Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Curr Opin Rheumatol ; 35(1): 55-60, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36350386

ABSTRACT

PURPOSE OF REVIEW: To review recent studies using 'Omics' approaches (genomics, proteomics, metabolomics, single cell analyses) in patient populations and animal models of osteoarthritis (OA), with the goal of identifying disease-modifying mechanisms that could serve as therapeutic and diagnostic targets. RECENT FINDINGS: The number of genes, pathways and molecules with potential roles in OA pathogenesis has grown substantially over the last 18 months. Studies have expanded from their traditional focus on cartilage and gene expression to other joint tissues, proteins and metabolites. Single cell approaches provide unprecedented resolution and exciting insights into the heterogeneity of cellular activities in OA. Functional validation and investigation of underlying mechanisms in animal models of OA, in particular genetically engineered mice, link Omics findings to pathophysiology and potential therapeutic applications. SUMMARY: Although great progress has been made in the use of Omics approaches to OA, in both animal models and patient samples, much work remains to be done. In addition to filling gaps in data sets not yet existing, integration of data from the various approaches, mechanistic investigations, and linkage of Omics data to patient stratification remain significant challenges.


Subject(s)
Osteoarthritis , Mice , Animals , Humans , Osteoarthritis/genetics , Osteoarthritis/therapy , Osteoarthritis/metabolism , Proteomics , Genomics , Metabolomics , Proteins/metabolism
2.
Int J Obes (Lond) ; 46(4): 726-738, 2022 04.
Article in English | MEDLINE | ID: mdl-34897286

ABSTRACT

BACKGROUND: Pannexin 3 (PANX3) is a channel-forming glycoprotein that enables nutrient-induced inflammation in vitro, and genetic linkage data suggest that it regulates body mass index. Here, we characterized inflammatory and metabolic parameters in global Panx3 knockout (KO) mice in the context of forced treadmill running (FEX) and high-fat diet (HFD). METHODS: C57BL/6N (WT) and KO mice were randomized to either a FEX running protocol or no running (SED) from 24 until 30 weeks of age. Body weight was measured biweekly, and body composition was measured at 24 and 30 weeks of age. Male WT and KO mice were fed a HFD from 12 to 28 weeks of age. Metabolic organs were analyzed for a panel of inflammatory markers and PANX3 expression. RESULTS: In females there were no significant differences in body composition between genotypes, which could be due to the lack of PANX3 expression in female white adipose tissue, while male KOs fed a chow diet had lower body weight and lower fat mass at 24 and 30 weeks of age, which was reduced to the same extent as 6 weeks of FEX in WT mice. In addition, male KO mice exhibited significantly lower expression of multiple pro-inflammatory genes in white adipose tissue compared to WT mice. While on a HFD body weight differences were insignificant, multiple inflammatory genes were significantly different in quadriceps muscle and white adipose tissue resulting in a more anti-inflammatory phenotype in KO mice compared to WT. The lower fat mass in male KO mice may be due to significantly fewer adipocytes in their subcutaneous fat compared to WT mice. Mechanistically, adipose stromal cells (ASCs) cultured from KO mice grow significantly slower than WT ASCs. CONCLUSION: PANX3 is expressed in male adult mouse adipose tissue and may regulate adipocyte numbers, influencing fat accumulation and inflammation.


Subject(s)
Adipose Tissue , Obesity , Adipose Tissue/metabolism , Animals , Body Weight/physiology , Diet, High-Fat , Female , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism
3.
Int J Mol Sci ; 22(3)2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33499145

ABSTRACT

Pannexin 3 (Panx3) is a mechanosensitive, channel-forming glycoprotein implicated in the progression of post-traumatic osteoarthritis. Despite evidence for Panx3 expression in the intervertebral disc (IVD), its function in this cartilaginous joint structure remained unknown. Using Panx3 knockout mice, this study investigated the role of Panx3 in age-associated IVD degeneration and degeneration induced by annulus fibrosus (AF) needle puncture. Loss of Panx3 did not significantly impact the progression of age-associated histopathological IVD degeneration; however, loss of Panx3 was associated with decreased gene expression of Acan, Col1a1, Mmp13 and Runx2 and altered localization of COLX in the IVD at 19 months-of-age. Following IVD injury in the caudal spine, histological analysis of wild-type mice revealed clusters of hypertrophic cells in the AF associated with increased pericellular proteoglycan accumulation, disruptions in lamellar organization and increased lamellar thickness. In Panx3 knockout mice, hypertrophic AF cells were rarely detected and AF structure was largely preserved post-injury. Interestingly, uninjured IVDs adjacent to the site of injury more frequently showed evidence of early nucleus pulposus degeneration in Panx3 knockout mice but remained healthy in wild-type mice. These findings suggest a role for Panx3 in mediating the adaptive cellular responses to altered mechanical stress in the IVD, which may buffer aberrant loads transferred to adjacent motion segments.


Subject(s)
Annulus Fibrosus/injuries , Connexins/metabolism , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc/injuries , Nucleus Pulposus/pathology , Proteoglycans/metabolism , Aging , Animals , Annulus Fibrosus/pathology , Disease Models, Animal , Gene Expression Regulation , Genotype , Intervertebral Disc/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Punctures , Stress, Mechanical
4.
Br J Sports Med ; 54(13): 771-775, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31848152

ABSTRACT

OBJECTIVES: We systemically reviewed published studies that evaluated aerobic exercise interventions in patients with knee osteoarthritis (OA) to: (1) report the frequency, intensity, type and time (FITT) of exercise prescriptions and (2) quantify the changes in markers of cardiovascular health and systemic inflammation. DATA SOURCES: PubMed, CINAHL, Scopus; inception to January 2019. ELIGIBILITY CRITERIA: Randomised clinical trials (RCT), cohort studies, case series. DESIGN: We summarised exercise prescriptions for all studies and calculated effect sizes with 95% CIs for between-group (RCTs that compared exercise and control groups) and within-group (pre-post exercise) differences in aerobic capacity (VO2), heart rate (HR), systolic blood pressure (SBP), diastolic blood pressure (DBP) and inflammatory markers (interleukin-6 (IL-6), tumour necrosis factor-alpha). We pooled results where possible using random effects models. RESULTS: Interventions from 49 studies were summarised; 8% (4/49) met all FITT guidelines; 16% (8/49) met all or most FITT guidelines. Fourteen studies (10 RCTs) reported at least one marker of cardiovascular health or systemic inflammation. Mean differences (95% CI) indicated a small to moderate increase in VO2 (0.84 mL/min/kg; 95% CI 0.37 to 1.31), decrease in HR (-3.56 beats per minute; 95% CI -5.60 to -1.52) and DBP (-4.10 mm Hg; 95% CI -4.82 to -3.38) and no change in SBP (-0.36 mm Hg; 95% CI -3.88 to 3.16) and IL-6 (0.37 pg/mL; 95% CI -0.11 to 0.85). Within-group differences were also small to moderate. CONCLUSIONS: In studies of aerobic exercise in patients with knee OA, very few interventions met guideline-recommended dose; there were small to moderate changes in markers of cardiovascular health and no decrease in markers of systemic inflammation. These findings question whether aerobic exercise is being used to its full potential in patients with knee OA. PROSPERO REGISTRATION NUMBER: CRD42018087859.


Subject(s)
Cardiorespiratory Fitness/physiology , Exercise Therapy/methods , Inflammation/physiopathology , Osteoarthritis, Knee/physiopathology , Osteoarthritis, Knee/rehabilitation , Exercise , Exercise Tolerance , Humans
6.
Int J Mol Sci ; 20(20)2019 Oct 20.
Article in English | MEDLINE | ID: mdl-31635173

ABSTRACT

Longitudinal bone growth occurs through endochondral ossification (EO), controlled by various signaling molecules. Retinoid X Receptor (RXR) is a nuclear receptor with important roles in cell death, development, and metabolism. However, little is known about its role in EO. In this study, the agonist SR11237 was used to evaluate RXR activation in EO. Rats given SR11237 from post-natal day 5 to post-natal day 15 were harvested for micro-computed tomography (microCT) scanning and histology. In parallel, newborn CD1 mouse tibiae were cultured with increasing concentrations of SR11237 for histological and whole-mount evaluation. RXR agonist-treated rats had shorter long bones than the controls and developed dysmorphia of the growth plate. Cells invading the calcified and dysmorphic growth plate appeared pre-hypertrophic in size and shape, in correspondence with p57 immunostaining. Additionally, SOX9-positive cells were found surrounding the calcified tissue. The epiphysis of SR11237-treated bones showed increased TRAP staining and additional TUNEL staining at the osteo-chondral junction. MicroCT revealed morphological disorganization in the long bones of the treated animals. This study suggests that stimulation of RXR causes irregular ossification, premature closure of the growth plate, and disrupted long bone growth in rodent models.


Subject(s)
Benzoates/pharmacology , Bone Development/drug effects , Gene Expression Regulation, Developmental/drug effects , Growth Plate/drug effects , Retinoid X Receptors/agonists , Retinoids/pharmacology , Animals , Female , Male , Mice , Rats , Rats, Sprague-Dawley , Signal Transduction
7.
Curr Opin Rheumatol ; 29(1): 96-102, 2017 01.
Article in English | MEDLINE | ID: mdl-27906752

ABSTRACT

PURPOSE OF REVIEW: Despite the tremendous individual suffering and socioeconomic burden caused by osteoarthritis, there are currently no effective disease-modifying treatment options. This is in part because of our incomplete understanding of osteoarthritis disease mechanism. This review summarizes recent developments in therapeutic targets identified from surgical animal models of osteoarthritis that provide novel insight into osteoarthritis pathology and possess potential for progression into preclinical studies. RECENT FINDINGS: Several candidate pathways and processes that have been identified include chondrocyte autophagy, growth factor signaling, inflammation, and nociceptive signaling. Major strategies that possess therapeutic potential at the cellular level include inhibiting autophagy suppression and decreasing reactive oxygen species (ROS) production. Cartilage anabolism and prevention of cartilage degradation has been shown to result from growth factor signaling modulation, such as TGF-ß, TGF-α, and FGF; however, the results are context-dependent and require further investigation. Pain assessment studies in rodent surgical models have demonstrated potential in employing anti-NGF strategies for minimizing osteoarthritis-associated pain. SUMMARY: Studies of potential therapeutic targets in osteoarthritis using animal surgical models are helping to elucidate osteoarthritis pathology and propel therapeutics development. Further studies should continue to elucidate pathological mechanisms and therapeutic targets in various joint tissues to improve overall joint health.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Experimental/drug therapy , Molecular Targeted Therapy/methods , Osteoarthritis/drug therapy , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Autophagy/drug effects , Autophagy/physiology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/pathology , Humans , Inflammation Mediators/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
8.
Oncologist ; 22(7): 811-822, 2017 07.
Article in English | MEDLINE | ID: mdl-28526718

ABSTRACT

Squamous cell carcinoma of the head and neck (SCCHN) is the sixth most common cancer worldwide. It has recently been appreciated that human papillomavirus (HPV) status (or p16 status, which is a frequently used surrogate for HPV status) is prognostic for oropharyngeal SCCHN. Here, we review and contextualize existing p16 and HPV data, focusing on the cetuximab registration trials in previously untreated, locoregionally advanced, nonmetastatic SCCHN (LA SCCHN) and in recurrent and/or metastatic SCCHN (R/M SCCHN): the IMCL-9815 and EXTREME clinical trials, respectively. Taken together, the available data suggest that, while p16 and HPV are prognostic biomarkers in patients with LA SCCHN and R/M SCCHN, it could not be shown that they are predictive for the outcomes of the described cetuximab-containing trial regimens. Consequently, although HPV status provides prognostic information, it is not shown to predict therapy response, and so is not helpful for assigning first-line therapy in patients with SCCHN. In addition, we discuss assays currently used to assess p16 and HPV status, as well as the differentiation between these two biomarkers. Ultimately, we believe HPV E6/E7 polymerase chain reaction-based mRNA testing may represent the most informative technique for assessing HPV status in patients with SCCHN. While p16 is a valid surrogate for HPV status in oropharyngeal carcinoma (OPC), there is a higher risk of discordance between p16 and HPV status in non-OPC SCCHN. Collectively, these discussions hold key implications for the clinical management of SCCHN. IMPLICATIONS FOR PRACTICE: Human papillomavirus (HPV) status (or its commonly utilized surrogate p16) is a known prognostic biomarker in oropharyngeal squamous-cell carcinoma of the head and neck (SCCHN). We evaluated implications of the available evidence, including cetuximab registration trials in previously untreated locoregionally advanced (LA) SCCHN and recurrent and/or metastatic (R/M) SCCHN. We conclude that, although p16 and HPV are prognostic biomarkers for both LA and R/M SCCHN, they have not been shown to be predictive of response to the described cetuximab-containing regimens for either indication. Thus, current evidence suggests that benefits of cetuximab are observed in both p16-/HPV-positive and -negative SCCHN.


Subject(s)
Carcinoma, Squamous Cell/virology , Cetuximab/therapeutic use , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Head and Neck Neoplasms/virology , Papillomavirus Infections/complications , Antineoplastic Agents, Immunological , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Humans , Prognosis
9.
Connect Tissue Res ; 58(1): 37-48, 2017 01.
Article in English | MEDLINE | ID: mdl-27028588

ABSTRACT

Osteoarthritis (OA) is a major clinical problem across the world, in part due to the lack of disease-modifying drugs resulting, to a significant degree, from our incomplete understanding of the underlying molecular mechanisms of the disease. Emerging evidence points to a role of epigenetics in the pathogenesis of OA, but research in this area is still in its early stages. In order to summarize current knowledge and to facilitate the potential coordination of future research activities, the first international workshop on the epigenetics of OA was held in Amsterdam in October 2015. Recent findings on DNA methylation and hydroxymethylation, histone modifications, noncoding RNAs, and other epigenetic mechanisms were presented and discussed. The workshop demonstrated the advantage of bringing together those working in this nascent field and highlights from the event are summarized in this report in the form of summaries from invited speakers and organizers.


Subject(s)
Epigenomics , Osteoarthritis , Animals , Congresses as Topic , Denmark , Humans
10.
Proc Natl Acad Sci U S A ; 111(7): 2590-5, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24550287

ABSTRACT

A deficiency of mitogen-inducible gene-6 (Mig-6) in mice leads to the development of an early-onset, osteoarthritis (OA)-like disorder in multiple synovial joints, underlying its importance in maintaining joint homeostasis. Here we determined what joint tissues Mig-6 is expressed in and what role chondrocytes play in the Mig-6-deficient OA-like disorder. A Mig-6/lacZ reporter mouse strain expressing ß-galactosidase under the control of the Mig-6 gene promoter was generated to determine Mig-6 expression in joint tissues. By ß-galactosidase staining, we demonstrated that Mig-6 was uniquely expressed in the cells across the entire surface of the synovial joint cavity, including chondrocytes in the superficial zone of articular cartilage and in the meniscus, as well as synovial lining cells. By crossing Mig-6-floxed mice to Col2a1-Cre transgenic mice, to generate cartilage-specific deletion of Mig-6, we demonstrated that deficiency of Mig-6 in the chondrocytes results in a joint phenotype that only partially recapitulates the OA-like disorder of the Mig-6-deficient mice: Ubiquitous deletion of Mig-6 led to the OA-like disorder in multiple joints, whereas cartilage-specific deletion affected the knees but rarely other joints. Furthermore, chondrocytes with Mig-6 deficiency showed excessive proliferative activities along with enhanced EGF receptor signaling in the articular cartilage and in the abnormally formed osteophytes. Our findings provide insight into the crucial requirement for Mig-6 in maintaining joint homeostasis and in regulating chondrocyte activities in the synovial joints. Our data also suggest that other cell types are required for fully developing the Mig-6-deficient OA-like disorder.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/physiology , Intracellular Signaling Peptides and Proteins/deficiency , Osteoarthritis/genetics , Animals , Cell Proliferation , Genetic Vectors , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Osteoarthritis/pathology , Reverse Transcriptase Polymerase Chain Reaction , beta-Galactosidase
11.
J Anat ; 228(5): 746-56, 2016 May.
Article in English | MEDLINE | ID: mdl-26749194

ABSTRACT

Pannexins form single-membrane channels that allow passage of small molecules between the intracellular and extracellular compartments. Of the three pannexin family members, Pannexin3 (Panx3) is the least studied but is highly expressed in skeletal tissues and is thought to play a role in the regulation of chondrocyte and osteoblast proliferation and differentiation. The purpose of our study is to closely examine the in vivo effects of Panx3 ablation on long bone morphology using micro-computed tomography. Using Panx3 knockout (KO) and wildtype (WT) adult mice, we measured and compared aspects of phenotypic shape, bone mineral density (BMD), cross-sectional geometric properties of right femora and humeri, and lean mass. We found that KO mice have absolutely and relatively shorter diaphyseal shafts compared with WT mice, and relatively larger areas of muscle attachment sites. No differences in BMD or lean mass were found between WT and KO mice. Interestingly, KO mice had more robust femora and humeri compared with WT mice when assessed in cross-section at the midshaft. Our results clearly show that Panx3 ablation produces phenotypic effects in mouse femora and humeri, and support the premise that Panx3 has a role in regulating long bone growth and development.


Subject(s)
Connexins/deficiency , Femur/anatomy & histology , Humerus/anatomy & histology , Animals , Bone Density/physiology , Cross-Sectional Studies , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , X-Ray Microtomography
12.
FASEB J ; 29(10): 4107-21, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26092928

ABSTRACT

We investigated the role of periostin, an extracellular matrix protein, in the pathophysiology of osteoarthritis (OA). In OA, dysregulated gene expression and phenotypic changes in articular chondrocytes culminate in progressive loss of cartilage from the joint surface. The molecular mechanisms underlying this process are poorly understood. We examined periostin expression by immunohistochemical analysis of lesional and nonlesional cartilage from human and rodent OA knee cartilage. In addition, we used small interfering (si)RNA and adenovirus transduction of chondrocytes to knock down and up-regulate periostin levels, respectively, and analyzed its effect on matrix metalloproteinase (MMP)-13, a disintegrin and MMP with thrombospondin motifs (ADAMTS)-4, and type II collagen expression. We found high periostin levels in human and rodent OA cartilage. Periostin increased MMP-13 expression dose [1-10 µg/ml (EC50 0.5-1 µg/ml)] and time (24-72 h) dependently, significantly enhanced expression of ADAMTS4 mRNA, and promoted cartilage degeneration through collagen and proteoglycan degradation. Periostin induction of MMP-13 expression was inhibited by CCT031374 hydrobromide, an inhibitor of the canonical Wnt/ß-catenin signaling pathway. In addition, siRNA-mediated knockdown of endogenous periostin blocked constitutive MMP-13 expression. These findings implicate periostin as a catabolic protein that promotes cartilage degeneration in OA by up-regulating MMP-13 through canonical Wnt signaling.


Subject(s)
Cartilage, Articular/metabolism , Cell Adhesion Molecules/metabolism , Extracellular Matrix/metabolism , Matrix Metalloproteinase 13/metabolism , Osteoarthritis/metabolism , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS4 Protein , Aged , Aged, 80 and over , Animals , Blotting, Western , Cattle , Cell Adhesion Molecules/genetics , Cells, Cultured , Chondrocytes/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Matrix Metalloproteinase 13/genetics , Mice, Inbred C57BL , Middle Aged , Osteoarthritis/genetics , Procollagen N-Endopeptidase/genetics , Procollagen N-Endopeptidase/metabolism , RNA Interference , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
13.
Biochim Biophys Acta ; 1840(7): 2112-22, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24637075

ABSTRACT

BACKGROUND: Choline kinase has three isoforms encoded by the genes Chka and Chkb. Inactivation of Chka in mice results in embryonic lethality, whereas Chkb(-/-) mice display neonatal forelimb bone deformations. METHODS: To understand the mechanisms underlying the bone deformations, we compared the biology and biochemistry of bone formation from embryonic to young adult wild-type (WT) and Chkb(-/-) mice. RESULTS: The deformations are specific to the radius and ulna during the late embryonic stage. The radius and ulna of Chkb(-/-) mice display expanded hypertrophic zones, unorganized proliferative columns in their growth plates, and delayed formation of primary ossification centers. The differentiation of chondrocytes of Chkb(-/-) mice was impaired, as was chondrocyte proliferation and expression of matrix metalloproteinases 9 and 13. In chondrocytes from Chkb(-/-) mice, phosphatidylcholine was slightly lower than in WT mice whereas the amount of phosphocholine was decreased by approximately 75%. In addition, the radius and ulna from Chkb(-/-) mice contained fewer osteoclasts along the cartilage/bone interface. CONCLUSIONS: Chkb has a critical role in the normal embryogenic formation of the radius and ulna in mice. GENERAL SIGNIFICANCE: Our data indicate that choline kinase beta plays an important role in endochondral bone formation by modulating growth plate physiology.


Subject(s)
Cell Differentiation/genetics , Choline Kinase/genetics , Growth Plate/growth & development , Osteogenesis/genetics , Animals , Choline Kinase/metabolism , Chondrocytes/enzymology , Embryo, Mammalian/enzymology , Embryonic Development/genetics , Forelimb/embryology , Forelimb/enzymology , Forelimb/growth & development , Growth Plate/enzymology , Humans , Mice , Mice, Knockout , Phosphatidylcholines/metabolism
14.
Hum Mol Genet ; 22(24): 5015-25, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23892236

ABSTRACT

ATR-X syndrome is a rare genetic disorder caused by mutations in the ATRX gene. Affected individuals are cognitively impaired and display a variety of developmental abnormalities, including skeletal deformities. To investigate the function of ATRX during skeletal development, we selectively deleted the gene in the developing forelimb mesenchyme of mice. The absence of ATRX in the limb mesenchyme resulted in shorter digits, or brachydactyly, a defect also observed in a subset of ATR-X patients. This phenotype persisted until adulthood, causing reduced grip strength and altered gait in mutant mice. Examination of the embryonic ATRX-null forelimbs revealed a significant increase in apoptotic cell death, which could explain the reduced digit length. In addition, staining for the DNA damage markers γ-histone 2A family member X (γ-H2AX) and 53BP1 demonstrated a significant increase in the number of cells with DNA damage in the embryonic ATRX-null forepaw. Strikingly, only one large bright DNA damage event was observed per nucleus in proliferating cells. These large γ-H2AX foci were located in close proximity to the nuclear lamina and remained largely unresolved after cell differentiation. In addition, ATRX-depleted forelimb mesenchymal cells did not exhibit hypersensitivity to DNA fork-stalling compounds, suggesting that the nature as well as the response to DNA damage incurred by loss of ATRX in the developing limb fundamentally differs from other tissues. Our data suggest that DNA damage-induced apoptosis is a novel cellular mechanism underlying brachydactyly that might be relevant to additional skeletal syndromes.


Subject(s)
Brachydactyly/genetics , DNA Helicases/genetics , Forelimb/abnormalities , Mesoderm/metabolism , Nuclear Proteins/genetics , Animals , Brachydactyly/metabolism , Cell Death/genetics , Chondrocytes/metabolism , DNA Helicases/deficiency , DNA Helicases/metabolism , Disease Models, Animal , Female , Forelimb/embryology , Forelimb/physiopathology , Genetic Association Studies , Histones/genetics , Histones/metabolism , Hydroxyurea/pharmacology , Limb Buds/embryology , Limb Buds/metabolism , Male , Mesoderm/drug effects , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , Phenotype , X-linked Nuclear Protein
15.
Curr Rheumatol Rep ; 17(8): 50, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26113010

ABSTRACT

Osteoarthritis causes tremendous individual suffering and staggering societal costs, but due to our limited understanding of the underlying molecular and cellular mechanisms, our avenues for treating this disease are very restricted. Recent years have seen a drastic increase in the use of genetically modified mice to characterize the pathophysiology of osteoarthritis. Many new players and mechanisms driving osteoarthritis pathogenesis have been elucidated, some of which might be strong candidates as therapeutic targets for the human disease. The current review summarizes key findings (selected subjectively by the authors) from mouse osteoarthritis studies over recent years.


Subject(s)
Arthritis, Experimental/pathology , Osteoarthritis/pathology , Animals , Arthritis, Experimental/physiopathology , Autophagy/physiology , Bone Remodeling/physiology , Cartilage, Articular/pathology , Homeostasis/physiology , Mice , Osteoarthritis/physiopathology , Osteophyte/pathology , Osteophyte/physiopathology , Proteolysis , Synovial Membrane/pathology
16.
Biochem J ; 464(3): 355-64, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25310312

ABSTRACT

Biomineralization is a complex process in the development of mineralized tissues such as bone and pathological calcifications such as atherosclerotic plaques, kidney stones and gout. Osteopontin (OPN), an anionic phosphoprotein, is expressed in mineralizing tissues and has previously been demonstrated to be a potent inhibitor of hydroxyapatite formation. The OPN-deficient (Opn-/-) mouse displays a hypermineralized bone phenotype starting at 12 weeks postnatally. By isolating and culturing Opn-/- and wild-type (WT) osteoblasts, we sought to determine the role of OPN and two of its functional peptides in osteoblast development and mineralization. Opn-/- osteoblasts had significantly increased mineral deposition relative to their WT counterparts, with no physiologically relevant change in gene expression of osteogenic markers. Supplementation with bovine milk OPN (mOPN) led to a dramatic reduction in mineral deposition by the Opn-/- osteoblasts. Treatment with OPN-derived peptides corresponding to phosphorylated OPN-(220-235) (P3) and non-phosphorylated OPN-(65-80) (OPAR) also rescued the hypermineralization phenotype of Opn-/- osteogenic cultures. Supplementation with mOPN or the OPN-derived peptides did not alter the expression of terminal osteogenic markers. These data suggest that OPN plays an important role in the regulation of biomineralization, but that OPN does not appear to affect osteoblast cell development in vitro.


Subject(s)
Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Osteoblasts/drug effects , Osteogenesis/drug effects , Osteopontin/pharmacology , Animals , Cells, Cultured , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/chemistry , Osteoblasts/physiology , Osteopontin/genetics , Peptide Fragments/pharmacology
17.
Birth Defects Res C Embryo Today ; 102(1): 74-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24677724

ABSTRACT

Most of our bones form through the process of endochondral ossification, which is tightly regulated by the activity of the cartilage growth plate. Chondrocyte maturation through the various stages of growth plate physiology ultimately results in hypertrophy. Chondrocyte hypertrophy is an essential contributor to longitudinal bone growth, but recent data suggest that these cells also play fundamental roles in signaling to other skeletal cells, thus coordinating endochondral ossification. On the other hand, ectopic hypertrophy of articular chondrocytes has been implicated in the pathogenesis of osteoarthritis. Thus, a better understanding of the processes that control chondrocyte hypertrophy in the growth plate as well as in articular cartilage is required for improved management of both skeletal growth disorders and osteoarthritis. This review summarizes recent findings on the regulation of hypertrophic chondrocyte differentiation, the cellular mechanisms involved in hypertrophy, and the role of chondrocyte hypertrophy in skeletal physiology and pathophysiology.


Subject(s)
Bone Development/physiology , Bone Diseases, Developmental/physiopathology , Bone and Bones/cytology , Chondrocytes/cytology , Chondrogenesis/physiology , Animals , Humans , Hypertrophy
18.
Ann Rheum Dis ; 73(8): 1575-84, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23928557

ABSTRACT

OBJECTIVE: To examine the expression of ADAMTS-7 during the progression of osteoarthritis (OA), defining its role in the pathogenesis of OA, and elucidating the molecular events involved. METHODS: ADAMTS-7 expression in cartilage of a rat OA model was assayed using immunohistochemistry. Cartilage-specific ADAMTS-7 transgenic mice and ADAMTS-7 small interfering (si)RNA knockdown mice were generated and used to analyse OA progression in both spontaneous and surgically induced OA models. Cartilage degradation and OA was evaluated using Safranin-O staining, immunohistochemistry, ELISA and western blotting. In addition, mRNA expression of tumour necrosis factor (TNF)-α and metalloproteinases known to be involved in cartilage degeneration in OA was analysed. Furthermore, the transactivation of ADAMTS-7 by TNF-α and its downstream NF-κB signalling was measured using reporter gene assay. RESULTS: ADAMTS-7 expression was elevated during disease progression in the surgically induced rat OA model. Targeted overexpression of ADAMTS-7 in chondrocytes led to chondrodysplasia characterised by short-limbed dwarfism and a delay in endochondral ossification in 'young mice' and a spontaneous OA-like phenotype in 'aged' mice. In addition, overexpression of ADAMTS-7 led to exaggerated breakdown of cartilage and accelerated OA progression, while knockdown of ADAMTS-7 attenuated degradation of cartilage matrix and protected against OA development, in surgically induced OA models. ADAMTS-7 upregulated TNF-α and metalloproteinases associated with OA; in addition, TNF-α induced ADAMTS-7 through NF-κB signalling. CONCLUSIONS: ADAMTS-7 and TNF-α form a positive feedback loop in the regulation of cartilage degradation and OA progression, making them potential molecular targets for prevention and treatment of joint degenerative diseases, including OA.


Subject(s)
ADAM Proteins/immunology , Feedback, Physiological , Osteoarthritis/immunology , Tumor Necrosis Factor-alpha/immunology , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS7 Protein , Aging/immunology , Animals , Cartilage/cytology , Cartilage/immunology , Cartilage/metabolism , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/immunology , Chondrocytes/metabolism , Disease Models, Animal , Disease Progression , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , NF-kappa B/metabolism , Osteoarthritis/metabolism , RNA, Messenger/metabolism , Rats , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
19.
Am J Pathol ; 182(4): 1099-106, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23375622

ABSTRACT

Osteoarthritis (OA) is an age-related progressive degenerative joint disease. Peroxisome proliferator-activated receptor gamma (PPARγ), a transcription factor, is suggested as an attractive therapeutic target to counteract degradative mechanisms associated with OA. Studies suggest that activation of PPARγ by its agonists can reduce the synthesis of OA catabolic and inflammatory factors and the development of cartilage lesions in OA animal models. Because these agonists impart several PPARγ-independent effects, the specific in vivo function of PPARγ in cartilage homeostasis and OA remains largely unknown. Herein, we describe the in vivo role of PPARγ in OA using cartilage-specific PPARγ knockout (KO) mice generated using the Cre-lox system. Adult PPARγ KO mice exhibited a spontaneous OA phenotype associated with enhanced cartilage degradation, hypocellularity, synovial and cartilage fibrosis, synovial inflammation, mononuclear cell influx in the synovium, and increased expression of catabolic factors, including matrix metalloproteinase-13, accompanied by an increase in staining for matrix metalloproteinase-generated aggrecan and type II collagen neoepitopes (VDIPEN and C1-2C). We demonstrate that PPARγ-deficient articular cartilage exhibits elevated expression of the additional catabolic factors hypoxia-inducible factor-2α, syndecan-4, and a disintegrin and metalloproteinase with thrombospondin motifs 5 and of the inflammatory factors cyclooxygenase-2 and inducible nitric oxide synthase. In conclusion, PPARγ is a critical regulator of cartilage health, the lack of which leads to an accelerated spontaneous OA phenotype.


Subject(s)
Aging/metabolism , Cartilage/metabolism , Cartilage/pathology , Osteoarthritis/metabolism , Osteoarthritis/pathology , PPAR gamma/deficiency , Animals , Biomarkers/metabolism , Fibrosis , Gene Deletion , Inflammation/pathology , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Organ Specificity , PPAR gamma/metabolism , Phenotype , Synovial Membrane/metabolism , Synovial Membrane/pathology
20.
Arthritis Rheum ; 65(12): 3153-64, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24022823

ABSTRACT

OBJECTIVE: Interleukin-10 (IL-10) is a pleiotropic immunoregulatory cytokine with a chondroprotective effect that is elevated in cartilage and synovium in patients with osteoarthritis. However, the role of IL-10 during endochondral bone formation and its mechanism of action have not been elucidated. METHODS: IL-10(-/-) mice and IL-10-treated tibial organ cultures were used to study loss and gain of IL-10 functions, respectively, during endochondral bone formation. Primary chondrocytes from the long bones of mouse embryos were cultured with and without IL-10. To assess the role of IL-10 in chondrogenic differentiation, we conducted mesenchymal cell micromass cultures. RESULTS: The lengths of whole skeletons from IL-10(-/-) mice were similar to those of their wild-type littermates, although their skull diameters were smaller. The tibial growth plates of IL-10(-/-) mice showed shortening of the proliferating zone. Treatment with IL-10 significantly increased tibial lengths in organ culture. IL-10 also induced chondrocyte proliferation and hypertrophic differentiation in primary chondrocytes in vitro. Mechanistically, IL-10 activated STAT-3 and the Smad1/5/8 and ERK-1/2 MAP kinase pathways and induced the expression of bone morphogenetic protein 2 (BMP-2) and BMP-6 in primary chondrocytes. Furthermore, the blocking of BMP signaling attenuated the IL-10-mediated induction of cyclin D1 and RUNX-2 in primary chondrocytes and suppressed Alcian blue and alkaline phosphatase staining in mesenchymal cell micromass cultures. CONCLUSION: These results indicate that IL-10 acts as a stimulator of chondrocyte proliferation and chondrogenic or hypertrophic differentiation via activation of the BMP signaling pathway.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Bone and Bones/metabolism , Chondrocytes/metabolism , Interleukin-10/metabolism , Smad Proteins/metabolism , Animals , Bone and Bones/cytology , Bone and Bones/drug effects , Cell Proliferation/drug effects , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrogenesis/drug effects , Chondrogenesis/physiology , Interleukin-10/genetics , Interleukin-10/pharmacology , Mice , Mice, Knockout , Osteogenesis/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL