Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Ann Rheum Dis ; 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38684323

RESUMO

Giant cell arteritis (GCA), the most common systemic vasculitis, is characterised by aberrant interactions between infiltrating and resident cells of the vessel wall. Ageing and breach of tolerance are prerequisites for GCA development, resulting in dendritic and T-cell dysfunction. Inflammatory cytokines polarise T-cells, activate resident macrophages and synergistically enhance vascular inflammation, providing a loop of autoreactivity. These events originate in the adventitia, commonly regarded as the biological epicentre of the vessel wall, with additional recruitment of cells that infiltrate and migrate towards the intima. Thus, GCA-vessels exhibit infiltrates across the vascular layers, with various cytokines and growth factors amplifying the pathogenic process. These events activate ineffective repair mechanisms, where dysfunctional vascular smooth muscle cells and fibroblasts phenotypically shift along their lineage and colonise the intima. While high-dose glucocorticoids broadly suppress these inflammatory events, they cause well known deleterious effects. Despite the emerging targeted therapeutics, disease relapse remains common, affecting >50% of patients. This may reflect a discrepancy between systemic and local mediators of inflammation. Indeed, temporal arteries and aortas of GCA-patients can show immune-mediated abnormalities, despite the treatment induced clinical remission. The mechanisms of persistence of vascular disease in GCA remain elusive. Studies in other chronic inflammatory diseases point to the fibroblasts (and their lineage cells including myofibroblasts) as possible orchestrators or even effectors of disease chronicity through interactions with immune cells. Here, we critically review the contribution of immune and stromal cells to GCA pathogenesis and analyse the molecular mechanisms by which these would underpin the persistence of vascular disease.

2.
Osteoarthritis Cartilage ; 32(2): 148-158, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37944663

RESUMO

Great progress continues to be made in our understanding of the multiple facets of osteoarthritis (OA) biology. Here, we review the major advances in this field and progress towards therapy development over the past year, highlighting a selection of relevant published literature from a PubMed search covering the year from the end of April 2022 to the end of April 2023. The selected articles have been arranged in themes. These include 1) molecular regulation of articular cartilage and implications for OA, 2) mechanisms of subchondral bone remodelling, 3) role of synovium and inflammation, 4) role of age-related changes including cartilage matrix stiffening, cellular senescence, mitochondrial dysfunction, metabolic dysfunction, and impaired autophagy, and 5) peripheral mechanisms of OA pain. Progress in the understanding of the cellular and molecular mechanisms responsible for the multiple aspects of OA biology is unravelling novel therapeutic targets for disease modification.


Assuntos
Cartilagem Articular , Osteoartrite , Humanos , Osteoartrite/metabolismo , Inflamação/metabolismo , Cartilagem Articular/metabolismo , Osso e Ossos/metabolismo , Biologia
3.
Ann Rheum Dis ; 82(3): 428-437, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36414376

RESUMO

OBJECTIVES: Fibroblasts in synovium include fibroblast-like synoviocytes (FLS) in the lining and Thy1+ connective-tissue fibroblasts in the sublining. We aimed to investigate their developmental origin and relationship with adult progenitors. METHODS: To discriminate between Gdf5-lineage cells deriving from the embryonic joint interzone and other Pdgfrα-expressing fibroblasts and progenitors, adult Gdf5-Cre;Tom;Pdgfrα-H2BGFP mice were used and cartilage injury was induced to activate progenitors. Cells were isolated from knees, fibroblasts and progenitors were sorted by fluorescence-activated cell-sorting based on developmental origin, and analysed by single-cell RNA-sequencing. Flow cytometry and immunohistochemistry were used for validation. Clonal-lineage mapping was performed using Gdf5-Cre;Confetti mice. RESULTS: In steady state, Thy1+ sublining fibroblasts were of mixed ontogeny. In contrast, Thy1-Prg4+ lining fibroblasts predominantly derived from the embryonic joint interzone and included Prg4-expressing progenitors distinct from molecularly defined FLS. Clonal-lineage tracing revealed compartmentalisation of Gdf5-lineage fibroblasts between lining and sublining. Following injury, lining hyperplasia resulted from proliferation and differentiation of Prg4-expressing progenitors, with additional recruitment of non-Gdf5-lineage cells, into FLS. Consistent with this, a second population of proliferating cells, enriched near blood vessels in the sublining, supplied activated multipotent cells predicted to give rise to Thy1+ fibroblasts, and to feed into the FLS differentiation trajectory. Transcriptional programmes regulating fibroblast differentiation trajectories were uncovered, identifying Sox5 and Foxo1 as key FLS transcription factors in mice and humans. CONCLUSIONS: Our findings blueprint a cell atlas of mouse synovial fibroblasts and progenitors in healthy and injured knees, and provide novel insights into the cellular and molecular principles governing the organisation and maintenance of adult synovial joints.


Assuntos
Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Sinoviócitos , Humanos , Adulto , Camundongos , Animais , Articulações , Membrana Sinovial , Fibroblastos
4.
Osteoarthritis Cartilage ; 31(9): 1154-1155, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37196976

RESUMO

In December 2022, Gerwin et al published in Nature Medicine that the C-terminal portion of angiopoietin-like 3, called LNA043, has chondroprotective and cartilage-regenerative properties. Molecular data from an experimental medicine phase I study suggested potential efficacy in humans. Here, we respond to and complement a commentary from Vincent and Conaghan and discuss unresolved issues and the potential of this molecule as a disease-modifying osteoarthritis drug.


Assuntos
Cartilagem Articular , Osteoartrite , Humanos , Cartilagem , Ensaios Clínicos Fase I como Assunto
5.
Osteoarthritis Cartilage ; 31(11): 1454-1468, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37392862

RESUMO

OBJECTIVE: To explore the significance of BMP signaling in osteoarthritis (OA) etiology, and thereafter propose a disease-modifying therapy for OA. METHODS: To examine the role of the BMP signaling in pathogenesis of OA, an Anterior Cruciate Ligament Transection (ACLT) surgery was performed to incite OA in C57BL/6J mouse line at postnatal day 120 (P120). Thereafter, to investigate whether activation of BMP signaling is necessary and sufficient to induce OA, we have used conditional gain- and loss-of-function mouse lines in which BMP signaling can be activated or depleted, respectively, upon intraperitoneal injection of tamoxifen. Finally, we locally inhibited BMP signaling through intra-articular injection of LDN-193189 pre- and post-onset surgically induced OA. The majority of the investigation has been conducted using micro-CT, histological staining, and immuno histochemistry to assess the disease etiology. RESULTS: Upon induction of OA, depletion of SMURF1-an intra-cellular BMP signaling inhibitor in articular cartilage coincided with the activation of BMP signaling, as measured by pSMAD1/5/9 expression. In mouse articular cartilage, the BMP gain-of-function mutation is sufficient to induce OA even without surgery. Further, genetic, or pharmacological BMP signaling suppression also prevented pathogenesis of OA. Interestingly, inflammatory indicators were also significantly reduced upon LDN-193189 intra-articular injection which inhibited BMP signaling and slowed OA progression post onset. CONCLUSION: Our findings showed that BMP signaling is crucial to the etiology of OA and inhibiting BMP signaling locally can be a potent strategy for alleviating OA.


Assuntos
Cartilagem Articular , Osteoartrite do Joelho , Camundongos , Animais , Osteoartrite do Joelho/patologia , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Ligamento Cruzado Anterior/cirurgia , Ligamento Cruzado Anterior/metabolismo , Cartilagem Articular/patologia
6.
Ann Rheum Dis ; 81(2): 214-224, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34844926

RESUMO

OBJECTIVE: We aimed to understand the role of the transcriptional co-factor Yes-associated protein (Yap) in the molecular pathway underpinning the pathogenic transformation of synovial fibroblasts (SF) in rheumatoid arthritis (RA) to become invasive and cause joint destruction. METHODS: Synovium from patients with RA and mice with antigen-induced arthritis (AIA) was analysed by immunostaining and qRT-PCR. SF were targeted using Pdgfrα-CreER and Gdf5-Cre mice, crossed with fluorescent reporters for cell tracing and Yap-flox mice for conditional Yap ablation. Fibroblast phenotypes were analysed by flow cytometry, and arthritis severity was assessed by histology. Yap activation was detected using Yap-Tead reporter cells and Yap-Snail interaction by proximity ligation assay. SF invasiveness was analysed using matrigel-coated transwells. RESULTS: Yap, its binding partner Snail and downstream target connective tissue growth factor were upregulated in hyperplastic human RA and in mouse AIA synovium, with Yap detected in SF but not macrophages. Lineage tracing showed polyclonal expansion of Pdgfrα-expressing SF during AIA, with predominant expansion of the Gdf5-lineage SF subpopulation descending from the embryonic joint interzone. Gdf5-lineage SF showed increased expression of Yap and adopted an erosive phenotype (podoplanin+Thy-1 cell surface antigen-), invading cartilage and bone. Conditional ablation of Yap in Gdf5-lineage cells or Pdgfrα-expressing fibroblasts ameliorated AIA. Interleukin (IL)-6, but not tumour necrosis factor alpha (TNF-α) or IL-1ß, Jak-dependently activated Yap and induced Yap-Snail interaction. SF invasiveness induced by IL-6 stimulation or Snail overexpression was prevented by Yap knockdown, showing a critical role for Yap in SF transformation in RA. CONCLUSIONS: Our findings uncover the IL-6-Yap-Snail signalling axis in pathogenic SF in inflammatory arthritis.


Assuntos
Artrite Reumatoide/patologia , Fibroblastos/patologia , Membrana Sinovial/patologia , Proteínas de Sinalização YAP/metabolismo , Animais , Artrite Experimental/patologia , Artrite Reumatoide/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Interleucina-6/metabolismo , Camundongos , Transdução de Sinais/fisiologia , Fatores de Transcrição da Família Snail/metabolismo , Membrana Sinovial/metabolismo
7.
Ann Rheum Dis ; 79(12): 1625-1634, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32963046

RESUMO

OBJECTIVES: Osteophytes are highly prevalent in osteoarthritis (OA) and are associated with pain and functional disability. These pathological outgrowths of cartilage and bone typically form at the junction of articular cartilage, periosteum and synovium. The aim of this study was to identify the cells forming osteophytes in OA. METHODS: Fluorescent genetic cell-labelling and tracing mouse models were induced with tamoxifen to switch on reporter expression, as appropriate, followed by surgery to induce destabilisation of the medial meniscus. Contributions of fluorescently labelled cells to osteophytes after 2 or 8 weeks, and their molecular identity, were analysed by histology, immunofluorescence staining and RNA in situ hybridisation. Pdgfrα-H2BGFP mice and Pdgfrα-CreER mice crossed with multicolour Confetti reporter mice were used for identification and clonal tracing of mesenchymal progenitors. Mice carrying Col2-CreER, Nes-CreER, LepR-Cre, Grem1-CreER, Gdf5-Cre, Sox9-CreER or Prg4-CreER were crossed with tdTomato reporter mice to lineage-trace chondrocytes and stem/progenitor cell subpopulations. RESULTS: Articular chondrocytes, or skeletal stem cells identified by Nes, LepR or Grem1 expression, did not give rise to osteophytes. Instead, osteophytes derived from Pdgfrα-expressing stem/progenitor cells in periosteum and synovium that are descendants from the Gdf5-expressing embryonic joint interzone. Further, we show that Sox9-expressing progenitors in periosteum supplied hybrid skeletal cells to the early osteophyte, while Prg4-expressing progenitors from synovial lining contributed to cartilage capping the osteophyte, but not to bone. CONCLUSION: Our findings reveal distinct periosteal and synovial skeletal progenitors that cooperate to form osteophytes in OA. These cell populations could be targeted in disease modification for treatment of OA.


Assuntos
Osteoartrite/patologia , Osteófito/patologia , Periósteo/patologia , Células-Tronco/patologia , Membrana Sinovial/patologia , Animais , Linhagem da Célula , Camundongos
8.
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
9.
Ann Rheum Dis ; 75(7): 1386-91, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26275430

RESUMO

BACKGROUND: Myeloid inhibitory C-type lectin-like receptor (MICL, Clec12A) is a C-type lectin receptor (CLR) expressed predominantly by myeloid cells. Previous studies have suggested that MICL is involved in controlling inflammation. OBJECTIVE: To determine the role of this CLR in inflammatory pathology using Clec12A(-/-) mice. METHODS: Clec12A(-/-) mice were generated commercially and primarily characterised using the collagen antibody-induced arthritis (CAIA) model. Mechanisms and progress of disease were characterised by clinical scoring, histology, flow cytometry, irradiation bone-marrow chimera generation, administration of blocking antibodies and in vivo imaging. Characterisation of MICL in patients with rheumatoid arthritis (RA) was determined by immunohistochemistry and single nucleotide polymorphism analysis. Anti-MICL antibodies were detected in patient serum by ELISA and dot-blot analysis. RESULTS: MICL-deficient animals did not present with pan-immune dysfunction, but exhibited markedly exacerbated inflammation during CAIA, owing to the inappropriate activation of myeloid cells. Polymorphisms of MICL were not associated with disease in patients with RA, but this CLR was the target of autoantibodies in a subset of patients with RA. In wild-type mice the administration of such antibodies recapitulated the Clec12A(-/-) phenotype. CONCLUSIONS: MICL plays an essential role in regulating inflammation during arthritis and is an autoantigen in a subset of patients with RA. These data suggest an entirely new mechanism underlying RA pathogenesis, whereby the threshold of myeloid cell activation can be modulated by autoantibodies that bind to cell membrane-expressed inhibitory receptors.


Assuntos
Artrite Experimental/genética , Artrite Reumatoide/genética , Lectinas Tipo C/fisiologia , Receptores Mitogênicos/fisiologia , Animais , Artrite Reumatoide/sangue , Artrite Reumatoide/etiologia , Artrite Reumatoide/patologia , Autoanticorpos/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Lectinas Tipo C/deficiência , Lectinas Tipo C/imunologia , Camundongos , Células Mieloides/metabolismo , Polimorfismo Genético , Receptores Mitogênicos/deficiência , Receptores Mitogênicos/imunologia , Membrana Sinovial/patologia
10.
Ann Rheum Dis ; 74(12): 2207-15, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25135253

RESUMO

OBJECTIVE: ELR+ CXC chemokines are heparin-binding cytokines signalling through the CXCR1 and CXCR2 receptors. ELR+ CXC chemokines have been associated with inflammatory arthritis due to their capacity to attract inflammatory cells. Here, we describe an unsuspected physiological function of these molecules in articular cartilage homeostasis. METHODS: Chemokine receptors and ligands were detected by immunohistochemistry, western blotting and RT-PCR. Osteoarthritis was induced in wild-type and CXCR2(-/-) mice by destabilisation of the medial meniscus (DMM). CXCR1/2 signalling was inhibited in vitro using blocking antibodies or siRNA. Chondrocyte phenotype was analysed using Alcian blue staining, RT-PCR and western blotting. AKT phosphorylation and SOX9 expression were upregulated using constitutively active AKT or SOX9 plasmids. Apoptosis was detected by terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay. RESULTS: CXCL6 was expressed in healthy cartilage and was retained through binding to heparan sulfate proteoglycans. CXCR2(-/-) mice developed more severe osteoarthritis than wild types following DMM, with increased chondrocyte apoptosis. Disruption of CXCR1/2 in human and CXCR2 signalling in mouse chondrocytes led to a decrease in extracellular matrix production, reduced expression of chondrocyte differentiation markers and increased chondrocyte apoptosis. CXCR2-dependent chondrocyte homeostasis was mediated by AKT signalling since forced expression of constitutively active AKT rescued the expression of phenotypic markers and the apoptosis induced by CXCR2 blockade. CONCLUSIONS: Our study demonstrates an important physiological role for CXCR1/2 signalling in maintaining cartilage homeostasis and suggests that the loss of ELR+ CXC chemokines during cartilage breakdown in osteoarthritis contributes to the characteristic loss of chondrocyte phenotypic stability.


