Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Rheumatology (Oxford) ; 59(10): 2816-2828, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32047926

RESUMO

OBJECTIVES: We investigated the reciprocal relationship linking fibroblast-like synoviocytes (FLS) and T lymphocytes in the inflamed RA synovium and subsequently targeted cellular metabolic pathways in FLS to identify key molecular players in joint inflammation. METHODS: RA FLS were cultured with CD4 T cells or T cell conditioned medium (CD4CM); proliferation, expression of adhesion molecules and intracellular cytokines were examined by flow cytometry. FLS invasiveness and secreted cytokines were measured by transwell matrigel invasion chambers and ELISA, while metabolic profiles were determined by extracellular Seahorse flux analysis. Gene expression was quantified by real-time quantitative RT-PCR. RESULTS: Our results showed mutual activation between CD4 T cells and FLS, which resulted in increased proliferation and expression of intercellular adhesion molecule 1 and vascular cell adhesion molecule 1 by both CD4 T cells and FLS. Furthermore, interaction between CD4 T cells and FLS resulted in an increased frequency of TNF-α+, IFN-γ+ and IL-17A+ CD4 T cells and augmented TNF-α, IFN-γ, IL-17A, IL-6, IL-8 and VEGF secretion. Moreover, CD4CM promoted invasiveness and boosted glycolysis in FLS while downregulating oxidative phosphorylation, effects paralleled by increased glucose transporters GLUT1 and GLUT3; key glycolytic enzymes GSK3A, HK2, LDHA and PFKFB3; angiogenic factor VEGF and MMP-3 and MMP-9. Importantly, these effects were reversed by the glycolytic inhibitor 2-DG and AMP analogue 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR). CONCLUSION: This study demonstrates that CD4 T cells elicit an aggressive phenotype in FLS, which subsequently upregulate glycolysis to meet the increased metabolic demand. Accordingly, 2-DG and AICAR prevent this activation, suggesting that glycolytic manipulation could have clinical implications for RA treatment.


Assuntos
Artrite Reumatoide/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Metabolismo Energético , Membrana Sinovial/citologia , Sinoviócitos/metabolismo , Amina Oxidase (contendo Cobre)/metabolismo , Proteínas Angiogênicas/metabolismo , Artrite Reumatoide/imunologia , Linfócitos T CD4-Positivos/fisiologia , Moléculas de Adesão Celular/metabolismo , Ensaios de Migração Celular , Proliferação de Células , Meios de Cultivo Condicionados , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Glicólise/fisiologia , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Interferon gama/metabolismo , Interleucinas/metabolismo , Ativação Linfocitária , Fosforilação Oxidativa , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinoviócitos/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Front Immunol ; 10: 2056, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31555281

RESUMO

Objectives: Oncostatin M (OSM), a pleiotropic cytokine and a member of the gp130/IL-6 cytokine family, has been implicated in the pathogenesis of autoimmune diseases. Here we investigate the mechanisms by which its synergistic interactions with TNFα regulate the cellular bioenergetics and invasive function of synovial cells from patients with Rheumatoid Arthritis. Methods: Primary RA synovial fibroblasts (RAFLS) and human umbilical vein endothelial cells (HUVEC) were cultured with OSM alone or in combination with TNFα. Pro-inflammatory cytokines, angiogenic growth factors and adhesion molecules were quantified by real-time PCR and ELISA. Invasion, angiogenesis and cellular adhesion were quantified by Transwell invasion chambers, Matrigel tube formation assays, and adhesion binding assays. Cellular bioenergetics was assessed using the Seahorse XFe96 Analyser. Key metabolic genes (GLUT-1, HK2, PFKFB3, HIF1α, LDHA, PKM2) and transcription factor STAT3 were measured using real-time PCR and western blot. Results: OSM differentially regulates pro-inflammatory mediators in RAFLS and HUVEC, with IL-6, MCP-1, ICAM-1, and VEGF all significantly induced, in contrast to the observed inhibition of IL-8 and GROα, with opposing effects observed for VCAM-1 depending on cell type. Functionally, OSM significantly induced angiogenic network formation, adhesion, and invasive mechanisms. This was accompanied by a change in the cellular bioenergetic profile of the cells, where OSM significantly increased the ECAR/OCR ratio in favor of glycolysis, paralleled by induction of the glucose transporter GLUT-1 and key glycolytic enzymes (HK2, PFKFB3, HIF1α). OSM synergizes with TNFα to differentially regulate pro-inflammatory mechanisms in RAFLS and HUVEC. Interestingly, OSM differentially synergizes with TNFα to regulate metabolic reprogramming, where induction of glycolytic activity with concomitant attenuation of mitochondrial respiration and ATP activity was demonstrated in RAFLS but not in HUVEC. Finally, we identified a mechanism, whereby the combination of OSM with TNFα induces transcriptional activity of STAT3 only in RAFLS, with no effect observed in HUVEC. Conclusion: STAT3 mediates the differential effects of OSM and TNFα on RAFLS and EC function. Targeting OSM or downstream signaling pathways may lead to new potential therapeutic or adjuvant strategies, particularly for those patients who have sub-optimal responses to TNFi.


Assuntos
Artrite Reumatoide/etiologia , Artrite Reumatoide/metabolismo , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Oncostatina M/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Artrite Reumatoide/patologia , Adesão Celular , Células Cultivadas , Citocinas/metabolismo , Humanos , Janus Quinases/metabolismo , Neovascularização Fisiológica , Fosforilação , Transdução de Sinais , Membrana Sinovial
3.
Nat Commun ; 8: 15040, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28508891

RESUMO

The stem cells that safeguard synovial joints in adulthood are undefined. Studies on mesenchymal stromal/stem cells (MSCs) have mainly focused on bone marrow. Here we show that lineage tracing of Gdf5-expressing joint interzone cells identifies in adult mouse synovium an MSC population largely negative for the skeletal stem cell markers Nestin-GFP, Leptin receptor and Gremlin1. Following cartilage injury, Gdf5-lineage cells underpin synovial hyperplasia through proliferation, are recruited to a Nestin-GFPhigh perivascular population, and contribute to cartilage repair. The transcriptional co-factor Yap is upregulated after injury, and its conditional ablation in Gdf5-lineage cells prevents synovial lining hyperplasia and decreases contribution of Gdf5-lineage cells to cartilage repair. Cultured Gdf5-lineage cells exhibit progenitor activity for stable chondrocytes and are able to self-organize three-dimensionally to form a synovial lining-like layer. Finally, human synovial MSCs transduced with Bmp7 display morphogenetic properties by patterning a joint-like organ in vivo. Our findings further the understanding of the skeletal stem/progenitor cells in adult life.


Assuntos
Cartilagem Articular/fisiologia , Condrócitos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Regeneração/fisiologia , Membrana Sinovial/citologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Animais , Cartilagem Articular/citologia , Cartilagem Articular/lesões , Proteínas de Ciclo Celular , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Feminino , Fator 5 de Diferenciação de Crescimento/metabolismo , Humanos , Hiperplasia/fisiopatologia , Artropatias/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese/fisiologia , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Membrana Sinovial/lesões , Membrana Sinovial/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Sinalização YAP
4.
Curr Opin Rheumatol ; 29(2): 201-207, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27941390

RESUMO

PURPOSE OF REVIEW: Mesenchymal stromal/stem cells (MSCs) have potent anti-inflammatory and immunomodulatory properties, in addition to their ability to form cartilage and bone. The purpose of this review is to highlight recent developments and current knowledge gaps in our understanding of the protective effects of MSCs against inflammatory arthritis, and to discuss their clinical exploitation for the treatment of rheumatoid arthritis (RA). RECENT FINDINGS: The weight of evidence for protective mechanisms of exogenously administered MSCs is on immunomodulatory effects, including inhibition of dendritic cell maturation, polarization of macrophages to an anti-inflammatory phenotype, and activation of regulatory T cells, thereby dampening inflammation and preventing joint damage. Evidence for direct effects on tissue repair is scant. Recent studies have identified MSC subsets in vivo and an important question is whether MSCs in their native tissues have similar immunoregulatory functions. Recent proof-of-concept clinical studies have shown a satisfactory safety profile of allogeneic MSC therapy in RA patients with promising trends for clinical efficacy. SUMMARY: Allogeneic MSCs could be effective in RA. Larger, multicentre clinical studies are needed to provide robust evidence, and MSC treatment at early stages of RA should be explored to 'reset' the immune system.


Assuntos
Artrite Reumatoide/terapia , Transplante de Células-Tronco Mesenquimais , Artrite Reumatoide/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Humanos , Inflamação , Ativação Linfocitária/imunologia , Células-Tronco Mesenquimais/imunologia , Linfócitos T Reguladores/imunologia
5.
Ann Biomed Eng ; 44(5): 1832-44, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26438451

RESUMO

Cartilage tissue engineering is a multifactorial problem requiring a wide range of material property requirements from provision of biological cues to facilitation of mechanical support in load-bearing diarthrodial joints. The study aim was to design, fabricate and characterize a template to promote endogenous cell recruitment for enhanced cartilage repair. A polylactic acid poly-ε-caprolactone (PLCL) support structure was fabricated using laser micromachining technology and thermal crimping to create a functionally-graded open pore network scaffold with a compressive modulus of 9.98 ± 1.41 MPa and a compressive stress at 50% strain of 8.59 ± 1.35 MPa. In parallel, rabbit mesenchymal stem cells were isolated and their growth characteristics, morphology and multipotency confirmed. Sterilization had no effect on construct chemical structure and cellular compatibility was confirmed. After four weeks implantation in an osteochondral defect in a rabbit model to assess biocompatibility, there was no evidence of inflammation or giant cells. Moreover, acellular constructs performed better than cell-seeded constructs with endogenous progenitor cells homing through microtunnels, differentiating to form neo-cartilage and strengthening integration with native tissue. These results suggest, albeit at an early stage of repair, that by modulating the architecture of a macroporous scaffold, pre-seeding with MSCs is not necessary for hyaline cartilage repair.


Assuntos
Substitutos Ósseos/química , Cartilagem Hialina , Teste de Materiais , Células-Tronco Mesenquimais/metabolismo , Poliésteres/química , Tíbia , Alicerces Teciduais/química , Animais , Modelos Animais de Doenças , Cartilagem Hialina/lesões , Cartilagem Hialina/metabolismo , Cartilagem Hialina/patologia , Masculino , Células-Tronco Mesenquimais/patologia , Porosidade , Coelhos , Tíbia/lesões , Tíbia/metabolismo , Tíbia/patologia
6.
J Control Release ; 179: 42-51, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24491910

RESUMO

Human mesenchymal stem cells (hMSCs) have been identified as a viable cell source for cartilage tissue engineering. However, to undergo chondrogenic differentiation hMSCs require growth factors, in particular members of the transforming growth factor beta (TGF-ß) family. While in vitro differentiation is feasible through continuous supplementation of TGF-ß3, mechanisms to control and drive hMSCs down the chondrogenic lineage in their native microenvironment remain a significant challenge. The release of TGF-ß3 from an injectable microsphere composed of the cartilage-associated extracellular matrix molecule hyaluronan represents a readily translatable approach for in situ differentiation of hMSCs for cartilage repair. In this study, chondromimetic hyaluronan microspheres were used as a growth factor delivery source for hMSC chondrogenesis. Cellular compatibility of the microspheres (1.2 and 14.1 µm) with hMSCs was shown and release of TGF-ß3 from the most promising 14.1 µm microspheres to control differentiation of hMSCs was evaluated. Enhanced accumulation of cartilage-associated glycosaminoglycans by hMSCs incubated with TGF-ß3-loaded microspheres was seen and positive staining for collagen type II and proteoglycan confirmed successful in vitro chondrogenesis. Gene expression analysis showed significantly increased expression of the chondrocyte-associated genes, collagen type II and aggrecan. This delivery platform resulted in significantly less collagen type X expression, suggesting the generation of a more stable cartilage phenotype. When evaluated in an ex vivo osteoarthritic cartilage model, implanted hMSCs with TGF-ß3-loaded HA microspheres were detected within cartilage fibrillations and increased proteoglycan staining was seen in the tissue. In summary, data presented here demonstrate that TGF-ß3-bound hyaluronan microspheres provide a suitable delivery system for induction of hMSC chondrogenesis and their use may represent a clinically feasible tissue engineering approach for the treatment of articular cartilage defects.


Assuntos
Biomimética , Diferenciação Celular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Portadores de Fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Engenharia Tecidual , Fator de Crescimento Transformador beta3/farmacologia , Adolescente , Adulto , Agrecanas/genética , Agrecanas/metabolismo , Animais , Linhagem Celular , Condrócitos/metabolismo , Condrócitos/transplante , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Colágeno Tipo X/metabolismo , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Humanos , Ácido Hialurônico/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Microesferas , Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia , Osteoartrite do Joelho/terapia , Fenótipo , Fatores de Tempo , Fator de Crescimento Transformador beta3/toxicidade , Adulto Jovem
7.
Eur Cell Mater ; 23: 310-8; discussion 318-9, 2012 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-22522285

RESUMO

Multipotent mesenchymal stem cells (MSCs) have been suggested as a suitable cell source for cell-based treatments for diseases such as osteoarthritis due to their ability to differentiate towards chondrogenic and osteogenic lineages. MSCs can be obtained from a variety of tissue sources, are scalable for mass-production and immuno-privileged enabling their use for allogeneic cell therapy. However, recent pre-clinical studies and clinical trials point to the necessity of increasing engraftment and efficacy of MSCs. This review explores how cell surface modification of the cells can improve homing of MSCs and summarises the use of nanoparticles to enable gene delivery by stem cells as well as facilitate in vivo imaging. The use of advanced biomaterials and how they can be applied to reduce the overall dose of MSCs during therapeutic interventions while achieving optimal targeting efficiency of cells to the diseased sites are addressed. Particular attention is paid to methods that improve engraftment of MSCs to cartilage and research describing combinatorial approaches of particle-based cell therapies for improved regeneration of this tissue is reviewed. The use of such approaches will add to the array of potential regenerative therapeutics for treatment of osteoarthritis.


Assuntos
Diferenciação Celular , Condrócitos/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Osteoartrite/cirurgia , Animais , Cartilagem/patologia , Cartilagem/cirurgia , Humanos , Medicina Regenerativa/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...