Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
1.
J Bone Miner Res ; 39(8): 1162-1173, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-38836497

ABSTRACT

X-linked hypophosphatemia (XLH) is caused by mutations in PHEX, leading to rickets and osteomalacia. Adults affected with XLH develop a mineralization of the bone-tendon attachment site (enthesis), called enthesopathy, which causes significant pain and impaired movement. Entheses in mice with XLH (Hyp) have enhanced bone morphogenetic protein (BMP) and Indian hedgehog (IHH) signaling. Treatment of Hyp mice with the BMP signaling blocker palovarotene attenuated BMP/IHH signaling in Hyp entheses, thus indicating that BMP signaling plays a pathogenic role in enthesopathy development and that IHH signaling is activated by BMP signaling in entheses. It was previously shown that mRNA expression of growth/differentiation factor 5 (Gdf5) is enhanced in Hyp entheses at P14. Thus, to determine a role for GDF5 in enthesopathy development, Gdf5 was deleted globally in Hyp mice and conditionally in Scx + cells of Hyp mice. In both murine models, BMP/IHH signaling was similarly decreased in Hyp entheses, leading to decreased enthesopathy. BMP/IHH signaling remained unaffected in WT entheses with decreased Gdf5 expression. Moreover, deletion of Gdf5 in Hyp entheses starting at P30, after enthesopathy has developed, partially reversed enthesopathy. Taken together, these results demonstrate that while GDF5 is not essential for modulating BMP/IHH signaling in WT entheses, inappropriate GDF5 activity in Scx + cells contributes to XLH enthesopathy development. As such, inhibition of GDF5 signaling may be beneficial for the treatment of XLH enthesopathy.


X-linked hypophosphatemia (XLH) is a rare bone disorder that leads to short stature and poorly mineralized bones. As adults, patients with XLH often develop a mineralization of the bone-tendon attachment site, called enthesopathy, which results in significant pain. We previously showed that Achilles bone-tendon attachment sites (entheses) in mice with XLH (Hyp) have an enthesopathy characterized by increased bone morphogenetic protein (BMP) signaling. In the current studies, we show that treating Hyp mice with the BMP signaling inhibitor palovarotene prevents enthesopathy, demonstrating that the increased BMP signaling in Hyp entheses leads to enthesopathy development. We also reported that gene expression of Gdf5, which activates BMP signaling, is enhanced in Hyp entheses. Therefore, to determine if the enhanced Gdf5 expression leads to the increased BMP signaling seen Hyp entheses, Gdf5 was deleted from Hyp mice and also deleted specifically in the entheses of Hyp mice. In both mouse models, enthesopathy development was attenuated, demonstrating that the increased Gdf5 expression in Hyp entheses plays a role in enthesopathy development. These data indicate that blocking GDF5 and BMP signaling may prevent enthesopathy in patients with XLH.


Subject(s)
Enthesopathy , Familial Hypophosphatemic Rickets , Growth Differentiation Factor 5 , Animals , Mice , Bone Morphogenetic Proteins/metabolism , Disease Models, Animal , Enthesopathy/genetics , Enthesopathy/metabolism , Enthesopathy/pathology , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/metabolism , Familial Hypophosphatemic Rickets/pathology , Growth Differentiation Factor 5/metabolism , Growth Differentiation Factor 5/genetics , Signal Transduction
2.
Cardiovasc Pathol ; 68: 107581, 2024.
Article in English | MEDLINE | ID: mdl-37838075

ABSTRACT

BACKGROUND: We previously showed that growth differentiation factor 5 (GDF5) limits infarct expansion post-myocardial infarction (MI). We now examine the acute post-MI role of GDF5 in cardiac rupture. METHODS AND RESULTS: Following permanent ligation of the left anterior descending artery, GDF5 deficiency (i.e., GDF5 knockout mice) reduced the incidence of cardiac rupture (4/24 vs. 17/24; P < .05), and improved survival over 28-d compared to wild-type (WT) mice (79% vs. 25%; P < .0001). Moreover, at 3-d post-MI, GDF5-deficient mice manifest: (a) reduced heart weight/body weight ratio (P < .0001) without differences in infarct size or cardiomyocyte size; (b) increased infarct zone expression of Col1a1 (P < .05) and Col3a1 (P < .01), suggesting increased myocardial fibrosis; and (c) reduced aortic and left ventricular peak systolic pressures (P ≤ .05), suggesting reduced afterload. Despite dysregulated inflammatory markers and reduced circulating monocytes in GDF5-deficient mice at 3-d post-MI, reciprocal bone marrow transplantation (BMT) failed to implicate GDF5 in BM-derived cells, suggesting the involvement of tissue-resident GDF5 expression in cardiac rupture. CONCLUSIONS: Loss of GDF5 reduces cardiac rupture post-MI with increased myocardial fibrosis and lower afterload, albeit at the cost of chronic adverse remodeling.


Subject(s)
Growth Differentiation Factor 5 , Heart Rupture , Myocardial Infarction , Animals , Mice , Disease Models, Animal , Fibrosis , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Heart Rupture/genetics , Heart Rupture/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/complications , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardium/pathology
3.
Sci Rep ; 13(1): 22778, 2023 12 20.
Article in English | MEDLINE | ID: mdl-38123662

ABSTRACT

Growth differentiation factor 5 (GDF5), a BMP family member, is highly expressed in the surface layer of articular cartilage. The GDF5 gene is a key risk locus for osteoarthritis and Gdf5-deficient mice show abnormal joint development, indicating that GDF5 is essential in joint development and homeostasis. In this study, we aimed to identify transcription factors involved in Gdf5 expression by performing two-step screening. We first performed microarray analyses to find transcription factors specifically and highly expressed in the superficial zone (SFZ) cells of articular cartilage, and isolated 11 transcription factors highly expressed in SFZ cells but not in costal chondrocytes. To further proceed with the identification, we generated Gdf5-HiBiT knock-in (Gdf5-HiBiT KI) mice, by which we can easily and reproducibly monitor Gdf5 expression, using CRISPR/Cas9 genome editing. Among the 11 transcription factors, Hoxa10 clearly upregulated HiBiT activity in the SFZ cells isolated from Gdf5-HiBiT KI mice. Hoxa10 overexpression increased Gdf5 expression while Hoxa10 knockdown decreased it in the SFZ cells. Moreover, ChIP and promoter assays proved the direct regulation of Gdf5 expression by HOXA10. Thus, our results indicate the important role played by HOXA10 in Gdf5 regulation and the usefulness of Gdf5-HiBiT KI mice for monitoring Gdf5 expression.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Mice , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , Transcription Factors/metabolism
4.
Med Sci (Paris) ; 39 Hors série n° 1: 47-53, 2023 Nov.
Article in French | MEDLINE | ID: mdl-37975770

ABSTRACT

Sarcopenia is a complex age-related muscular disease affecting 10 to 16 % of people over 65 years old. It is characterized by excessive loss of muscle mass and strength. Despite a plethora of studies aimed at understanding the physiological mechanisms underlying this pathology, the pathophysiology of sarcopenia remains poorly understood. To date, there is no pharmacological treatment for this disease. In this context, our team develop therapeutic approaches based on the GDF5 protein to counteract the loss of muscle mass and function in various pathological conditions, including sarcopenia. After deciphering one of the molecular mechanisms governing GDF5 expression, we have demonstrated the therapeutic potential of this protein in the preservation of muscle mass and strength in aged mice.


Title: GDF5 - Un candidat thérapeutique dans la lutte contre la sarcopénie. Abstract: La sarcopénie est une maladie musculaire complexe liée à l'âge qui affecte entre 10 à 16 % des personnes âgées de plus 65 ans. Elle se caractérise par une perte excessive de la masse musculaire et de la force. Malgré la multitude d'études visant à comprendre les mécanismes physiologiques qui sous-tendent cette pathologie, la physiopathologie de la sarcopénie reste encore mal comprise. A ce jour, il n'existe pas de traitement pharmacologique pour lutter contre cette pathologie. Dans ce contexte, notre équipe développe des approches thérapeutiques basées sur l'utilisation de la protéine GDF5 pour contrecarrer la perte de la masse et de la fonction musculaire dans diverses conditions pathologiques dont la sarcopénie. Après avoir décrypté un des mécanismes moléculaires régulant l'expression du GDF5, nous avons démontré le potentiel thérapeutique de cette protéine dans la préservation de la masse et la force musculaire chez les souris âgées.


Subject(s)
Sarcopenia , Aged , Animals , Humans , Mice , Growth Differentiation Factor 5/metabolism , Muscle, Skeletal/pathology , Sarcopenia/drug therapy , Sarcopenia/genetics
5.
Mol Biol Rep ; 50(8): 6337-6347, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37310547

ABSTRACT

BACKGROUND: Degenerative disc disease(DDD)is one of the most important causes of low back pain (LBP). Programmed death of human nucleus pulposus mesenchymal stem cells (NPMSCs) plays an important role in the progression of DDD. Growth differentiation factor-5 (GDF-5) is a protein that promotes chondrogenic differentiation, and has been reported to slow the expression of inflammatory factors in nucleus pulposus cells. Compared with those in normal rats, MRI T2-weighted images show hypointense in the central nucleus pulposus region of the intervertebral disc in GDF-5 knockout rats. METHODS AND RESULTS: We aimed to evaluate the role of GDF-5 and Ras homolog family member A (RhoA) in NPMSCs. We used lipopolysaccharide (LPS) to simulate the inflammatory environment in degenerative disc disease, and performed related experiments on the effects of GDF-5 on NPMSCs, including the effects of pyroptosis, RhoA protein, and the expression of extracellular matrix components, and the effects of GDF-5, on NPMSCs. In addition, the effect of GDF-5 on chondroid differentiation of NPMSCs was included. The results showed that the addition of GDF-5 inhibited the LPS-induced pyroptosis of NPMSCs, and further analysis of its mechanism showed that this was achieved by activating the RhoA signaling pathway. CONCLUSION: These findings suggest that GDF-5 plays an important role in inhibiting the pyroptosis of NPMSCs and GDF-5 may have potential for degenerative disc disease gene-targeted therapy in the future.


Subject(s)
Intervertebral Disc Degeneration , Mesenchymal Stem Cells , Nucleus Pulposus , Animals , Humans , Rats , Growth Differentiation Factor 5/metabolism , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/therapy , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Mesenchymal Stem Cells/metabolism , Nucleus Pulposus/metabolism , Pyroptosis , rhoA GTP-Binding Protein/metabolism , Signal Transduction
6.
Int J Mol Sci ; 24(5)2023 Feb 25.
Article in English | MEDLINE | ID: mdl-36901991

ABSTRACT

Skeletal muscle-fat interaction is essential for maintaining organismal energy homeostasis and managing obesity by secreting cytokines and exosomes, but the role of the latter as a new mediator in inter-tissue communication remains unclear. Recently, we discovered that miR-146a-5p was mainly enriched in skeletal muscle-derived exosomes (SKM-Exos), 50-fold higher than in fat exosomes. Here, we investigated the role of skeletal muscle-derived exosomes regulating lipid metabolism in adipose tissue by delivering miR-146a-5p. The results showed that skeletal muscle cell-derived exosomes significantly inhibited the differentiation of preadipocytes and their adipogenesis. When the skeletal muscle-derived exosomes co-treated adipocytes with miR-146a-5p inhibitor, this inhibition was reversed. Additionally, skeletal muscle-specific knockout miR-146a-5p (mKO) mice significantly increased body weight gain and decreased oxidative metabolism. On the other hand, the internalization of this miRNA into the mKO mice by injecting skeletal muscle-derived exosomes from the Flox mice (Flox-Exos) resulted in significant phenotypic reversion, including down-regulation of genes and proteins involved in adipogenesis. Mechanistically, miR-146a-5p has also been demonstrated to function as a negative regulator of peroxisome proliferator-activated receptor γ (PPARγ) signaling by directly targeting growth and differentiation factor 5 (GDF5) gene to mediate adipogenesis and fatty acid absorption. Taken together, these data provide new insights into the role of miR-146a-5p as a novel myokine involved in the regulation of adipogenesis and obesity via mediating the skeletal muscle-fat signaling axis, which may serve as a target for the development of therapies against metabolic diseases, such as obesity.


Subject(s)
Exosomes , MicroRNAs , Mice , Animals , PPAR gamma/metabolism , Adipogenesis/genetics , Adipose Tissue/metabolism , MicroRNAs/genetics , Muscle, Skeletal/metabolism , Obesity/metabolism , Exosomes/metabolism , Growth Differentiation Factor 5/metabolism
7.
Hand (N Y) ; 18(3): 436-445, 2023 05.
Article in English | MEDLINE | ID: mdl-34340572

ABSTRACT

BACKGROUND: As hand surgeons, tendon injuries and lacerations are a particularly difficult problem to treat, as poor healing potential and adhesions hamper optimal recovery. Adipose-derived stem cells (ADSCs) have been shown to aid in rat Achilles tendon healing after a puncture defect, and this model can be used to study tendon healing in the upper extremity. We hypothesized that ADSCs cultured with growth differentiation factor 5 (GDF5) and platelet-derived growth factor (PDGF) would improve tendon healing after a transection injury. METHODS: Rat Achilles tendons were transected and then left either unrepaired or repaired. Both groups were treated with a hydrogel alone, a hydrogel with ADSCs, or a hydrogel with ADSCs that were cultured with GDF5 and PDGF prior to implantation. Tissue harvested from the tendons was evaluated for gene expression of several genes known to play an important role in successful tendon healing. Histological examination of the tendon healing was also performed. RESULTS: In both repaired and unrepaired tendons, those treated with ADSCs cultured with GDF5/PDGF prior to implantation showed the best tendon fiber organization, the smallest gaps, and the most organized blood vessels. Treatment with GDF5/PDGF increased expression of the protenogenesis gene SOX9, promoted cell-to-cell connections, improved cellular proliferation, and enhanced tissue remodeling. CONCLUSIONS: Adipose-derived stem cells cultured with GDF5/PDGF prior to implantation can promote tendon repair by improving cellular proliferation, tenogenesis, and vascular infiltration. This effect results in a greater degree of organized tendon healing.


Subject(s)
Achilles Tendon , Platelet-Derived Growth Factor , Rats , Animals , Platelet-Derived Growth Factor/metabolism , Growth Differentiation Factor 5/metabolism , Hydrogels/metabolism , Stem Cells
8.
Article in English | MEDLINE | ID: mdl-35675541

ABSTRACT

OBJECTIVE: To explore the role of WNT family member 1 (WNT1) in the development of dysplasia of the hip (DDH) and the molecular mechanism involved in this process. Methods: Si-WNT1, pcDNA3.1-WNT1 or corresponding negative controls were transfected into human osteoblast hFOB1.19 and human chondrocyte C28/I2, respectively. The proliferation of cells was measured by EdU assay. The relative expressions of human noggin gene (NOG), growth differentiating factor 5 (GDF5), WNT1, and WNT1-inducible-signaling pathway protein 2 (WISP2) were determined by immunofluorescence analysis. The protein expressions of RNA-binding protein of multiple splice forms 2 (RBPMS2), NOG, bone morphogenetic protein 2 (BMP2), BMP4, WNT1 and WISP2 were determined by western blot. Animal experiment was also performed and the morphological development of hip joint was observed. Results: Overexpression of WNT1 promoted osteoblast proliferation and inhibited chondrocyte proliferation, while knockdown of WNT1 inhibited osteoblast proliferation. In chondrocytes, knockdown of WNT1 upregulated NOG expression, while overexpression of WNT1 downregulated its expression. In osteoblasts and chondrocytes, overexpression of WNT1 increased BMP2, BMP4, WNT1, and WISP2 expression. RBPMS2 and NOG were slightly expressed in each group. Conclusion: Overexpression of WNT1 promoted osteoblast proliferation, inhibited chondrocyte proliferation, and increased the expressions of BMP2, BMP4, WNT1, and WISP2. Therefore, WNT1 may be a new therapeutic target for DDH.


Subject(s)
Hip Dislocation, Congenital , Osteoblasts , Wnt1 Protein , Animals , Bone Morphogenetic Protein 2/metabolism , CCN Intercellular Signaling Proteins/metabolism , Cell Differentiation , Cell Proliferation , Factor V/metabolism , Growth Differentiation Factor 5/metabolism , Hip Dislocation, Congenital/metabolism , Humans , Osteoblasts/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Wnt1 Protein/metabolism
9.
Development ; 149(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35451016

ABSTRACT

It has been established in the mouse model that during embryogenesis joint cartilage is generated from a specialized progenitor cell type, distinct from that responsible for the formation of growth plate cartilage. We recently found that mesodermal progeny of human pluripotent stem cells gave rise to two types of chondrogenic mesenchymal cells in culture: SOX9+ and GDF5+ cells. The fast-growing SOX9+ cells formed in vitro cartilage that expressed chondrocyte hypertrophy markers and readily underwent mineralization after ectopic transplantation. In contrast, the slowly growing GDF5+ cells derived from SOX9+ cells formed cartilage that tended to express low to undetectable levels of chondrocyte hypertrophy markers, but expressed PRG4, a marker of embryonic articular chondrocytes. The GDF5+-derived cartilage remained largely unmineralized in vivo. Interestingly, chondrocytes derived from the GDF5+ cells seemed to elicit these activities via non-cell-autonomous mechanisms. Genome-wide transcriptomic analyses suggested that GDF5+ cells might contain a teno/ligamento-genic potential, whereas SOX9+ cells resembled neural crest-like progeny-derived chondroprogenitors. Thus, human pluripotent stem cell-derived GDF5+ cells specified to generate permanent-like cartilage seem to emerge coincidentally with the commitment of the SOX9+ progeny to the tendon/ligament lineage.


Subject(s)
Cartilage, Articular , Chondrocytes , Pluripotent Stem Cells , Animals , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cell Differentiation , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrogenesis , Growth Differentiation Factor 5/metabolism , Humans , Hypertrophy , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
10.
Stem Cell Res Ther ; 13(1): 130, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35346361

ABSTRACT

BACKGROUND: The treatment of bone loss has posed a challenge to clinicians for decades. Thus, it is of great significance to identify more effective methods for bone regeneration. However, the role and mechanisms of long non-coding RNA small nucleolar RNA host gene 5 (SNHG5) during osteogenic differentiation remain unclear. METHODS: We investigated the function of SNHG5, Yin Yang 1 (YY1), miR-212-3p and growth differentiation factor 5 (GDF5) in osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) in vitro and in vivo. Molecular mechanisms were clarified by chromatin immunoprecipitation assay and dual luciferase reporter assay. RESULTS: We found SNHG5 expression was upregulated during osteogenesis of hBMSCs. Knockdown of SNHG5 in hBMSCs inhibited osteogenic differentiation while overexpression of SNHG5 promoted osteogenesis. Moreover, YY1 transcription factor directly bound to the promoter region of SNHG5 and regulated SNHG5 expression to promote osteogenesis. Dual luciferase reporter assay confirmed that SNHG5 acted as a miR-212-3p sponge and miR-212-3p directly targeted GDF5 and further activated Smad1/5/8 phosphorylation. miR-212-3p inhibited osteogenic differentiation, while GDF5 promoted osteogenic differentiation of hBMSCs. In addition, calvarial defect experiments showed knockdown of SNHG5 and GDF5 inhibited new bone formation in vivo. CONCLUSION: Our results demonstrated that the novel pathway YY1/SNHG5/miR-212-3p/GDF5/Smad regulates osteogenic differentiation of hBMSCs and may serve as a potential target for the treatment of bone loss.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Osteogenesis , RNA, Long Noncoding , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Humans , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , RNA, Long Noncoding/genetics
11.
Dev Dyn ; 251(9): 1535-1549, 2022 09.
Article in English | MEDLINE | ID: mdl-34242444

ABSTRACT

BACKGROUND: The development of the vertebrate limb skeleton requires a complex interaction of multiple factors to facilitate the correct shaping and positioning of bones and joints. Growth and differentiation factor 5 (Gdf5) is involved in patterning appendicular skeletal elements including joints. Expression of gdf5 in zebrafish has been detected in fin mesenchyme condensations and segmentation zones as well as the jaw joint, however, little is known about the functional role of Gdf5 outside of Amniota. RESULTS: We generated CRISPR/Cas9 knockout of gdf5 in zebrafish and analyzed the resulting phenotype at different developmental stages. Homozygous gdf5 mutant zebrafish displayed changes in segmentation of the endoskeletal disc and, as a consequence, loss of posterior radials in the pectoral fins. Mutant fish also displayed disorganization and reduced length of endoskeletal elements in the median fins, while joints and mineralization seemed unaffected. CONCLUSIONS: Our study demonstrates the importance of Gdf5 in the development of the zebrafish pectoral and median fin endoskeleton and reveals that the severity of the effect increases from anterior to posterior elements. Our findings are consistent with phenotypes observed in the human and mouse appendicular skeleton in response to Gdf5 knockout, suggesting a broadly conserved role for Gdf5 in Osteichthyes.


Subject(s)
Gene Expression Regulation, Developmental , Growth Differentiation Factor 5 , Zebrafish , Animal Fins/metabolism , Animals , Bone and Bones/metabolism , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Mice , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Int J Mol Sci ; 22(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34681754

ABSTRACT

Osteoarthritis (OA) is a common articular disease manifested by the destruction of cartilage and compromised chondrogenesis in the aging population, with chronic inflammation of synovium, which drives OA progression. Importantly, the activated synovial fibroblast (AF) within the synovium facilitates OA through modulating key molecules, including regulatory microRNAs (miR's). To understand OA associated pathways, in vitro co-culture system, and in vivo papain-induced OA model were applied for this study. The expression of key inflammatory markers both in tissue and blood plasma were examined by qRT-PCR, western blot, immunohistochemistry, enzyme-linked immunosorbent assay (ELISA) and immunofluorescence assays. Herein, our result demonstrated, AF-activated human chondrocytes (AC) exhibit elevated NFκB, TNF-α, IL-6, and miR-21 expression as compared to healthy chondrocytes (HC). Importantly, AC induced the apoptosis of HC and inhibited the expression of chondrogenesis inducers, SOX5, TGF-ß1, and GDF-5. NFκB is a key inflammatory transcription factor elevated in OA. Therefore, SC75741 (an NFκB inhibitor) therapeutic effect was explored. SC75741 inhibits inflammatory profile, protects AC-educated HC from apoptosis, and inhibits miR-21 expression, which results in the induced expression of GDF-5, SOX5, TGF-ß1, BMPR2, and COL4A1. Moreover, ectopic miR-21 expression in fibroblast-like activated chondrocytes promoted osteoblast-mediated differentiation of osteoclasts in RW264.7 cells. Interestingly, in vivo study demonstrated SC75741 protective role, in controlling the destruction of the articular joint, through NFκB, TNF-α, IL-6, and miR-21 inhibition, and inducing GDF-5, SOX5, TGF-ß1, BMPR2, and COL4A1 expression. Our study demonstrated the role of NFκB/miR-21 axis in OA progression, and SC75741's therapeutic potential as a small-molecule inhibitor of miR-21/NFκB-driven OA progression.


Subject(s)
Benzimidazoles/pharmacology , Chondrocytes/drug effects , Fibroblasts/drug effects , NF-kappa B/antagonists & inhibitors , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Piperidines/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Animals , Benzimidazoles/chemistry , Cell Differentiation/genetics , Chondrocytes/metabolism , Chondrocytes/pathology , Collagen Type IV/genetics , Collagen Type IV/metabolism , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Humans , Interleukin-1beta/pharmacology , MicroRNAs/metabolism , NF-kappa B/metabolism , Osteoarthritis/pathology , Piperidines/chemistry , Pyrimidines/chemistry , Rats, Wistar , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism , Signal Transduction/drug effects , Synovial Membrane/pathology , Thiazoles/chemistry
13.
Nat Commun ; 12(1): 5363, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34508093

ABSTRACT

The activity of epiphyseal growth plates, which drives long bone elongation, depends on extensive changes in chondrocyte size and shape during differentiation. Here, we develop a pipeline called 3D Morphometric Analysis for Phenotypic significance (3D MAPs), which combines light-sheet microscopy, segmentation algorithms and 3D morphometric analysis to characterize morphogenetic cellular behaviors while maintaining the spatial context of the growth plate. Using 3D MAPs, we create a 3D image database of hundreds of thousands of chondrocytes. Analysis reveals broad repertoire of morphological changes, growth strategies and cell organizations during differentiation. Moreover, identifying a reduction in Smad 1/5/9 activity together with multiple abnormalities in cell growth, shape and organization provides an explanation for the shortening of Gdf5 KO tibias. Overall, our findings provide insight into the morphological sequence that chondrocytes undergo during differentiation and highlight the ability of 3D MAPs to uncover cellular mechanisms that may regulate this process.


Subject(s)
Chondrocytes/physiology , Growth Differentiation Factor 5/metabolism , Growth Plate/growth & development , Animals , Animals, Newborn , Cell Differentiation , Cell Proliferation , Embryo, Mammalian , Female , Growth Differentiation Factor 5/economics , Growth Plate/cytology , Growth Plate/diagnostic imaging , Imaging, Three-Dimensional , Intravital Microscopy , Mice, Knockout , Models, Animal , Tibia/cytology , Tibia/drug effects , Tibia/growth & development , X-Ray Microtomography
14.
Nat Commun ; 12(1): 4161, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34230488

ABSTRACT

Given the pleiotropic nature of coding sequences and that many loci exhibit multiple disease associations, it is within non-coding sequence that disease-specificity likely exists. Here, we focus on joint disorders, finding among replicated loci, that GDF5 exhibits over twenty distinct associations, and we identify causal variants for two of its strongest associations, hip dysplasia and knee osteoarthritis. By mapping regulatory regions in joint chondrocytes, we pinpoint two variants (rs4911178; rs6060369), on the same risk haplotype, which reside in anatomical site-specific enhancers. We show that both variants have clinical relevance, impacting disease by altering morphology. By modeling each variant in humanized mice, we observe joint-specific response, correlating with GDF5 expression. Thus, we uncouple separate regulatory variants on a common risk haplotype that cause joint-specific disease. By broadening our perspective, we finally find that patterns of modularity at GDF5 are also found at over three-quarters of loci with multiple GWAS disease associations.


Subject(s)
Exons , Hip Dislocation/genetics , Hip Dislocation/metabolism , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/metabolism , Animals , Chondrocytes , Disease Models, Animal , Gene Expression Regulation , Genetic Predisposition to Disease , Genome-Wide Association Study , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Humans , Mice , Phenotype , Regulatory Sequences, Nucleic Acid
15.
Int J Rheum Dis ; 24(5): 694-700, 2021 May.
Article in English | MEDLINE | ID: mdl-33861510

ABSTRACT

AIM: Osteoarthritis (OA) is the most common chronic joint disorder, resulting from the breakdown of joint cartilage. It occurs in the knees, hands, and hips, leading to pain, stiffness, inflammation, and swelling. METHODS: In this study, 100 hand and knee OA patients, meeting the American College of Rheumatology criteria were included in the case group, and 100 healthy individuals were allocated to the control group. Blood samples were collected from the participants. After DNA extraction, genotyping was carried out for GDF5 rs143383 C/T polymorphism by allele-specific polymerase chain reaction (ASPCR) and for D-repeat alleles of asporin (ASPN) by conventional PCR assay. RESULTS: The results showed that the frequency of the D14 allele of ASPN was significantly higher than other alleles in the case group (P = .0001). Also, the frequency of the D14 allele among women was significantly higher than in men (P = .004). Moreover, the frequency of the TT allele in GDF5 rs143383 C/T polymorphism was significantly higher than the CC and CT alleles in the case group, compared with the control group (P = .001). A significant difference was found between the TT allele and other alleles in female and male patients compared with the control group (P = .02). CONCLUSIONS: The D14 allele of the ASPN gene and TT allele of the GDF5 gene (rs143383 + 104T/C) are associated with hand and knee OA in the Kurdish population, indicating that these alleles could be risk factors for OA, at least in our populations.


Subject(s)
Cartilage, Articular/physiopathology , Extracellular Matrix Proteins/genetics , Genetic Predisposition to Disease/genetics , Growth Differentiation Factor 5/genetics , Hand/physiopathology , Osteoarthritis, Knee/genetics , Polymorphism, Genetic/genetics , Aged , Alleles , Case-Control Studies , Extracellular Matrix Proteins/metabolism , Female , Genetic Predisposition to Disease/ethnology , Growth Differentiation Factor 5/metabolism , Humans , Iran/epidemiology , Male , Middle Aged , Osteoarthritis, Knee/epidemiology , Polymorphism, Single Nucleotide
16.
Orthop Surg ; 13(3): 734-741, 2021 May.
Article in English | MEDLINE | ID: mdl-33817978

ABSTRACT

Intervertebral disc degeneration (IDD) is widely recognized as the main cause of low back pain, which leads to disability in aging populations and induces great losses both socially and economically worldwide. Unfortunately, current treatments for IDD are aimed at relieving symptoms instead of preserving disc structure and function. Researchers are forged to find new promising biological therapeutics to stop, and even reverse, IVD degeneration. Recently, the injection of growth factors has been shown to be a promising biological therapy for IDD. A number of growth factors have been investigated to modulate the synthesis of the extracellular matrix (ECM) through a variety of pathogenetic biological mechanisms, including suppressing inflammatory process and down-regulating degrading enzymes. However, growth factors, including Transforming Growth Factor-ß (TGF-ß), Fibroblast Growth Factor (FGF), and Insulin-like Growth Factor-1 (IGF-1), may induce unwanted blood vessel in-growth, which accelerates the process of IDD. On the contrary, studies have demonstrated that injection of GDF-5 into the intervertebral disc of mice can effectively alleviate the degeneration of the intervertebral disc, which elicits their response via BMPRII and will not induce blood vessel in-growth. This finding suggests that GDF-5 is more suitable for use in IDD treatment compared with the three other growth factors. Substantial evidence has suggested that GDF-5 may maintain the structure and function of the intervertebral disc (IVD). GDF-5 plays an important role in IDD and is a very promising therapeutic agent for IDD. This review is focused on the mechanisms and functions of GDF-5 in IDD.


Subject(s)
Growth Differentiation Factor 5/metabolism , Intervertebral Disc Degeneration , Humans
17.
Cell Prolif ; 54(3): e12998, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33522652

ABSTRACT

Growth differentiation factor 5 (GDF-5) is essential for cartilage development and homeostasis. The expression and function of GDF-5 are highly associated with the pathogenesis of osteoarthritis (OA). OA, characterized by progressive degeneration of joint, particularly in cartilage, causes severe social burden. However, there is no effective approach to reverse the progression of this disease. Over the past decades, extensive studies have demonstrated the protective effects of GDF-5 against cartilage degeneration and defects. Here, we summarize the current literature describing the role of GDF-5 in development of cartilage and joints, and the association between the GDF-5 gene polymorphisms and OA susceptibility. We also shed light on the protective effects of GDF-5 against OA in terms of direct GDF-5 supplementation and modulation of the GDF-5-related signalling. Finally, we discuss the current limitations in the application of GDF-5 for the clinical treatment of OA. This review provides a comprehensive insight into the role of GDF-5 in cartilage and emphasizes GDF-5 as a potential therapeutic candidate in OA.


Subject(s)
Chondrocytes/metabolism , Growth Differentiation Factor 5/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Animals , Cartilage Diseases/drug therapy , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cell Differentiation/physiology , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/pharmacology , Humans
18.
J Cell Mol Med ; 25(4): 1939-1948, 2021 02.
Article in English | MEDLINE | ID: mdl-33369147

ABSTRACT

The migration of epidermal stem cells (EpSCs) is critical for wound re-epithelization and wound healing. Recently, growth/differentiation factor-5 (GDF-5) was discovered to have multiple biological effects on wound healing; however, its role in EpSCs remains unclear. In this work, recombinant mouse GDF-5 (rmGDF-5) was found via live imaging in vitro to facilitate the migration of mouse EpSCs in a wound-scratch model. Western blot and real-time PCR assays demonstrated that the expression levels of RhoA and matrix metalloproteinase-9 (MMP9) were correlated with rmGDF-5 concentration. Furthermore, we found that rmGDF-5 stimulated mouse EpSC migration in vitro by regulating MMP9 expression at the mRNA and protein levels through the RhoA signalling pathway. Moreover, in a deep partial-thickness scald mouse model in vivo, GDF-5 was confirmed to promote EpSC migration and MMP9 expression via RhoA, as evidenced by the tracking of cells labelled with 5-bromo-2-deoxyuridine (BrdU). The current study showed that rmGDF-5 can promote mouse EpSC migration in vitro and in vivo and that GDF-5 can trigger the migration of EpSCs via RhoA-MMP9 signalling.


Subject(s)
Cell Movement/genetics , Epidermal Cells/metabolism , Growth Differentiation Factor 5/metabolism , Matrix Metalloproteinase 9/metabolism , Stem Cells/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Gene Expression , Gene Expression Regulation , Matrix Metalloproteinase 9/genetics , Mice , RNA, Messenger/genetics
19.
Dev Biol ; 470: 136-146, 2021 02.
Article in English | MEDLINE | ID: mdl-33217406

ABSTRACT

The development of joints in the mammalian skeleton depends on the precise regulation of multiple interacting signaling pathways including the bone morphogenetic protein (BMP) pathway, a key regulator of joint development, digit patterning, skeletal growth, and chondrogenesis. Mutations in the BMP receptor ACVR1 cause the rare genetic disease fibrodysplasia ossificans progressiva (FOP) in which extensive and progressive extra-skeletal bone forms in soft connective tissues after birth. These mutations, which enhance BMP-pSmad1/5 pathway activity to induce ectopic bone, also affect skeletal development. FOP can be diagnosed at birth by symmetric, characteristic malformations of the great toes (first digits) that are associated with decreased joint mobility, shortened digit length, and absent, fused, and/or malformed phalanges. To elucidate the role of ACVR1-mediated BMP signaling in digit skeletal development, we used an Acvr1R206H/+;Prrx1-Cre knock-in mouse model that mimics the first digit phenotype of human FOP. We have determined that the effects of increased Acvr1-mediated signaling by the Acvr1R206H mutation are not limited to the first digit but alter BMP signaling, Gdf5+ joint progenitor cell localization, and joint development in a manner that differently affects individual digits during embryogenesis. The Acvr1R206H mutation leads to delayed and disrupted joint specification and cleavage in the digits and alters the development of cartilage and endochondral ossification at sites of joint morphogenesis. These findings demonstrate an important role for ACVR1-mediated BMP signaling in the regulation of joint and skeletal formation, show a direct link between failure to restrict BMP signaling in the digit joint interzone and failure of joint cleavage at the presumptive interzone, and implicate impaired, digit-specific joint development as the proximal cause of digit malformation in FOP.


Subject(s)
Activin Receptors, Type I/metabolism , Bone Morphogenetic Proteins/metabolism , Joints/embryology , Myositis Ossificans/embryology , Myositis Ossificans/metabolism , Toes/embryology , Animals , Body Patterning , Chondrogenesis , Disease Models, Animal , Forelimb/abnormalities , Forelimb/embryology , Growth Differentiation Factor 5/metabolism , Growth Plate/embryology , Hindlimb/abnormalities , Hindlimb/embryology , Joints/abnormalities , Joints/metabolism , Mice , Osteogenesis , Signal Transduction , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Stem Cells/physiology , Toes/abnormalities
20.
Eur Rev Med Pharmacol Sci ; 24(21): 10975-10983, 2020 11.
Article in English | MEDLINE | ID: mdl-33215411

ABSTRACT

OBJECTIVE: Destruction of extracellular matrix (ECM), especially collagen II and aggrecan, is an essential feature of intervertebral disc degeneration (IDD). This project planned to elucidate the role of cartilage-derived morphogenetic protein-1 (CDMP-1) in the collagen II and aggrecan synthesis of nucleus pulposus (NP) cells under the IL-1ß induced degeneration. PATIENTS AND METHODS: We cultured human primary NP cells in the different concentrations of IL-1ß medium and analyzed the CDMP-1 level. Recombinant human CDMP-1 protein was used to co-culture with IL-1ß to investigate its effects on collagen II and aggrecan synthesis of NP cells. Additionally, the bone morphogenetic protein type IB receptor (ALK6) gene silenced and upregulated NP cells were used to evaluate the function of ALK6 in the CDMP-1 treated NP cells. Collagen II, aggrecan, MMP9, MMP13, and TIMP4 expression level were analyzed to assess the ECM stability of NP cells. RESULTS: CDMP-1 gene expression decreased in the IL-1ß treated NP cells with a dose-dependent. Appropriate CDMP-1 protein supplement contributed to the collagen II and aggrecan production, the suppression of MMP9 and MMP13, and the upregulation of TIMP4. However, the silencing of ALK6 rejected the positive function of CDMP-1 on the collagen II and aggrecan; on the contrary, ALK6 upregulation magnified the CDMP-1 induced collagen II and aggrecan production. CONCLUSIONS: CDMP-1 is efficient in promoting the collagen II and aggrecan synthesis of NP cells, which is probably based on the mediation of ALK6.


Subject(s)
Aggrecans/biosynthesis , Bone Morphogenetic Protein Receptors, Type I/metabolism , Collagen Type II/biosynthesis , Growth Differentiation Factor 5/metabolism , Nucleus Pulposus/metabolism , Adult , Aggrecans/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Cells, Cultured , Collagen Type II/genetics , Female , Growth Differentiation Factor 5/genetics , Humans , Male , Nucleus Pulposus/cytology
SELECTION OF CITATIONS
SEARCH DETAIL