Assuntos
Cartilagem Articular/metabolismo , Osteoartrite/metabolismo , Receptores de Interleucina-8B/metabolismo , Animais , Apoptose , Western Blotting , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos/metabolismo , Condrócitos/patologia , Modelos Animais de Doenças , Homeostase , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais
11.
Rheumatology (Oxford) ; 54(2): 210-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25288785

RESUMO

Mesenchymal stem cells (MSCs) are multipotent cells with the capacity to undergo chondrogenic differentiation. Systemically administered MSCs have been shown to preferentially accumulate at sites of tissue damage and inflammation, thus MSC-based therapy holds great promise for the treatment of inflammatory diseases such as RA. Modulation of MSC homing may allow targeted delivery of systemically administered MSCs to damaged articular cartilage, where they can suppress immune-mediated cartilage destruction and contribute to cartilage repair via a combination of chondrogenic differentiation and paracrine stimulation of intrinsic residual repair. To harness the potential of MSC homing, a thorough understanding of the mechanism is key. This review discusses current knowledge of the mechanism of MSC homing to injured/inflamed tissue and its implications for targeted MSC-based therapy in arthritis.


Assuntos
Artrite Reumatoide/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Osteoartrite/terapia , Comunicação Celular/fisiologia , Movimento Celular/fisiologia , Quimiocinas/fisiologia , Humanos , Proteínas de Membrana/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia
12.
J Cell Sci ; 125(Pt 24): 6009-19, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23038772

RESUMO

Satellite cells are the resident stem cells of skeletal muscle. Mitotically quiescent in mature muscle, they can be activated to proliferate and generate myoblasts to supply further myonuclei to hypertrophying or regenerating muscle fibres, or self-renew to maintain the resident stem cell pool. Here, we identify the transcriptional co-factor Yap as a novel regulator of satellite cell fate decisions. Yap expression increases during satellite cell activation and Yap remains highly expressed until after the differentiation versus self-renewal decision is made. Constitutive expression of Yap maintains Pax7(+) and MyoD(+) satellite cells and satellite cell-derived myoblasts, promotes proliferation but prevents differentiation. In contrast, Yap knockdown reduces the proliferation of satellite cell-derived myoblasts by ≈40%. Consistent with the cellular phenotype, microarrays show that Yap increases expression of genes associated with Yap inhibition, the cell cycle, ribosome biogenesis and that it represses several genes associated with angiotensin signalling. We also identify known regulators of satellite cell function such as BMP4, CD34 and Myf6 (Mrf4) as genes whose expression is dependent on Yap activity. Finally, we confirm in myoblasts that Yap binds to Tead transcription factors and co-activates MCAT elements which are enriched in the proximal promoters of Yap-responsive genes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular , Processos de Crescimento Celular/fisiologia , Núcleo Celular/metabolismo , Embrião de Galinha , Via de Sinalização Hippo , Cavalos , Camundongos , Fosfoproteínas/genética , Transdução de Sinais , Transfecção , Proteínas de Sinalização YAP
13.
Proc Natl Acad Sci U S A ; 108(16): 6503-8, 2011 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-21464310

RESUMO

In many adult tissues, mesenchymal stem cells (MSCs) are closely associated with perivascular niches and coexpress many markers in common with pericytes. The ability of pericytes to act as MSCs, however, remains controversial. By using genetic lineage tracing, we show that some pericytes differentiate into specialized tooth mesenchyme-derived cells--odontoblasts--during tooth growth and in response to damage in vivo. As the pericyte-derived mesenchymal cell contribution to odontoblast differentiation does not account for all cell differentiation, we identify an additional source of cells with MSC-like properties that are stimulated to migrate toward areas of tissue damage and differentiate into odontoblasts. Thus, although pericytes are capable of acting as a source of MSCs and differentiating into cells of mesenchymal origin, they do so alongside other MSCs of a nonpericyte origin. This study identifies a dual origin of MSCs in a single tissue and suggests that the pericyte contribution to MSC-derived mesenchymal cells in any given tissue is variable and possibly dependent on the extent of the vascularity.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais , Odontoblastos , Pericitos , Regeneração/fisiologia , Dente , Animais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Transgênicos , Odontoblastos/citologia , Odontoblastos/fisiologia , Pericitos/citologia , Pericitos/fisiologia , Dente/citologia , Dente/crescimento & desenvolvimento
14.
Rheumatology (Oxford) ; 52(10): 1758-68, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23804221

RESUMO

OBJECTIVE: We previously reported the coexistence, within cultured mesenchymal stem cells (MSCs) from human synovial membrane, of single-cell-derived clonal cell populations with distinct differentiation potency. The aim of this study was to investigate marker sets for prospective purification of functionally distinct MSC subsets. METHODS: Cells were enzymatically released from human synovium and culture expanded. Phenotype analysis was performed by flow cytometry using combinations of MSC markers. Sorting was carried out using the FACS DiVA cell sorter. Sorted cell populations were assessed for clonogenicity, kinetics of growth, cell senescence and chondro-osteogenic potency. RESULTS: During culture expansion, the co-localization of CD39 within the CD73(+) cell population identified a small cell subset that was maintained from passage 1 (P1) up to at least P12 in all donors tested. The CD73(+)CD39(+) cell subset displayed higher expression levels of Sox9 and Runx2 and a significantly greater chondro-osteogenic potency than the CD73(+)CD39(-) cell subset. In contrast, it was less clonogenic and proliferative. There was no difference in cell senescence between the sorted MSC subsets and the parental MSCs. Notably, there were no detectable differences in chondro-osteogenic potency between the CD73(+)CD39(-) and CD73(+)CD39(+) cell subsets purified from fresh synovial cell populations. CONCLUSION: Our findings indicate that the combination of CD73 and CD39 allows the prospective purification from culture-expanded heterogeneous synovial MSC populations of a distinct MSC subset with greater chondro-osteogenic potency. We anticipate that such an approach will enhance the consistency of cell-based therapeutic protocols for the repair of osteochondral defects.


Assuntos
Condrogênese/fisiologia , Células-Tronco Mesenquimais/fisiologia , Osteogênese/fisiologia , Membrana Sinovial/citologia , 5'-Nucleotidase/análise , Antígenos CD/análise , Apirase/análise , Técnicas de Cultura de Células/métodos , Diferenciação Celular/imunologia , Separação Celular/métodos , Ensaio de Unidades Formadoras de Colônias , Estudos de Viabilidade , Citometria de Fluxo/métodos , Proteínas Ligadas por GPI/análise , Humanos , Células-Tronco Mesenquimais/imunologia , Estudos Prospectivos , Membrana Sinovial/imunologia
15.
J Bone Miner Res ; 38(5): 792-807, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36824055

RESUMO

Lipids play a crucial role in signaling and metabolism, regulating the development and maintenance of the skeleton. Membrane lipids have been hypothesized to act as intermediates upstream of orphan phosphatase 1 (PHOSPHO1), a major contributor to phosphate generation required for bone mineralization. Here, we spatially resolve the lipid atlas of the healthy mouse knee and demonstrate the effects of PHOSPHO1 ablation on the growth plate lipidome. Lipids spanning 17 subclasses were mapped across the knee joints of healthy juvenile and adult mice using matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS), with annotation supported by shotgun lipidomics. Multivariate analysis identified 96 and 80 lipid ions with differential abundances across joint tissues in juvenile and adult mice, respectively. In both ages, marrow was enriched in phospholipid platelet activating factors (PAFs) and related metabolites, cortical bone had a low lipid content, whereas lysophospholipids were strikingly enriched in the growth plate, an active site of mineralization and PHOSPHO1 activity. Spatially-resolved profiling of PHOSPHO1-knockout (KO) mice across the resting, proliferating, and hypertrophic growth plate zones revealed 272, 306, and 296 significantly upregulated, and 155, 220, and 190 significantly downregulated features, respectively, relative to wild-type (WT) controls. Of note, phosphatidylcholine, lysophosphatidylcholine, sphingomyelin, lysophosphatidylethanolamine, and phosphatidylethanolamine derived lipid ions were upregulated in PHOSPHO1-KO versus WT. Our imaging pipeline has established a spatially-resolved lipid signature of joint tissues and has demonstrated that PHOSPHO1 ablation significantly alters the growth plate lipidome, highlighting an essential role of the PHOSPHO1-mediated membrane phospholipid metabolism in lipid and bone homeostasis. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Assuntos
Lipidômica , Monoéster Fosfórico Hidrolases , Camundongos , Animais , Monoéster Fosfórico Hidrolases/metabolismo , Lâmina de Crescimento/metabolismo , Camundongos Knockout , Homeostase , Fosfolipídeos
16.
Arthritis Rheum ; 63(5): 1289-300, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21538315

RESUMO

OBJECTIVE: We previously reported that human synovium contains cells that, after culture expansion, display properties of mesenchymal stem cells (MSCs). The objective of this study was to identify MSCs in native synovium in vivo. METHODS: To identify stem cells in the synovium in vivo, a double nucleoside analog cell-labeling scheme was used in a mouse model of joint-surface injury. For labeling of slow-cycling cells, mice received iododeoxyuridine (IdU) for 30 days, followed by a 40-day washout period. For labeling of cells that proliferate after injury, mice underwent knee surgery to produce an articular cartilage defect and received chlorodeoxyuridine (CIdU) for 4 days, starting at multiple time points after surgery. Unoperated and sham-operated joints served as controls. Knee joint paraffin sections were analyzed by double and triple immunostaining to detect nucleoside analogs, conventional MSC markers, and chondrocyte-lineage markers. RESULTS: Long-term-retaining, slow-cycling IdU-positive cells were detected in the synovium. At 4 days and 8 days after injury, there was marked proliferation of IdU-positive cells, which costained for CIdU. IdU-positive cells were nonhematopoietic, nonendothelial stromal cells, were distinct from pericytes, and stained positive for MSC markers. MSCs were phenotypically heterogeneous and located in topographically distinct niches in the lining layer and the subsynovial tissue. Twelve days after injury, double nucleoside-labeled cells within synovium were embedded in cartilage-specific metachromatic extracellular matrix and costained positive for the chondrocyte-lineage markers Sox9 and type II collagen. CONCLUSION: Our findings provide the first evidence of the existence of resident MSCs in the knee joint synovium that undergo proliferation and chondrogenic differentiation following injury in vivo.


Assuntos
Condrogênese/fisiologia , Articulação do Joelho/citologia , Células-Tronco Mesenquimais/citologia , Nicho de Células-Tronco/citologia , Membrana Sinovial/citologia , Animais , Contagem de Células , Proliferação de Células , Imuno-Histoquímica , Articulação do Joelho/fisiologia , Camundongos , Nicho de Células-Tronco/fisiologia , Membrana Sinovial/fisiologia
17.
Cell Stem Cell ; 29(4): 528-544.e9, 2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-35276096

RESUMO

The autonomic nervous system is a master regulator of homeostatic processes and stress responses. Sympathetic noradrenergic nerve fibers decrease bone mass, but the role of cholinergic signaling in bone has remained largely unknown. Here, we describe that early postnatally, a subset of sympathetic nerve fibers undergoes an interleukin-6 (IL-6)-induced cholinergic switch upon contacting the bone. A neurotrophic dependency mediated through GDNF-family receptor-α2 (GFRα2) and its ligand, neurturin (NRTN), is established between sympathetic cholinergic fibers and bone-embedded osteocytes, which require cholinergic innervation for their survival and connectivity. Bone-lining osteoprogenitors amplify and propagate cholinergic signals in the bone marrow (BM). Moderate exercise augments trabecular bone partly through an IL-6-dependent expansion of sympathetic cholinergic nerve fibers. Consequently, loss of cholinergic skeletal innervation reduces osteocyte survival and function, causing osteopenia and impaired skeletal adaptation to moderate exercise. These results uncover a cholinergic neuro-osteocyte interface that regulates skeletogenesis and skeletal turnover through bone-anabolic effects.


Assuntos
Interleucina-6 , Osteogênese , Colinérgicos , Fibras Colinérgicas , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia
18.
J Cell Mol Med ; 15(5): 1054-65, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21199329

RESUMO

Cell-based tissue repair of the tooth and - tooth-supporting - periodontal ligament (PDL) is a new attractive approach that complements traditional restorative or surgical techniques for replacement of injured or pathologically damaged tissues. In such therapeutic approaches, stem cells and/or progenitor cells are manipulated in vitro and administered to patients as living and dynamic biological agents. In this review, we discuss the clonogenic potential of human dental and periodontal tissues such as the dental pulp and the PDL and their potential for tooth and periodontal repair and/or regeneration. We propose novel therapeutic approaches using stem cells or progenitor cells, which are targeted to regenerate the lost dental or periodontal tissue.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Doenças Periodontais/terapia , Regeneração , Engenharia Tecidual/métodos , Dente/fisiologia , Animais , Odontologia , Humanos , Doenças Periodontais/patologia
19.
Eur Cell Mater ; 22: 344-58, 2011 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-22125259

RESUMO

At sites of bone fracture, naturally-occurring electric fields (EFs) exist during healing and may guide cell migration. In this study, we investigated whether EFs could direct the migration of bone marrow mesenchymal stem cells (BM-MSCs), which are known to be key players in bone formation. Human BM-MSCs were cultured in direct current EFs of 10 to 600 mV/mm. Using time-lapse microscopy, we demonstrated that an EF directed migration of BM-MSCs mainly to the anode. Directional migration occurred at a low threshold and with a physiological EF of ~25 mV/mm. Increasing the EF enhanced the MSC migratory response. The migration speed peaked at 300 mV/mm, at a rate of 42 ±1 µm/h, around double the control (no EF) migration rate. MSCs showed sustained response to prolonged EF application in vitro up to at least 8 h. The electrotaxis of MSCs with either early (P3-P5) or late (P7-P10) passage was also investigated. Migration was passage-dependent with higher passage number showing reduced directed migration, within the range of passages examined. An EF of 200 mV/mm for 2 h did not affect cell senescence, phenotype, or osteogenic potential of MSCs, regardless of passage number within the range tested (P3-P10). Our findings indicate that EFs are a powerful cue in directing migration of human MSCs in vitro. An applied EF may be useful to control or enhance migration of MSCs during bone healing.


Assuntos
Células da Medula Óssea/fisiologia , Movimento Celular , Estimulação Elétrica , Células-Tronco Mesenquimais/fisiologia , Regeneração Óssea , Sobrevivência Celular , Células Cultivadas , Senescência Celular , Humanos , Microscopia de Vídeo , Pessoa de Meia-Idade , Osteogênese , Fenótipo , Imagem com Lapso de Tempo , Adulto Jovem
20.
Arthritis Rheum ; 62(4): 1007-16, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20131274

RESUMO

OBJECTIVE: To demonstrate that posttranslational modification of type II collagen (CII) by reactive oxygen species (ROS), which are known to be present in inflamed arthritic joints, can give rise to epitopes specific to damaged cartilage in rheumatoid arthritis (RA) and osteoarthritis (OA) and to establish a proof of concept that antibodies specific to ROS-modified CII can be used to target therapeutics specifically to inflamed arthritic joints. METHODS: We used a semisynthetic phage display human antibody library to raise single-chain variable fragments (scFv) specific to ROS-modified CII. The specificity of anti-ROS-modified CII scFv to damaged arthritic cartilage was assessed in vitro by immunostaining articular cartilage from RA and OA patients and from normal controls. The in vivo targeting potential was tested using mice with antigen-induced arthritis, in which localization of anti-ROS-modified CII scFv in the joints was determined. The therapeutic effect of anti-ROS-modified CII scFv fused to soluble murine tumor necrosis factor receptor II-Fc fusion protein (mTNFRII-Fc) was also investigated. RESULTS: The anti-ROS-modified CII scFv bound to damaged arthritic cartilage from patients with RA and OA but not to normal preserved cartilage. When systemically administered to arthritic mice, the anti-ROS-modified CII accumulated selectively at the inflamed joints. Importantly, when fused to mTNFRII-Fc, it significantly reduced inflammation in arthritic mice, as compared with the effects of mTNFRII-Fc alone or of mTNFRII-Fc fused to an irrelevant scFv. CONCLUSION: Our findings indicate that biologic therapeutics can be targeted specifically to arthritic joints and suggest a new approach for the development of novel treatments of arthritis.


Assuntos
Artrite Reumatoide/imunologia , Cartilagem/imunologia , Anticorpos de Cadeia Única/genética , Animais , Artrite Reumatoide/genética , Cartilagem/patologia , Cartilagem Articular/imunologia , Cartilagem Articular/patologia , Bovinos , Modelos Animais de Doenças , Epitopos/imunologia , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Osteoartrite/genética , Osteoartrite/imunologia , Osteoprotegerina/imunologia , Espécies Reativas de Oxigênio/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA