Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Ann Rheum Dis ; 82(7): 963-973, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36927643

RESUMO

OBJECTIVES: In osteoarthritis, methylation of lysine 79 on histone H3 (H3K79me), a protective epigenetic mechanism, is reduced. Histone methylation levels are dynamically regulated by histone methyltransferases and demethylases. Here, we aimed to identify which histone demethylases regulate H3K79me in cartilage and investigate whether their targeting protects against osteoarthritis. METHODS: We determined histone demethylase expression in human non-osteoarthritis and osteoarthritis cartilage using qPCR. The role of histone demethylase families and subfamilies on H3K79me was interrogated by treatment of human C28/I2 chondrocytes with pharmacological inhibitors, followed by western blot and immunofluorescence. We performed C28/I2 micromasses to evaluate effects on glycosaminoglycans by Alcian blue staining. Changes in H3K79me after destabilisation of the medial meniscus (DMM) in mice were determined by immunohistochemistry. Daminozide, a KDM2/7 subfamily inhibitor, was intra-articularly injected in mice upon DMM. Histone demethylases targeted by daminozide were individually silenced in chondrocytes to dissect their role on H3K79me and osteoarthritis. RESULTS: We documented the expression signature of histone demethylases in human non-osteoarthritis and osteoarthritis articular cartilage. Inhibition of Jumonji-C demethylase family increased H3K79me in human chondrocytes. Blockade of KDM2/7 histone demethylases with daminozide increased H3K79me and glycosaminoglycans. In mouse articular cartilage, H3K79me decayed rapidly upon induction of joint injury. Early and sustained intra-articular treatment with daminozide enhanced H3K79me and exerted protective effects in mice upon DMM. Individual silencing of KDM7A/B demethylases in human chondrocytes demonstrated that KDM7A/B mediate protective effects of daminozide on H3K79me and osteoarthritis. CONCLUSION: Targeting KDM7A/B histone demethylases could be an attractive strategy to protect joints against osteoarthritis.


Assuntos
Cartilagem Articular , Osteoartrite , Humanos , Camundongos , Animais , Histona Desmetilases/metabolismo , Histona Desmetilases/farmacologia , Metilação , Histona Desmetilases com o Domínio Jumonji , Osteoartrite/metabolismo , Condrócitos/metabolismo , Cartilagem Articular/metabolismo , Glicosaminoglicanos
2.
Rheumatology (Oxford) ; 62(1): 457-466, 2022 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-35383365

RESUMO

OBJECTIVES: To investigate whether the deiodinase inhibitor iopanoic acid (IOP) has chondroprotective properties, a mechanical stress induced model of human aged explants was used to test both repeated dosing and slow release of IOP. METHODS: Human osteochondral explants subjected to injurious mechanical stress (65%MS) were treated with IOP or IOP encapsulated in poly lactic-co-glycolic acid-polyethylene glycol nanoparticles (NP-IOP). Changes to cartilage integrity and signalling were determined by Mankin scoring of histology, sulphated glycosaminoglycan (sGAG) release and expression levels of catabolic, anabolic and hypertrophic markers. Subsequently, on a subgroup of samples, RNA sequencing was performed on 65%MS (n = 14) and 65%MS+IOP (n = 7) treated cartilage to identify IOP's mode of action. RESULTS: Damage from injurious mechanical stress was confirmed by increased cartilage surface damage in the Mankin score, increased sGAG release, and consistent upregulation of catabolic markers and downregulation of anabolic markers. IOP and, though less effective, NP-IOP treatment, reduced MMP13 and increased COL2A1 expression. In line with this, IOP and NP-IOP reduced cartilage surface damage induced by 65%MS, while only IOP reduced sGAG release from explants subjected to 65%MS. Lastly, differential expression analysis identified 12 genes in IOP's mode of action to be mainly involved in reducing metabolic processes (INSIG1, DHCR7, FADS1 and ACAT2) and proliferation and differentiation (CTGF, BMP5 and FOXM1). CONCLUSION: Treatment with the deiodinase inhibitor IOP reduced detrimental changes of injurious mechanical stress. In addition, we identified that its mode of action was likely on metabolic processes, cell proliferation and differentiation.


Assuntos
Cartilagem Articular , Glândula Tireoide , Humanos , Glândula Tireoide/metabolismo , Iodeto Peroxidase/metabolismo , Iodeto Peroxidase/farmacologia , Transdução de Sinais , Cartilagem Articular/metabolismo , Condrócitos/metabolismo
3.
Ann Rheum Dis ; 75(3): 571-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25550340

RESUMO

OBJECTIVE: To further explore deiodinase iodothyronine type 2 (DIO2) as a therapeutic target in osteoarthritis (OA) by studying the effects of forced mechanical loading on in vivo joint cartilage tissue homeostasis and the modulating effect herein of Dio2 deficiency. METHODS: Wild-type and C57BL/6-Dio2(-/-) -mice were subjected to a forced running regime for 1 h per day for 3 weeks. Severity of OA was assessed by histological scoring for cartilage damage and synovitis. Genome-wide gene expression was determined in knee cartilage by microarray analysis (Illumina MouseWG-6 v2). STRING-db analyses were applied to determine enrichment for specific pathways and to visualise protein-protein interactions. RESULTS: In total, 158 probes representing 147 unique genes showed significantly differential expression with a fold-change ≥1.5 upon forced exercise. Among these are genes known for their association with OA (eg, Mef2c, Egfr, Ctgf, Prg4 and Ctnnb1), supporting the use of forced running as an OA model in mice. Dio2-deficient mice showed significantly less cartilage damage and signs of synovitis. Gene expression response upon exercise between wild-type and knockout mice was significantly different for 29 genes. CONCLUSIONS: Mice subjected to a running regime have significant increased cartilage damage and synovitis scores. Lack of Dio2 protected against cartilage damage in this model and was reflected in a specific gene expression profile, and either mark a favourable effect in the Dio2 knockout (eg, Gnas) or an unfavourable effect in wild-type cartilage homeostasis (eg, Hmbg2 and Calr). These data further support DIO2 activity as a therapeutic target in OA.


Assuntos
Cartilagem Articular/metabolismo , Iodeto Peroxidase/genética , Articulação do Joelho/metabolismo , Osteoartrite do Joelho/genética , Condicionamento Físico Animal , RNA Mensageiro/metabolismo , Estresse Mecânico , Animais , Cartilagem Articular/patologia , Perfilação da Expressão Gênica , Articulação do Joelho/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia , Reação em Cadeia da Polimerase em Tempo Real , Iodotironina Desiodinase Tipo II
4.
Development ; 139(18): 3343-54, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22912414

RESUMO

The strength and spatiotemporal activity of Nodal signaling is tightly controlled in early implantation mouse embryos, including by autoregulation and feedback loops, and involves secreted and intracellular antagonists. These control mechanisms, which are established at the extra-embryonic/embryonic interfaces, are essential for anterior-posterior patterning of the epiblast and correct positioning of the primitive streak. Formation of an ectopic primitive streak, or streak expansion, has previously been reported in mutants lacking antagonists that target Nodal signaling. Here, we demonstrate that loss-of-function of a major bone morphogenetic protein (BMP) effector, Smad5, results in formation of an ectopic primitive streak-like structure in mutant amnion accompanied by ectopic Nodal expression. This suggests that BMP/Smad5 signaling contributes to negative regulation of Nodal. In cultured cells, we find that BMP-activated Smad5 antagonizes Nodal signaling by interfering with the Nodal-Smad2/4-Foxh1 autoregulatory pathway through the formation of an unusual BMP4-induced Smad complex containing Smad2 and Smad5. Quantitative expression analysis supports that ectopic Nodal expression in the Smad5 mutant amnion is induced by the Nodal autoregulatory loop and a slow positive-feedback loop. The latter involves BMP4 signaling and also induction of ectopic Wnt3. Ectopic activation of these Nodal feedback loops in the Smad5 mutant amnion results in the eventual formation of an ectopic primitive streak-like structure. We conclude that antagonism of Nodal signaling by BMP/Smad5 signaling prevents primitive streak formation in the amnion of normal mouse embryos.


Assuntos
Âmnio/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Proteína Nodal/metabolismo , Linha Primitiva/metabolismo , Proteína Smad5/metabolismo , Âmnio/citologia , Animais , Western Blotting , Proteínas Morfogenéticas Ósseas/genética , Linhagem Celular , Feminino , Humanos , Imuno-Histoquímica , Imunoprecipitação , Hibridização In Situ , Camundongos , Proteína Nodal/genética , Gravidez , Linha Primitiva/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad5/genética
5.
Proc Natl Acad Sci U S A ; 109(21): 8218-23, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22566624

RESUMO

Hip osteoarthritis (HOA) is one of the most disabling and common joint disorders with a large genetic component that is, however, still ill-defined. To date, genome-wide association studies (GWAS) in osteoarthritis (OA) and specifically in HOA have yielded only few loci, which is partly explained by heterogeneity in the OA definition. Therefore, we here focused on radiographically measured joint-space width (JSW), a proxy for cartilage thickness and an important underlying intermediate trait for HOA. In a GWAS of 6,523 individuals on hip-JSW, we identified the G allele of rs12982744 on chromosome 19p13.3 to be associated with a 5% larger JSW (P = 4.8 × 10(-10)). The association was replicated in 4,442 individuals from three United Kingdom cohorts with an overall meta-analysis P value of 1.1 × 10(-11). The SNP was also strongly associated with a 12% reduced risk for HOA (P = 1 × 10(-4)). The SNP is located in the DOT1L gene, which is an evolutionarily conserved histone methyltransferase, recently identified as a potentially dedicated enzyme for Wnt target-gene activation in leukemia. Immunohistochemical staining of the DOT1L protein in mouse limbs supports a role for DOT1L in chondrogenic differentiation and adult articular cartilage. DOT1L is also expressed in OA articular chondrocytes. Silencing of Dot1l inhibited chondrogenesis in vitro. Dot1l knockdown reduces proteoglycan and collagen content, and mineralization during chondrogenesis. In the ATDC5 chondrogenesis model system, DOT1L interacts with TCF and Wnt signaling. These data are a further step to better understand the role of Wnt-signaling during chondrogenesis and cartilage homeostasis. DOT1L may represent a therapeutic target for OA.


Assuntos
Condrócitos/fisiologia , Condrogênese/genética , Estudo de Associação Genômica Ampla , Metiltransferases/genética , Osteoartrite do Quadril/genética , Fatores Etários , Animais , Cartilagem Articular/patologia , Cartilagem Articular/fisiologia , Linhagem Celular , Condrócitos/citologia , Variação Genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Histona-Lisina N-Metiltransferase , Humanos , Metiltransferases/metabolismo , Camundongos , Osteoartrite do Quadril/epidemiologia , Osteoartrite do Quadril/patologia , Fatores de Risco , Via de Sinalização Wnt/fisiologia
6.
Bone Res ; 11(1): 49, 2023 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-37730805

RESUMO

Abnormal subchondral bone remodeling leading to sclerosis is a main feature of osteoarthritis (OA), and osteomodulin (OMD), a proteoglycan involved in extracellular matrix mineralization, is associated with the sclerotic phenotype. However, the functions of OMD remain poorly understood, specifically in vivo. We used Omd knockout and overexpressing male mice and mutant zebrafish to study its roles in bone and cartilage metabolism and in the development of OA. The expression of Omd is deeply correlated with bone and cartilage microarchitectures affecting the bone volume and the onset of subchondral bone sclerosis and spontaneous cartilage lesions. Mechanistically, OMD binds to RANKL and inhibits osteoclastogenesis, thus controlling the balance of bone remodeling. In conclusion, OMD is a key factor in subchondral bone sclerosis associated with OA. It participates in bone and cartilage homeostasis by acting on the regulation of osteoclastogenesis. Targeting OMD may be a promising new and personalized approach for OA.


Assuntos
Osteoartrite , Peixe-Zebra , Masculino , Animais , Camundongos , Regulação para Baixo , Esclerose , Proteoglicanas , Osteoartrite/genética
7.
Orphanet J Rare Dis ; 15(1): 119, 2020 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-32448375

RESUMO

BACKGROUND: Limb-girdle muscular dystrophy recessive 1 calpain3-related (LGMDR1), previously known as LGMD2A, is a disease caused by mutations in the CAPN3 gene. It is characterized by progressive weakness and muscle degeneration. Frizzled related protein (FRZB), upregulated in LGMDR1, was identified as a key regulator of the crosstalk between Wnt and integrin signalling pathways. FRZB gene silencing showed a recovery in the expression of some of the costamere protein levels in myotubes. RESULTS: Here, we performed a comprehensive characterization of Frzb-/- mice muscles to study the absence of Frzb in skeletal muscle and eventual links with the molecular characteristics of LGMDR1 patient muscles. Frzb-/- mice showed reduced muscle size and strength. Gait analysis showed that Frzb-/- mice moved more slowly but no impaired regeneration capacity was observed after muscle injury. Additionally, Frzb-/- mice muscle showed an increased number of mesoangioblasts. Lack of Frzb gene in Frzb-/- mice and its increased expression in LGMDR1 patients, showed contrary regulation of Rora, Slc16a1, Tfrc and Capn3 genes. The reciprocal regulation of Frzb and Capn3 genes further supports this axis as a potential target for LGMDR1 patients. CONCLUSIONS: Our data confirm a role for Frzb in the regulation of Rora, Slc16a1, Tfrc, and Capn3 genes in muscle cells. In vivo, reduced muscle strength and gait in the Frzb-/- mice are intriguing features. The reciprocal relationship between FRZB and CAPN3 further supports a key role for this axis in patients with LGMDR1.


Assuntos
Distrofia Muscular do Cíngulo dos Membros , Deficiência de Proteína , Animais , Calpaína/genética , Marcha , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Proteínas Musculares , Força Muscular , Músculo Esquelético , Distrofia Muscular do Cíngulo dos Membros/genética
8.
Sci Transl Med ; 10(458)2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30209244

RESUMO

Osteoarthritis is the most common joint disorder with increasing global prevalence due to aging of the population. Current therapy is limited to symptom relief, yet there is no cure. Its multifactorial etiology includes oxidative stress and overproduction of reactive oxygen species, but the regulation of these processes in the joint is insufficiently understood. We report that ANP32A protects the cartilage against oxidative stress, preventing osteoarthritis development and disease progression. ANP32A is down-regulated in human and mouse osteoarthritic cartilage. Microarray profiling revealed that ANP32A protects the joint by promoting the expression of ATM, a key regulator of the cellular oxidative defense. Antioxidant treatment reduced the severity of osteoarthritis, osteopenia, and cerebellar ataxia features in Anp32a-deficient mice, revealing that the ANP32A/ATM axis discovered in cartilage is also present in brain and bone. Our findings indicate that modulating ANP32A signaling could help manage oxidative stress in cartilage, brain, and bone with therapeutic implications for osteoarthritis, neurological disease, and osteoporosis.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Osso e Ossos/metabolismo , Encéfalo/metabolismo , Cartilagem/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Osteoartrite/metabolismo , Estresse Oxidativo , Animais , Antioxidantes/farmacologia , Osso e Ossos/patologia , Encéfalo/patologia , Cartilagem/patologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Suscetibilidade a Doenças , Masculino , Camundongos , Proteínas Nucleares/deficiência , Osteoartrite/patologia , Estresse Oxidativo/efeitos dos fármacos , Proteínas de Ligação a RNA
9.
Nat Commun ; 8: 15889, 2017 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-28627522

RESUMO

Osteoarthritis is the most prevalent and crippling joint disease, and lacks curative treatment, as the underlying molecular basis is unclear. Here, we show that DOT1L, an enzyme involved in histone methylation, is a master protector of cartilage health. Loss of DOT1L disrupts the molecular signature of healthy chondrocytes in vitro and causes osteoarthritis in mice. Mechanistically, the protective function of DOT1L is attributable to inhibition of Wnt signalling, a pathway that when hyper-activated can lead to joint disease. Unexpectedly, DOT1L suppresses Wnt signalling by inhibiting the activity of sirtuin-1 (SIRT1), an important regulator of gene transcription. Inhibition of SIRT1 protects against osteoarthritis triggered by loss of DOT1L activity. Modulating the DOT1L network might therefore be a therapeutic approach to protect the cartilage against osteoarthritis.


Assuntos
Cartilagem/metabolismo , Metiltransferases/metabolismo , Osteoartrite/patologia , Animais , Benzimidazóis/farmacologia , Benzimidazóis/toxicidade , Cartilagem/patologia , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Feminino , Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase , Homeostase , Masculino , Metilação , Metiltransferases/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoartrite/induzido quimicamente , Osteoartrite/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo , Via de Sinalização Wnt
10.
PLoS One ; 11(5): e0154999, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27163789

RESUMO

OBJECTIVE: To identify intrinsic differences in cartilage gene expression profiles between wild-type- and Dio2-/--mice, as a mechanism to investigate factors that contribute to prolonged healthy tissue homeostasis. METHODS: Previously generated microarray-data (Illumina MouseWG-6 v2) of knee cartilage of wild-type and Dio2 -/- -mice were re-analyzed to identify differential expressed genes independent of mechanical loading conditions by forced treadmill-running. RT-qPCR and western blot analyses of overexpression and knockdown of Calr in mouse chondro-progenitor cells (ATDC5) were applied to assess the direct effect of differential Calr expression on cartilage deposition. RESULTS: Differential expression analyses of articular cartilage of Dio2-/- (N = 9) and wild-type-mice (N = 11) while applying a cutoff threshold (P < 0.05 (FDR) and FC > |1,5|) resulted in 1 probe located in Calreticulin (Calr) that was found significantly downregulated in Dio2-/- mice (FC = -1.731; P = 0.044). Furthermore, overexpression of Calr during early chondrogenesis in ATDC5 cells leads to decreased proteoglycan deposition and corresponding lower Aggrecan expression, whereas knocking down Calr expression does not lead to histological differences of matrix composition. CONCLUSION: We here demonstrate that the beneficial homeostatic state of articular cartilage in Dio2-/- mice is accompanied with significant lower expression of Calr. Functional analyses further showed that upregulation of Calr expression could act as an initiator of cartilage destruction. The consistent association between Calr and Dio2 expression suggests that enhanced expression of these genes facilitate detrimental effects on cartilage integrity.


Assuntos
Calreticulina/genética , Cartilagem Articular/metabolismo , Iodeto Peroxidase/genética , Osteoartrite/genética , Articulação Patelofemoral/metabolismo , Agrecanas/genética , Agrecanas/metabolismo , Animais , Calreticulina/metabolismo , Cartilagem Articular/patologia , Condrócitos/metabolismo , Condrócitos/patologia , Teste de Esforço , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Iodeto Peroxidase/deficiência , Masculino , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Osteoartrite/metabolismo , Osteoartrite/patologia , Articulação Patelofemoral/patologia , Proteoglicanas/genética , Proteoglicanas/metabolismo , Células-Tronco/metabolismo , Células-Tronco/patologia , Iodotironina Desiodinase Tipo II
11.
Discov Med ; 12(63): 129-39, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21878190

RESUMO

Osteoarthritis is the most common chronic musculoskeletal disorder. Although osteoarthritis is a multifactorial disease, it is known to have a significant genetic contribution. Identified genes so far, contributing to the prevalence and progression of the disease, belong to signaling pathways such as the bone morphogenetic protein and the wingless-type signaling families. These pathways are important during development and appear to be reactivated in postnatal joint homeostasis and repair. Among the most consistently associated genes, so far, are GDF5 and FRZB, which were both originally identified from a chondrogenic extract of articular cartilage. We focus on the functional aspects of these susceptibility genes and summarize recent evidence, obtained in specific animal models, for their possible roles in osteoarthritis. These data support the view that all tissues within the joint may contribute to osteoarthritis and that pathways important in skeletal development are also involved in the disease processes thereby presenting attractive therapeutic targets.


Assuntos
Osteoartrite/metabolismo , Animais , Predisposição Genética para Doença , Glicoproteínas/genética , Glicoproteínas/metabolismo , Fator 5 de Diferenciação de Crescimento/genética , Fator 5 de Diferenciação de Crescimento/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Modelos Biológicos , Osteoartrite/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA