Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Inflamm Res ; 72(12): 2145-2153, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37874359

RESUMO

OBJECTIVE AND DESIGN: 15-Lipoxygenase-1 (15-LOX-1) catalyzes the biosynthesis of many anti-inflammatory and immunomodulatory lipid mediators and was reported to have protective properties in several inflammatory conditions, including osteoarthritis (OA). This study was designed to evaluate the expression of 15-LOX-1 in cartilage from normal donors and patients with OA, and to determine whether it is regulated by DNA methylation. METHODS: Cartilage samples were obtained at autopsy from normal knee joints and from OA-affected joints at the time of total knee joint replacement surgery. The expression of 15-LOX-1 was evaluated using real-time polymerase chain reaction (PCR). The role of DNA methylation in 15-LOX-1 expression was assessed using the DNA methyltransferase inhibitor 5-Aza-2'-desoxycytidine (5-Aza-dC). The effect of CpG methylation on 15-LOX-1 promoter activity was evaluated using a CpG-free luciferase vector. The DNA methylation status of the 15-LOX-1 promoter was determined by pyrosequencing. RESULTS: Expression of 15-LOX-1 was upregulated in OA compared to normal cartilage. Treatment with 5-Aza-dC increased 15-LOX-1 mRNA levels in chondrocytes, and in vitro methylation decreased 15-LOX-1 promoter activity. There was no difference in the methylation status of the 15-LOX-1 gene promoter between normal and OA cartilage. CONCLUSION: The expression level of 15-LOX-1 was elevated in OA cartilage, which may be part of a repair process. The upregulation of 15-LOX-1 in OA cartilage was not associated with the methylation status of its promoter, suggesting that other mechanisms are involved in its upregulation.


Assuntos
Araquidonato 15-Lipoxigenase , Osteoartrite , Humanos , Araquidonato 15-Lipoxigenase/genética , Araquidonato 15-Lipoxigenase/metabolismo , Condrócitos/metabolismo , Metilação de DNA , Epigênese Genética , Osteoartrite/genética , Osteoartrite/metabolismo , Receptores Depuradores Classe E/genética , Receptores Depuradores Classe E/metabolismo
2.
Health Promot Int ; 38(4)2023 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-34967862

RESUMO

Vaccine hesitancy is a global health issue and can be affected by several variables. We explored the predictive factors and causes of vaccine hesitancy among adults in Saudi Arabia. An online survey method with multiple regression analysis was used to identify factors predicting of vaccine hesitancy in 558 adults (46.24% women and 53.76% men). The prevalence of vaccine hesitancy is 20.6%, with higher rates among females, young people and single people. About 70% of the participants believe that vaccine hesitancy is due to concerns about the safety and efficacy of the vaccine, a lack of information about the disease and vaccine or social media. The vaccine acceptance rate is 71.3%; 17.2% are not willing to accept a COVID-19 vaccine and 11.5% are unsure. Males and married people are more accepting of the vaccine. The risk factors that predict vaccine hesitancy include age, gender, belief in conspiracy theories and psychosocial factors. Meanwhile, age, gender, belief in conspiracy theories, concerns about the safety and efficacy of the vaccine and psychosocial factors significantly predict vaccine acceptance. The high rate of vaccine hesitancy could undermine efforts to combat COVID-19. Factors predicting vaccine hesitancy can be used in interventions to address this issue during major epidemics.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Adulto , Masculino , Feminino , Humanos , Adolescente , Arábia Saudita , Hesitação Vacinal , COVID-19/epidemiologia , COVID-19/prevenção & controle , Vacinação
3.
Int J Mol Sci ; 22(14)2021 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-34298927

RESUMO

Adult human subcutaneous adipose tissue (AT) harbors a rich population of mesenchymal stromal cells (MSCs) that are of interest for tissue repair. For this purpose, it is of utmost importance to determine the response of AT-MSCs to proliferative and inflammatory signals within the damaged tissue. We have characterized the transcriptional profile of cytokines, regulatory mediators and Toll-like receptors (TLR) relevant to the response of MSCs. AT-MSCs constitutively present a distinct profile for each gene and differentially responded to inflammation and cell-passaging. Inflammation leads to an upregulation of IL-6, IL-8, IL-1ß, TNFα and CCL5 cytokine expression. Inflammation and cell-passaging increased the expression of HGF, IDO1, PTGS1, PTGS2 and TGFß. The expression of the TLR pattern was differentially modulated with TLR 1, 2, 3, 4, 9 and 10 being increased, whereas TLR 5 and 6 downregulated. Functional enrichment analysis demonstrated a complex interplay between cytokines, TLR and regulatory mediators central for tissue repair. This profiling highlights that following a combination of inflammatory and proliferative signals, the sensitivity and responsive capacity of AT-MSCs may be significantly modified. Understanding these transcriptional changes may help the development of novel therapeutic approaches.


Assuntos
Citocinas/genética , Regulação da Expressão Gênica/genética , Inflamação/genética , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais/genética , Receptores Toll-Like/genética , Transcrição Gênica/genética , Proliferação de Células/genética , Regulação para Baixo/genética , Humanos , Gordura Subcutânea/metabolismo , Regulação para Cima/genética
4.
J Cell Physiol ; 234(11): 21145-21152, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31041809

RESUMO

Adipose tissue-derived mesenchymal stromal cells (ASCs) hold the promise of achieving successful immunotherapeutic results due to their ability to regulate different T-cell fate. ASCs also show significant adaptability to environmental stresses by modulating their immunologic profile. Cell-based therapy for inflammatory diseases requires a detailed understanding of the molecular relation between ASCs and Th17 lymphocytes taking into account the influence of inflammation and cell ratio on such interaction. Accordingly, a dose-dependent increase in Th17 generation was only observed in high MSC:T-cell ratio with no significant impact of inflammatory priming. IL-23 receptor (IL-23R) expression by T cells was not modulated by ASCs when compared to levels in activated T cells, while ROR-γt expression was significantly increased reaching a maximum in high (1:5) unprimed ASC:T-cell ratio. Finally, multiplex immunoassay showed substantial changes in the secretory profile of 15 cytokines involved in the Th17 immune response (IL-1ß, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-22, IL-21, IL-23, IL-25, IL-31, IL-33, IFN-γ, sCD40, and TNF-α), which was modulated by both cell ratio and inflammatory priming. These findings suggest that Th17 lymphocyte pathway is significantly modulated by ASCs that may lead to immunological changes. Therefore, future ASC-based immunotherapy should take into account the complex and detailed molecular interactions that depend on several factors including inflammatory priming and cell ratio.


Assuntos
Células-Tronco Mesenquimais/imunologia , Células Th17/imunologia , Diferenciação Celular/imunologia , Humanos , Ativação Linfocitária/imunologia
5.
Inflamm Res ; 68(2): 167-176, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30426152

RESUMO

OBJECTIVE AND DESIGN: Bone marrow mesenchymal stromal cells (BM-MSCs) are referred as a promising immunotherapeutic cell product. New approaches using empowered MSCs should be developed as for the treatment or prevention of different immunological diseases. Such preconditioning by new licensing stimuli will empower the immune fate of BM-MSCs and, therefore, promote a better and more efficient biological. Here, our main goal was to establish the immunological profile of BM-MSCs following inflammatory priming and in particular their capacity to adjust their immune-related proteome and transcriptome. MATERIAL AND METHODS: To run this study, we have used BM-MSC cell cultures, a pro-inflammatory cytokine cocktail priming, flow cytometry analysis, qPCR and ELISA techniques. RESULTS: Different expression levels of several immunological mediators such as COX-1, COX-2, LIF, HGF, Gal-1, HO-1, IL-11, IL-8, IL-6 and TGF-ß were constitutively observed in BM-MSCs. Inflammation priming substantially but differentially modulated the gene and protein expression profiles of these mediators. Thus, expressions of COX-2, LIF, HGF, IL-11, IL-8 and IL-6 were highly increased/induced and those of COX-1, Gal-1, and TGF-ß were reduced. CONCLUSIONS: Collectively, we demonstrated that BM-MSCs are endowed with a specific and modular regulatory machinery which is potentially involved in immunomodulation. Moreover, BM-MSCs are highly sensitive to inflammation and respond to such signal by properly adjusting their gene and protein expression of regulatory factors. Using such preconditioning may empower the immune fate of MSCs and, therefore, enhance their value for cell-based immunotherapy.


Assuntos
Células da Medula Óssea/imunologia , Inflamação/genética , Inflamação/imunologia , Células-Tronco Mesenquimais/imunologia , Citocinas/biossíntese , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Humanos , Imunomodulação/genética , Imunomodulação/fisiologia , Mediadores da Inflamação/metabolismo , Transplante de Células-Tronco Mesenquimais , Reação em Cadeia da Polimerase , Processamento de Proteína Pós-Traducional , Proteoma/genética , Transcriptoma/genética
6.
Inflamm Res ; 68(3): 203-213, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30506263

RESUMO

OBJECTIVE AND DESIGN: The objective of the study is to uncover the influence of human bone marrow-derived mesenchymal stem cells (BM-MSCs) on the generation of Th17 lymphocytes in co-cultures of both BM-MSCs and T cells. MATERIALS AND METHODS: BM-MSCs, characterized according to the international society for cellular therapy (ISCT) criteria, were co-cultured with T cells isolated from peripheral blood. The expression levels of IL-17 receptor, RORγt and IL-23 receptor were evaluated using flow cytometry. The levels of cytokines involved in Th17 immunomodulation were measured using multiplex assay. TREATMENT: Inflammatory primed and non-primed BM-MSCs were co-cultured with either activated or non-activated T cells either at (1/80) and (1/5) ratio respectively. RESULTS: MSC/T-cell ratio and inflammation significantly influenced the effect of BM-MSCs on the generation of Th17 lymphocytes. Cocultures of either primed or non-primed BM-MSCs with activated T cells significantly induced IL-17A-expressing lymphocytes. Interestingly, the expression of the transcription factor RORγt was significantly increased when compared to levels in activated T cells. Finally, both cell ratio and priming of BM-MSCs with cytokines substantially influenced the cytokine profile of BM-MSCs and T cells. CONCLUSION: Our findings suggest that BM-MSCs significantly modulate the Th17 lymphocyte pathway in a complex manner.


Assuntos
Células-Tronco Mesenquimais/imunologia , Células Th17/imunologia , Células da Medula Óssea/citologia , Técnicas de Cocultura , Citocinas/imunologia , Humanos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Receptores de Interleucina/imunologia
7.
Rheumatology (Oxford) ; 57(10): 1851-1860, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29982662

RESUMO

Objective: This study explored the role of the adipokine adipsin in OA. Methods: Control and OA articular tissues, cells and serum were obtained from human individuals. Serum adipsin levels of human OA individuals were compared with cartilage volume loss as assessed by MRI at 48 months. Human adipsin expression was determined by PCR, its production in tissues by immunohistochemistry, and in SF and serum by a specific assay. OA was surgically induced in wild-type (Df+/+) and adipsin-deficient (Df-/-) mice, and synovial membrane and cartilage processed for histology and immunohistochemistry. Results: Adipsin levels were significantly increased in human OA serum, SF, synovial membrane and cartilage compared with controls, but the expression was similar in chondrocytes, synoviocytes and osteoblasts. Multivariate analysis demonstrated that human serum adipsin levels were significantly associated (P = 0.045) with cartilage volume loss in the lateral compartment of the knee. Destabilization of the medial meniscus-Df-/- mice showed a preservation of the OA synovial membrane and cartilage lesions (P ⩽ 0.026), the latter corroborated by the decreased production of cartilage degradation products and proteases (P ⩽ 0.047). The adipsin effect is likely due to a deficient alternative complement pathway (P ⩽ 0.036). Conclusion: In human OA, higher serum adipsin levels were associated with greater cartilage volume loss in the lateral compartment, and adipsin deficiency led to a preservation of knee structure. Importantly, we documented an association between adipsin and OA synovial membrane and cartilage degeneration through the activation of the complement pathway. This study highlights the clinical relevance of adipsin as a valuable biomarker and potential therapeutic target for OA.


Assuntos
Fator D do Complemento/metabolismo , Articulação do Joelho/metabolismo , Joelho/patologia , Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia , Animais , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Humanos , Joelho/diagnóstico por imagem , Articulação do Joelho/diagnóstico por imagem , Imageamento por Ressonância Magnética , Camundongos , Osteoartrite do Joelho/diagnóstico por imagem , Osteoblastos/metabolismo , Membrana Sinovial/citologia , Membrana Sinovial/metabolismo , Sinoviócitos/metabolismo
8.
J Cell Biochem ; 118(5): 1003-1013, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27463229

RESUMO

Protandim and 6-gingerol, two potent nutraceuticals, have been shown to decrease free radicals production through enhancing endogenous antioxidant enzymes. In this study, we evaluated the effects of these products on the expression of different factors involved in osteoarthritis (OA) process. Human OA chondrocytes were treated with 1 ng/ml IL-1ß in the presence or absence of protandim (0-10 µg/ml) or 6-gingerol (0-10 µM). OA was induced surgically in mice by destabilization of the medial meniscus (DMM). The animals were treated weekly with an intraarticular injection of 10 µl of vehicle or protandim (10 µg/ml) for 8 weeks. Sham-operated mice served as controls. In vitro, we demonstrated that protandim and 6-gingerol preserve cell viability and mitochondrial metabolism and prevented 4-hydroxynonenal (HNE)-induced cell mortality. They activated Nrf2 transcription factor, abolished IL-1ß-induced NO, PGE2 , MMP-13, and HNE production as well as IL-ß-induced GSTA4-4 down-regulation. Nrf2 overexpression reduced IL-1ß-induced HNE and MMP-13 as well as IL-1ß-induced GSTA4-4 down-regulation. Nrf2 knockdown following siRNA transfection abolished protandim protection against oxidative stress and catabolism. The activation of MAPK and NF-κB by IL-1ß was not affected by 6-gingerol. In vivo, we observed that Nrf2 and GSTA4-4 expression was significantly lower in OA cartilage from humans and mice compared to normal controls. Interestingly, protandim administration reduced OA score in DMM mice. Altogether, our data indicate that protandim and 6-gingerol are essential in preserving cartilage and abolishing a number of factors known to be involved in OA pathogenesis. J. Cell. Biochem. 118: 1003-1013, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Anti-Inflamatórios/administração & dosagem , Catecóis/administração & dosagem , Condrócitos/efeitos dos fármacos , Medicamentos de Ervas Chinesas/administração & dosagem , Álcoois Graxos/administração & dosagem , Osteoartrite/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Catecóis/farmacologia , Sobrevivência Celular , Células Cultivadas , Condrócitos/citologia , Suplementos Nutricionais , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/farmacologia , Álcoois Graxos/farmacologia , Glutationa Transferase/metabolismo , Humanos , Injeções Intra-Articulares , Interleucina-1beta/efeitos adversos , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Inflamm Res ; 66(8): 637-651, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28447122

RESUMO

BACKGROUND: Over the years, many theories have been proposed and examined to better explain the etiology and development of osteoarthritis (OA). The characteristics of joint destruction are one of the most important aspects in disease progression. Therefore, investigating different factors and signaling pathways involved in the alteration of extracellular matrix (ECM) turnover, and the subsequent catabolic damage to cartilage holds chief importance in understanding OA development. Among these factors, reactive oxygen species (ROS) have been at the forefront of the physiological and pathophysiological OA investigation. FINDINGS: In the last decades, research studies provided an enormous volume of data supporting the involvement of ROS in OA. Most interestingly, published data regarding the effect of exogenous antioxidant therapy in OA lack conclusive results from clinical trials to back up in vitro data. Accordingly, it is rational to suggest that there are other reactive species in OA that are not taken into account. Thus, our present review is focused on our current understanding of the involvement of lipid peroxidation-derived 4-hydroxynonenal (HNE) in OA. CONCLUSION: Our findings, like those in the literature, illustrate the central role played by HNE in the regulation of a number of factors involved in joint homeostasis. HNE could thus be considered as an attractive therapeutic target in OA.


Assuntos
Aldeídos/metabolismo , Peroxidação de Lipídeos , Osteoartrite/metabolismo , Animais , Apoptose , Condrócitos , Humanos , Estresse Oxidativo
10.
Am J Pathol ; 185(2): 335-46, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25453723

RESUMO

Osteoarthritis (OA) is characterized by progressive joint destruction, including synovial membrane alteration. EphB4 and its ligand ephrin-B2 were found in vitro to positively affect OA subchondral bone and cartilage. In vivo in an experimental mouse model overexpressing bone-specific Ephb4 (TgEphB4), a protective effect was found on both the subchondral bone and cartilage during OA. We investigated in the TgEphB4 mouse model the in vivo effect on synovial membrane during OA. Knee OA was surgically induced by destabilization of the medial meniscus (DMM). Synovial membrane was evaluated using histology, histomorphometry, IHC, and real-time PCR. Compared to DMM-wild-type (WT) mice, DMM-TgEphB4 mice had a significant decrease in synovial membrane thickness, vascular endothelial growth factor, and the profibrotic markers fibrin, type 1 procollagen, type 3 collagen, connective tissue growth factor, smooth muscle actin-α, cartilage oligomeric matrix protein, and procollagen-lysine, and 2-oxoglutarate 5-dioxygenase 2. Moreover, factors known to modulate transforming growth factor-ß signaling, transforming growth factor receptor 1/ALK1, phosphorylated Smad-1, and heat shock protein 90ß were significantly decreased in DMM-TgEphB4 compared with DMM-WT mice. Ephb4 overexpression also exhibited a protective effect on synovial membrane thickness of aged (24-month-old) mice. Overexpression of bone-specific Ephb4 clearly demonstrated prevention of the development and/or progression of fibrosis in OA synovial membrane, reinforcing the hypothesis that protecting the subchondral bone prophylactically and during OA reduces the pathologic changes in other articular tissues.


Assuntos
Regulação da Expressão Gênica , Osteoartrite , Receptor EphB4 , Membrana Sinovial , Animais , Fibrose , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/genética , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite/patologia , Osteoartrite/prevenção & controle , Receptor EphB4/biossíntese , Receptor EphB4/genética , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia
11.
Inflamm Res ; 65(8): 635-45, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27056390

RESUMO

OBJECTIVE AND DESIGN: Resolvin D1 (RvD1), an omega-3 fatty acid derivative, has shown remarkable properties in resolving inflammation, promoting tissue repair and preserving tissue integrity. In this study, we investigated RvD1 effects on major processes involved in osteoarthritis (OA) pathophysiology. MATERIALS AND METHODS: Human OA chondrocytes were treated with either 1 ng/ml interleukin-1ß (IL-1ß) or 20 µM 4-hydroxynonenal (HNE), then treated or not with increased concentrations of RvD1 (0-10 µM). RvD1 levels were measured by enzyme immunoassay in synovial fluids from experimental dog model of OA and sham operated dogs obtained from our previous study. Cell viability was evaluated by 3-(4,5-dimethyl-thiazoyl)-2,5-diphenyl-SH-tetrazolium bromide assay. Parameters related to inflammation, catabolism and apoptosis were determined by enzyme-linked immunosorbent assay, Western blotting, and quantitative polymerase chain reaction. Glutathione (GSH) was assessed by commercial kit. The activation of mitogen-activated protein kinases and nuclear factor-kappaB (NF-κB) pathways was evaluated by Western blot. RESULTS: We showed that RvD1 levels were higher in synovial fluids from OA joint compared to controls. In OA human chondrocytes, we demonstrated that RvD1 was not toxic up to 10 µM and stifled IL-1ß-induced cyclooxygenase 2, prostaglandin E2, inducible nitric oxide synthase, nitric oxide, and matrix metalloproteinase-13. Our study of signalling pathways revealed that RvD1 suppressed IL-1ß-induced activation of NF-κB/p65, p38/MAPK and JNK(1/2). Moreover, RvD1 prevented HNE-induced cell apoptosis and oxidative stress, as indicated by inactivation of caspases, inhibition of lactate dehydrogenase release, and increased levels of Bcl2 and AKT, as well as GSH. CONCLUSION: This is the first in vitro study demonstrating the beneficial effect of RvD1 in OA. That RvD1 abolishing a number of factors known to be involved in OA pathogenesis renders it a clinically valuable agent in prevention of the disease.


Assuntos
Antioxidantes/farmacologia , Condrócitos/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/farmacologia , Osteoartrite/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Condrócitos/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/genética , Dinoprostona/metabolismo , Cães , Humanos , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Líquido Sinovial/metabolismo
12.
Ann Rheum Dis ; 74(7): 1432-40, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24651621

RESUMO

OBJECTIVES: Mammalian target of rapamycin (mTOR) (a serine/threonine protein kinase) is a major repressor of autophagy, a cell survival mechanism. The specific in vivo mechanism of mTOR signalling in OA pathophysiology is not fully characterised. We determined the expression of mTOR and known autophagy genes in human OA cartilage as well as mouse and dog models of experimental OA. We created cartilage-specific mTOR knockout (KO) mice to determine the specific role of mTOR in OA pathophysiology and autophagy signalling in vivo. METHODS: Inducible cartilage-specific mTOR KO mice were generated and subjected to mouse model of OA. Human OA chondrocytes were treated with rapamycin and transfected with Unc-51-like kinase 1 (ULK1) siRNA to determine mTOR signalling. RESULTS: mTOR is overexpressed in human OA cartilage as well as mouse and dog experimental OA. Upregulation of mTOR expression co-relates with increased chondrocyte apoptosis and reduced expression of key autophagy genes during OA. Subsequently, we show for the first time that cartilage-specific ablation of mTOR results in increased autophagy signalling and a significant protection from destabilisation of medial meniscus (DMM)-induced OA associated with a significant reduction in the articular cartilage degradation, apoptosis and synovial fibrosis. Furthermore, we show that regulation of ULK1/adenosine monophosphate-activated protein kinase (AMPK) signalling pathway by mTOR may in part be responsible for regulating autophagy signalling and the balance between catabolic and anabolic factors in the articular cartilage. CONCLUSIONS: This study provides a direct evidence of the role of mTOR and its downstream modulation of autophagy in articular cartilage homeostasis.


Assuntos
Autofagia/fisiologia , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Osteoartrite/metabolismo , Osteoartrite/prevenção & controle , Serina-Treonina Quinases TOR/deficiência , Regulação para Cima/fisiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose/fisiologia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Cartilagem Articular/efeitos dos fármacos , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Modelos Animais de Doenças , Cães , Inativação Gênica , Humanos , Imunossupressores/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Osteoartrite/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
13.
Ann Rheum Dis ; 74(3): 569-78, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25573665

RESUMO

OBJECTIVES: We have previously shown that peroxisome proliferator-activated receptor gamma (PPARγ), a transcription factor, is essential for the normal growth and development of cartilage. In the present study, we created inducible cartilage-specific PPARγ knockout (KO) mice and subjected these mice to the destabilisation of medial meniscus (DMM) model of osteoarthritis (OA) to elucidate the specific in vivo role of PPARγ in OA pathophysiology. We further investigated the downstream PPARγ signalling pathway responsible for maintaining cartilage homeostasis. METHODS: Inducible cartilage-specific PPARγ KO mice were generated and subjected to DMM model of OA. We also created inducible cartilage-specific PPARγ/mammalian target for rapamycin (mTOR) double KO mice to dissect the PPARγ signalling pathway in OA. RESULTS: Compared with control mice, PPARγ KO mice exhibit accelerated OA phenotype with increased cartilage degradation, chondrocyte apoptosis, and the overproduction of OA inflammatory/catabolic factors associated with the increased expression of mTOR and the suppression of key autophagy markers. In vitro rescue experiments using PPARγ expression vector reduced mTOR expression, increased expression of autophagy markers and reduced the expression of OA inflammatory/catabolic factors, thus reversing the phenotype of PPARγ KO mice chondrocytes. To dissect the in vivo role of mTOR pathway in PPARγ signalling, we created and subjected PPARγ-mTOR double KO mice to the OA model to see if the genetic deletion of mTOR in PPARγ KO mice (double KO) can rescue the accelerated OA phenotype observed in PPARγ KO mice. Indeed, PPARγ-mTOR double KO mice exhibit significant protection/reversal from OA phenotype. SIGNIFICANCE: PPARγ maintains articular cartilage homeostasis, in part, by regulating mTOR pathway.


Assuntos
Cartilagem Articular/metabolismo , Osteoartrite do Joelho/metabolismo , PPAR gama/genética , Serina-Treonina Quinases TOR/genética , Animais , Modelos Animais de Doenças , Meniscos Tibiais/cirurgia , Camundongos , Camundongos Knockout , PPAR gama/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
14.
Am J Pathol ; 182(4): 1099-106, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23375622

RESUMO

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.


Assuntos
Envelhecimento/metabolismo , Cartilagem/metabolismo , Cartilagem/patologia , Osteoartrite/metabolismo , Osteoartrite/patologia , PPAR gama/deficiência , Animais , Biomarcadores/metabolismo , Fibrose , Deleção de Genes , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout , Especificidade de Órgãos , PPAR gama/metabolismo , Fenótipo , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia
15.
Inflamm Res ; 63(8): 691-701, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24858301

RESUMO

OBJECTIVE AND DESIGN: Our study was designed to elucidate the precise molecular mechanisms by which sorbitol-modified hyaluronic acid (HA/sorbitol) exerts beneficial effects in osteoarthritis (OA). METHODS: Human OA chondrocytes were treated with increasing doses of HA/sorbitol ± anti-CD44 antibody or with sorbitol alone and thereafter with or without interleukin-1beta (IL-1ß) or hydrogen peroxide (H2O2). Signal transduction pathways and parameters related to oxidative stress, apoptosis, inflammation, and catabolism were investigated. RESULTS: HA/sorbitol prevented IL-1ß-induced oxidative stress, as measured by reactive oxygen species, p47-NADPH oxidase phosphorylation, 4-hydroxynonenal (HNE) production and HNE-metabolizing glutathione-S-transferase A4-4 expression. Moreover, HA/sorbitol stifled IL-1ß-induced metalloproteinase-13, nitric oxide (NO) and prostaglandin E2 release as well as inducible NO synthase expression. Study of the apoptosis process revealed that this gel significantly attenuated cell death, caspase-3 activation and DNA fragmentation elicited by exposure to a cytotoxic H2O2 dose. Examination of signaling pathway components disclosed that HA/sorbitol prevented IL-1ß-induced p38 mitogen-activated protein kinase and nuclear factor-kappa B activation, but not that of extracellular signal-regulated kinases 1 and 2. Interestingly, the antioxidant as well as the anti-inflammatory and anti-catabolic effects of HA/sorbitol were attributed to sorbitol and HA, respectively. CONCLUSIONS: Altogether, our findings support a beneficial effect of HA/sorbitol in OA through the restoration of redox status and reduction of apoptosis, inflammation and catabolism involved in cartilage damage.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Condrócitos/efeitos dos fármacos , Ácido Hialurônico/farmacologia , Sorbitol/química , Idoso , Aldeídos/metabolismo , Anti-Inflamatórios/química , Antioxidantes/química , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Condrócitos/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Dinoprostona/metabolismo , Glutationa Transferase/metabolismo , Humanos , Ácido Hialurônico/química , Peróxido de Hidrogênio , Interleucina-1beta , Metaloproteinase 13 da Matriz/metabolismo , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Osteoartrite/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
16.
Antioxidants (Basel) ; 13(4)2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38671833

RESUMO

In osteoarthritis (OA), oxidative stress plays a crucial role in maintaining and sustaining cartilage degradation. Current OA management requires a combination of pharmaceutical and non-pharmacological strategies, including intraarticular injections of hyaluronic acid (HA). However, several lines of evidence reported that HA oxidation by reactive oxygen species (ROS) is linked with HA cleavage and fragmentation, resulting in reduced HA viscosity. Resolvin D1 (RvD1) is a lipid mediator that is biosynthesized from omega-3 polyunsaturated fatty acids and is a good candidate with the potential to regulate a panoply of biological processes, including tissue repair, inflammation, oxidative stress, and cell death in OA. Herein, newly designed and synthesized imidazole-derived RvD1 analogues were introduced to compare their potential antioxidant properties with commercially available RvD1. Their antioxidant capacities were investigated by several in vitro chemical assays including oxygen radical absorbance capacity, 2,2-diphenyl-1-picrylhydrazyl radical scavenging, ferric ion reducing antioxidant power, hydroxyl radical scavenging, and HA fragmentation assay. All results proved that imidazole-derived RvD1 analogues showed excellent antioxidant performance compared to RvD1 due to their structural modifications. Interestingly, they scavenged the formed reactive oxygen species (ROS) and protected HA from degradation, as verified by agarose gel electrophoresis and gel permission chromatography. A computational study using Gaussian 09 with DFT calculations and a B3LYP/6-31 G (d, p) basis set was also employed to study the relationship between the antioxidant properties and chemical structures as well as calculation of the molecular structures, frontier orbital energy, molecular electrostatic potential, and bond length. The results showed that the antioxidant activity of our analogues was higher than that of RvD1. In conclusion, the findings suggest that imidazole-derived RvD1 analogues can be good candidates as antioxidant molecules for the treatment of oxidative stress-related diseases like OA. Therefore, they can prolong the longevity of HA in the knee and thus may improve the mobility of the articulation.

17.
Cells ; 13(2)2024 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-38247860

RESUMO

As a form of immunomodulatory therapeutics, mesenchymal stromal/stem cells (MSCs) from umbilical cord (UC) tissue were assessed for their dynamic interplay with the Th-17 immune response pathway. UC-MSCs were able to modulate lymphocyte response by promoting a Th-17-like profile. Such modulation depended on the cell ratio of the cocultures as well as the presence of an inflammatory setting underlying their plasticity. UC-MSCs significantly increased the expression of IL-17A and RORγt but differentially modulated T cell expression of IL-23R. In parallel, the secretion profile of the fifteen factors (IL1ß, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-22, IL-21, IL-23, IL-25, IL-31, IL-33, INF-γ, sCD40, and TNF-α) involved in the Th-17 immune response pathway was substantially altered during these cocultures. The modulation of these factors demonstrates the capacity of UC-MSCs to sense and actively respond to tissue challenges. Protein network and functional enrichment analysis indicated that several biological processes, molecular functions, and cellular components linked to distinct Th-17 signaling interactions are involved in several trophic, inflammatory, and immune network responses. These immunological changes and interactions with the Th-17 pathway are likely critical to tissue healing and may help to identify molecular targets that will improve therapeutic strategies involving UC-MSCs.


Assuntos
Interleucina-17 , Células-Tronco Mesenquimais , Células Th17 , Técnicas de Cocultura , Imunomodulação
18.
Biomolecules ; 14(7)2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39062566

RESUMO

BACKGROUND: AT-MSCs display great immunoregulatory features, making them potential candidates for cell-based therapy. This study aimed to evaluate the "RBC lysis buffer" isolation protocol and immunological profiling of the so-obtained AT-MSCs. METHODS: We established an immune-comparative screening of AT-MSCs throughout in vitro cell expansion (PM, P1, P2, P3, P4) and inflammatory priming regarding the expression of 28 cell-surface markers, 6 cytokines/chemokines, and 10 TLR patterns. FINDINGS: AT-MSCs were highly expandable and sensitive to microenvironment challenges, hereby showing plasticity in distinct expression profiles. Both cell expansion and inflammation differentially modulated the expression profile of CD34, HLA-DR, CD40, CD62L, CD200 and CD155, CD252, CD54, CD58, CD106, CD274 and CD112. Inflammation resulted in a significant increase in the expression of the cytokines IL-6, IL-8, IL-1ß, IL-1Ra, CCL5, and TNFα. Depending on the culture conditions, the expression of the TLR pattern was distinctively altered with TLR1-4, TLR7, and TLR10 being increased, whereas TLR6 was downregulated. Protein network and functional enrichment analysis showed that several trophic and immune responses are likely linked to these immunological changes. CONCLUSIONS: AT-MSCs may sense and actively respond to tissue challenges by modulating distinct and specific pathways to create an appropriate immuno-reparative environment. These mechanisms need to be further characterized to identify and assess a molecular target that can enhance or impede the therapeutic ability of AT-MSCs, which therefore will help improve the quality, safety, and efficacy of the therapeutic strategy.


Assuntos
Tecido Adiposo , Citocinas , Inflamação , Células-Tronco Mesenquimais , Humanos , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Citocinas/metabolismo , Tecido Adiposo/citologia , Proliferação de Células , Células Cultivadas , Adulto , Receptores Toll-Like/metabolismo , Feminino
19.
Ann Rheum Dis ; 72(2): 250-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23065732

RESUMO

OBJECTIVE: To explore the disease-modifying effect, under therapeutic conditions, of strontium ranelate (SrRan) on the progression of joint structural changes and on the major pathophysiological pathways in an experimental osteoarthritis dog model. METHODS: Dogs underwent sectioning of the anterior cruciate ligament, and 4 weeks after surgery received oral treatment of SrRan 25, 50 or 75 mg/kg per day, or placebo for 12 weeks. Methods included macroscopy, picrosirius red staining, histology, subchondral bone histomorphometry, quantitative PCR, and ELISA for CTX-II level in serum. Strontium plasma and synovial fluid levels were also measured. RESULTS: At steady state, strontium blood exposures were within the clinical therapeutic range of osteoarthritis patients and correlated with strontium concentrations in synovial fluid. SrRan treatment significantly reduced the osteoarthritis cartilage lesions at all doses tested (p≤0.05). Significantly better preservation of the collagen network was also found in SrRan-treated dogs at 50 and 75 mg/kg per day (p=0.03). The osteoarthritis subchondral bone thickening observed in osteoarthritis-placebo dogs was significantly reduced by SrRan at 50 mg/kg per day (p=0.02). The increased gene expression levels of MMP-1, MMP-13 and cathepsin K in osteoarthritis cartilage were all significantly reduced by SrRan at 75 mg/kg per day (p≤0.03) as were, in osteoarthritis synovium, IL-1ß at 50 and 75 mg/kg per day (p=0.05) and MMP-3 at all doses tested (p≤0.02). The serum level of CTX-II was reduced (p≤0.04) by SrRan at 16 weeks in dogs treated with 50 and 75 mg/kg per day. CONCLUSIONS: This study is the first to demonstrate in vivo in an animal model that SrRan reduced the progression of osteoarthritis structural changes. The inhibition of several key proteases as well as IL-1ß may have contributed to the beneficial effect of SrRan.


Assuntos
Antirreumáticos/farmacologia , Cartilagem Articular/efeitos dos fármacos , Compostos Organometálicos/farmacologia , Osteoartrite/tratamento farmacológico , Membrana Sinovial/efeitos dos fármacos , Tiofenos/farmacologia , Animais , Cartilagem Articular/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Cães , Feminino , Expressão Gênica/efeitos dos fármacos , Interleucina-1beta/biossíntese , Osteoartrite/metabolismo , Osteoartrite/patologia , Peptídeo Hidrolases/biossíntese , Reação em Cadeia da Polimerase em Tempo Real , Membrana Sinovial/metabolismo
20.
Arthritis Rheum ; 64(11): 3614-25, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22833446

RESUMO

OBJECTIVE: In vitro activation of the receptor EphB4 positively affects human osteoarthritis (OA) articular cell metabolism. However, the specific in vivo role of this ephrin receptor in OA remains unknown. We investigated in mice the in vivo effect of bone-specific EphB4 overexpression on OA pathophysiology. METHODS: Morphometric, morphologic, and radiologic evaluations were performed on postnatal day 5 (P5) mice and on 10-week-old mice. Knee OA was induced surgically by destabilization of the medial meniscus (DMM) in 10-week-old male EphB4 homozygous transgenic (EphB4-Tg) and wild-type (WT) mice. Medial compartment evaluations of cartilage were performed using histology and immunohistochemistry, and evaluations of subchondral bone using histomorphometry, osteoclast staining, and micro-computed tomography. RESULTS: There was no obvious phenotype difference in skeletal development between EphB4-Tg mice and WT mice at P5 or at 10 weeks. At 8 and 12 weeks post-DMM, the EphB4-Tg mice demonstrated significantly less cartilage alteration in the medial tibial plateau and the femoral condyle than did the WT mice. This was associated with a significant reduction of aggrecan and type II collagen degradation products, type X collagen, and collagen fibril disorganization in the operated EphB4-Tg mice. The medial tibial subchondral bone demonstrated a significant reduction in sclerosis, bone volume, trabecular thickness, and number of tartrate-resistant acid phosphatase-positive osteoclasts at both times assessed post-DMM in the EphB4-Tg mice than in the WT mice. CONCLUSION: This is the first in vivo evidence that bone-specific EphB4 overexpression exerts a protective effect on OA joint structural changes. The findings of this study stress the in vivo importance of subchondral bone biology in cartilage integrity.


Assuntos
Cartilagem Articular/patologia , Fêmur/patologia , Osteoartrite do Joelho , Receptor EphB4/genética , Tíbia/patologia , Animais , Biomarcadores/metabolismo , Cartilagem Articular/imunologia , Cartilagem Articular/metabolismo , Modelos Animais de Doenças , Edema/imunologia , Edema/patologia , Feminino , Fêmur/imunologia , Fêmur/metabolismo , Expressão Gênica/imunologia , Genótipo , Articulação do Joelho/imunologia , Articulação do Joelho/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoartrite do Joelho/genética , Osteoartrite do Joelho/imunologia , Osteoartrite do Joelho/patologia , Fenótipo , Receptor EphB4/imunologia , Tíbia/imunologia , Tíbia/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa