condensing mesenchyme, regulates limb vasculature via a direct long-range mechanism. Finally, we provide evidence for the involvement of SOX9 in the regulation of Vegf expression in the condensing mesenchyme. This study establishes Vegf expression in the condensing mesenchyme as the mechanism by which the skeleton patterns limb vasculature.


Subject(s)
Body Patterning , Bone and Bones/blood supply , Bone and Bones/metabolism , Limb Buds/blood supply , Limb Buds/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Bone and Bones/embryology , Gene Expression Regulation, Developmental , Limb Buds/embryology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/genetics
20.
J Cell Biochem ; 89(4): 837-47, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12858348

ABSTRACT

Retinoic acid (RA) is a well-known regulator of chondrocyte phenotype. RA inhibits chondrogenic differentiation of mesenchymal cells and also causes loss of differentiated chondrocyte phenotype. The present study investigated the mechanisms underlying RA regulation of chondrogenesis. RA treatment in chondrifying mesenchymal cells did not affect precartilage condensation, but blocked progression from precartilage condensation to cartilage nodule formation. This inhibitory effect of RA was independent of protein kinase C and extracellular signal-regulated protein kinase, which are positive and negative regulators of cartilage nodule formation, respectively. The progression from precartilage condensation to cartilage nodule requires downregulation of N-cadherin expression. However, RA treatment caused sustained expression of N-cadherin and its associated proteins including alpha- and beta-catenin suggesting that modulation of expression of these molecules is associated with RA-induced inhibition of chondrogenesis. This hypothesis was supported by the observation that disruption of the actin cytoskeleton by cytochalasin D (CD) blocks RA-induced sustained expression of cell adhesion molecules and overcomes RA-induced inhibition of chondrogenesis. Taken together, our results suggest RA inhibits chondrogenesis by stabilizing cell-to-cell interactions at the post-precartilage condensation stage.


Subject(s)
Cadherins/biosynthesis , Chondrogenesis/drug effects , Mesoderm/metabolism , Tretinoin/pharmacology , Animals , Cartilage/drug effects , Cartilage/metabolism , Cells, Cultured , Chick Embryo , Collagen/analogs & derivatives , Collagen/biosynthesis , Cytochalasin D/pharmacology , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Limb Buds , Mesoderm/cytology , Mesoderm/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Proteoglycans/analysis , Trans-Activators/biosynthesis , Trans-Activators/drug effects , Tretinoin/antagonists & inhibitors , alpha Catenin , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...