Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Br J Cancer ; 130(10): 1659-1669, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38480935

RESUMO

BACKGROUND: Vestibular schwannomas (VSs) remain a challenge due to their anatomical location and propensity to growth. Macrophages are present in VS but their roles in VS pathogenesis remains unknown. OBJECTIVES: The objective was to assess phenotypic and functional profile of macrophages in VS with single-cell RNA sequencing (scRNAseq). METHODS: scRNAseq was carried out in three VS samples to examine characteristics of macrophages in the tumour. RT-qPCR was carried out on 10 VS samples for CD14, CD68 and CD163 and a panel of macrophage-associated molecules. RESULTS: scRNAseq revealed macrophages to be a major constituent of VS microenvironment with three distinct subclusters based on gene expression. The subclusters were also defined by expression of CD163, CD68 and IL-1ß. AREG and PLAUR were expressed in the CD68+CD163+IL-1ß+ subcluster, PLCG2 and NCKAP5 were expressed in CD68+CD163+IL-1ß- subcluster and AUTS2 and SPP1 were expressed in the CD68+CD163-IL-1ß+ subcluster. RT-qPCR showed expression of several macrophage markers in VS of which CD14, ALOX15, Interleukin-1ß, INHBA and Colony Stimulating Factor-1R were found to have a high correlation with tumour volume. CONCLUSIONS: Macrophages form an important component of VS stroma. scRNAseq reveals three distinct subsets of macrophages in the VS tissue which may have differing roles in the pathogenesis of VS.


Assuntos
Macrófagos , Neuroma Acústico , Análise de Sequência de RNA , Análise de Célula Única , Humanos , Neuroma Acústico/genética , Neuroma Acústico/patologia , Neuroma Acústico/metabolismo , Análise de Célula Única/métodos , Macrófagos/metabolismo , Macrófagos/patologia , Microambiente Tumoral/genética , Feminino , Masculino , Pessoa de Meia-Idade , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo
2.
Nat Immunol ; 25(4): 682-692, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38396288

RESUMO

Fibroblasts are important regulators of inflammation, but whether fibroblasts change phenotype during resolution of inflammation is not clear. Here we use positron emission tomography to detect fibroblast activation protein (FAP) as a means to visualize fibroblast activation in vivo during inflammation in humans. While tracer accumulation is high in active arthritis, it decreases after tumor necrosis factor and interleukin-17A inhibition. Biopsy-based single-cell RNA-sequencing analyses in experimental arthritis show that FAP signal reduction reflects a phenotypic switch from pro-inflammatory MMP3+/IL6+ fibroblasts (high FAP internalization) to pro-resolving CD200+DKK3+ fibroblasts (low FAP internalization). Spatial transcriptomics of human joints indicates that pro-resolving niches of CD200+DKK3+ fibroblasts cluster with type 2 innate lymphoid cells, whereas MMP3+/IL6+ fibroblasts colocalize with inflammatory immune cells. CD200+DKK3+ fibroblasts stabilized the type 2 innate lymphoid cell phenotype and induced resolution of arthritis via CD200-CD200R1 signaling. Taken together, these data suggest a dynamic molecular regulation of the mesenchymal compartment during resolution of inflammation.


Assuntos
Artrite , Imunidade Inata , Humanos , Metaloproteinase 3 da Matriz , Interleucina-6/metabolismo , Linfócitos/metabolismo , Inflamação/metabolismo , Fibroblastos/metabolismo
3.
Arthritis Res Ther ; 25(1): 176, 2023 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-37730663

RESUMO

BACKGROUND: Synovial fibroblasts in rheumatoid arthritis (RAFLS) exhibit a pathological aberration of glycolysis and glutaminolysis. Henceforth, we aimed to investigate if dual inhibition of these pathways by phytobiological compound c28MS has the potential of synergistic therapy for arthritis by targeting both glucose and glutamine metabolism. METHODS: The presence of HK2 and GLS across various cell types and associated gene expression in human synovial cells and a murine model of arthritis was evaluated by scRNA-seq. The metabolic profiling of RAFLS cells was done using H1-nuclear magnetic resonance spectroscopy under glycolytic and glutaminolytic inhibitory conditions by incubating with 3-bromopyruvate, CB839, or dual inhibitor c28MS. FLS functional analysis was conducted under similar conditions. ELISA was employed for the quantification of IL-6, CCL2, and MMP3. K/BxN sera was administered to mice to induce arthritis for in vivo arthritis experiments. RESULTS: scRNA-seq analysis revealed that many fibroblasts expressed Hk2 along with Gls with several genes including Ptgs2, Hif1a, Timp1, Cxcl5, and Plod2 only associated with double-positive fibroblasts, suggesting that dual inhibition can be an attractive target for fibroblasts. Metabolomic and functional analysis revealed that c28MS decreased the aggressive behavior of RAFLS by targeting both upregulated glycolysis and glutaminolysis. c28MS administered in vivo significantly decreased the severity of arthritis in the K/BxN model. CONCLUSION: Our findings imply that dual inhibition of glycolysis and glutaminolysis could be an effective approach for the treatment of RA. It also suggests that targeting more than one metabolic pathway can be a novel treatment approach in non-cancer diseases.


Assuntos
Artrite Reumatoide , Humanos , Animais , Camundongos , Artrite Reumatoide/tratamento farmacológico , Metabolômica , Glicólise , Ciclo-Oxigenase 2 , Ensaio de Imunoadsorção Enzimática
4.
Front Immunol ; 14: 1103231, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37529037

RESUMO

Background: Glucose metabolism, specifically, hexokinase 2 (HK2), has a critical role in rheumatoid arthritis (RA) fibroblast-like synoviocyte (FLS) phenotype. HK2 localizes not only in the cytosol but also in the mitochondria, where it protects mitochondria against stress. We hypothesize that mitochondria-bound HK2 is a key regulator of RA FLS phenotype. Methods: HK2 localization was evaluated by confocal microscopy after FLS stimulation. RA FLSs were infected with Green fluorescent protein (GFP), full-length (FL)-HK2, or HK2 lacking its mitochondrial binding motif (HK2ΔN) expressing adenovirus (Ad). RA FLS was also incubated with methyl jasmonate (MJ; 2.5 mM), tofacitinib (1 µM), or methotrexate (1 µM). RA FLS was tested for migration and invasion and gene expression. Gene associations with HK2 expression were identified by examining single-cell RNA sequencing (scRNA-seq) data from murine models of arthritis. Mice were injected with K/BxN serum and given MJ. Ad-FLHK2 or Ad-HK2ΔN was injected into the knee of wild-type mice. Results: Cobalt chloride (CoCl2) and platelet-derived growth factor (PDGF) stimulation induced HK2 mitochondrial translocation. Overexpression of the HK2 mutant and MJ incubation reversed the invasive and migrative phenotype induced by FL-HK2 after PDGF stimulation, and MJ also decreased the expression of C-X-C Motif Chemokine Ligand 1 (CXCL1) and Collagen Type I Alpha 1 Chain (COL1A1). Of interest, tofacitinib but not methotrexate had an effect on HK2 dissociation from the mitochondria. In murine models, MJ treatment significantly decreased arthritis severity, whereas HK2FL was able to induce synovial hypertrophy as opposed to HK2ΔN. Conclusion: Our results suggest that mitochondrial HK2 regulates the aggressive phenotype of RA FLS. New therapeutic approaches to dissociate HK2 from mitochondria offer a safer approach than global glycolysis inhibition.


Assuntos
Artrite Reumatoide , Sinoviócitos , Sinovite , Camundongos , Animais , Sinoviócitos/metabolismo , Hexoquinase/metabolismo , Artrite Reumatoide/metabolismo , Sinovite/metabolismo , Metotrexato/uso terapêutico , Fibroblastos/metabolismo
5.
Cancer Discov ; 13(2): 364-385, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36351055

RESUMO

A lack of models that recapitulate the complexity of human bone marrow has hampered mechanistic studies of normal and malignant hematopoiesis and the validation of novel therapies. Here, we describe a step-wise, directed-differentiation protocol in which organoids are generated from induced pluripotent stem cells committed to mesenchymal, endothelial, and hematopoietic lineages. These 3D structures capture key features of human bone marrow-stroma, lumen-forming sinusoids, and myeloid cells including proplatelet-forming megakaryocytes. The organoids supported the engraftment and survival of cells from patients with blood malignancies, including cancer types notoriously difficult to maintain ex vivo. Fibrosis of the organoid occurred following TGFß stimulation and engraftment with myelofibrosis but not healthy donor-derived cells, validating this platform as a powerful tool for studies of malignant cells and their interactions within a human bone marrow-like milieu. This enabling technology is likely to accelerate the discovery and prioritization of novel targets for bone marrow disorders and blood cancers. SIGNIFICANCE: We present a human bone marrow organoid that supports the growth of primary cells from patients with myeloid and lymphoid blood cancers. This model allows for mechanistic studies of blood cancers in the context of their microenvironment and provides a much-needed ex vivo tool for the prioritization of new therapeutics. See related commentary by Derecka and Crispino, p. 263. This article is highlighted in the In This Issue feature, p. 247.


Assuntos
Medula Óssea , Neoplasias Hematológicas , Humanos , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , Organoides , Microambiente Tumoral
6.
Front Immunol ; 12: 715894, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34539648

RESUMO

Rheumatoid arthritis (RA) is a chronic prototypic immune-mediated inflammatory disease which is characterized by persistent synovial inflammation, leading to progressive joint destruction. Whilst the introduction of targeted biological drugs has led to a step change in the management of RA, 30-40% of patients do not respond adequately to these treatments, regardless of the mechanism of action of the drug used (ceiling of therapeutic response). In addition, many patients who acheive clinical remission, quickly relapse following the withdrawal of treatment. These observations suggest the existence of additional pathways of disease persistence that remain to be identified and targeted therapeutically. A major barrier for the identification of therapeutic targets and successful clinical translation is the limited understanding of the cellular mechanisms that operate within the synovial microenvironment to sustain joint inflammation. Recent insights into the heterogeneity of tissue resident synovial cells, including macropahges and fibroblasts has revealed distinct subsets of these cells that differentially regulate specific aspects of inflammatory joint pathology, paving the way for targeted interventions to specifically modulate the behaviour of these cells. In this review, we will discuss the phenotypic and functional heterogeneity of tissue resident synovial cells and how this cellular diversity contributes to joint inflammation. We discuss how critical interactions between tissue resident cell types regulate the disease state by establishing critical cellular checkpoints within the synovium designed to suppress inflammation and restore joint homeostasis. We propose that failure of these cellular checkpoints leads to the emergence of imprinted pathogenic fibroblast cell states that drive the persistence of joint inflammation. Finally, we discuss therapeutic strategies that could be employed to specifically target pathogenic subsets of fibroblasts in RA.


Assuntos
Artrite/etiologia , Artrite/metabolismo , Fibroblastos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Membrana Sinovial/imunologia , Membrana Sinovial/patologia , Animais , Artrite/patologia , Artrite/terapia , Artrite Reumatoide/etiologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Artrite Reumatoide/terapia , Biomarcadores , Comunicação Celular/imunologia , Suscetibilidade a Doenças , Fibroblastos/patologia , Regulação da Expressão Gênica , Humanos , Macrófagos/patologia , Receptores Notch/metabolismo , Recidiva , Transdução de Sinais , Sinoviócitos/metabolismo , Sinoviócitos/patologia
7.
Ann Rheum Dis ; 77(11): 1636-1643, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30061164

RESUMO

OBJECTIVES: Recent studies indicate that glucose metabolism is altered in rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLS). Hexokinases (HKs) catalyse the first step in glucose metabolism, and HK2 constitutes the principal HK inducible isoform. We hypothesise that HK2 contributes to the synovial lining hypertrophy and plays a critical role in bone and cartilage damage. METHODS: HK1 and HK2 expression were determined in RA and osteoarthritis (OA) synovial tissue by immunohistochemistry. RA FLS were transfected with either HK1 or HK2 siRNA, or infected with either adenovirus (ad)-GFP, ad-HK1 or ad-HK2. FLS migration and invasion were assessed. To study the role of HK2 in vivo, 108 particles of ad-HK2 or ad-GFP were injected into the knee of wild-type mice. K/BxN serum transfer arthritis was induced in HK2F/F mice harbouring Col1a1-Cre (HK2Col1), to delete HK2 in non-haematopoietic cells. RESULTS: HK2 is particular of RA histopathology (9/9 RA; 1/8 OA) and colocalises with FLS markers. Silencing HK2 in RA FLS resulted in a less invasive and migratory phenotype. Consistently, overexpression of HK2 resulted in an increased ability to migrate and invade. It also increased extracellular lactate production. Intra-articular injection of ad-HK2 in normal knees dramatically increased synovial lining thickness, FLS activation and proliferation. HK2 was highly expressed in the synovial lining after K/BxN serum transfer arthritis. HK2Col1 mice significantly showed decreased arthritis severity, bone and cartilage damage. CONCLUSION: HK2 is specifically expressed in RA synovial lining and regulates FLS aggressive functions. HK2 might be an attractive selective metabolic target safer than global glycolysis for RA treatment.


Assuntos
Artrite Reumatoide/enzimologia , Hexoquinase/metabolismo , Animais , Artrite Experimental/enzimologia , Artrite Experimental/genética , Artrite Experimental/patologia , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Movimento Celular/fisiologia , Regulação da Expressão Gênica , Hexoquinase/genética , Humanos , Mediadores da Inflamação/metabolismo , Camundongos Transgênicos , Osteoartrite/enzimologia , Osteoartrite/genética , Osteoartrite/patologia , RNA Interferente Pequeno/genética , Membrana Sinovial/enzimologia , Sinoviócitos/enzimologia , Sinoviócitos/fisiologia , Sinovite/enzimologia , Sinovite/patologia
8.
Arthritis Res Ther ; 18(1): 270, 2016 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-27863512

RESUMO

BACKGROUND: We investigated two distinct synovial fibroblast populations that were located preferentially in the lining or sub-lining layers and defined by their expression of either podoplanin (PDPN) or CD248, and explored their ability to undergo self-assembly and transmigration in vivo. METHODS: Synovial fibroblasts (SF) were cultured in vitro and phenotypic changes following stimulation with interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, and transforming growth factor (TGF)-ß1 were examined. To examine the phenotype of SF in vivo, a severe combined immunodeficiency (SCID) human-mouse model of cartilage destruction was utilised. RESULTS: SF in the lining layer in rheumatoid arthritis (RA) expressed high levels of PDPN compared to the normal synovium, whereas CD248 expression was restricted to sub-lining layer cells. TNF-α or IL1 stimulation in vitro resulted in an increased expression of PDPN. In contrast, stimulation with TGF-ß1 induced CD248 expression. In the SCID human-mouse model, rheumatoid SF recapitulated the expression of PDPN and CD248. Fibroblasts adjacent to cartilage expressed PDPN, and attached to, invaded, and degraded cartilage. PDPN+ CD248- SF preceded the appearance of PDPN- CD248+ cells in contralateral implants. CONCLUSIONS: We have identified two distinct SF populations identified by expression of either PDPN or CD248 which are located within different anatomical compartments of the inflamed synovial membrane. These markers discriminate between SF subsets with distinct biological properties. As PDPN-expressing cells are associated with early fibroblast migration and cartilage erosion in vivo, we propose that PDPN-expressing cells may be an attractive therapeutic target in RA.


Assuntos
Artrite Reumatoide/patologia , Fibroblastos/citologia , Membrana Sinovial/citologia , Idoso , Animais , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Cartilagem Articular/metabolismo , Diferenciação Celular/fisiologia , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Xenoenxertos , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos SCID , Microscopia de Fluorescência , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Migração Transendotelial e Transepitelial/fisiologia
9.
BMC Pulm Med ; 16(1): 51, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-27080864

RESUMO

BACKGROUND: CD248 or Endosialin is a transmembrane molecule expressed in stromal cells binding to extracellular matrix (ECM) components. It has been previously implicated in kidney fibrosis, rheumatoid arthritis as well as in tumour-stromal interactions. This study investigates the role of CD248 in the pathogenesis of fibrotic diseases in Idiopathic Pulmonary Fibrosis (IPF). METHODS: CD248 quantitative immunohistochemistry (IHC) was performed on lung samples from 22 IPF patients and its expression was assayed in cultured pulmonary fibroblasts and epithelial cells. Effects of CD248 silencing was evaluated on fibroblast proliferation and myofibroblast differentiation. RESULTS: IHC revealed strong CD248 expression in mesenchymal cells of normal lung structures such as pleura and adventitia but not in epithelium. Fibrotic areas showed markedly stronger staining than unaffected lung tissue. The extent of CD248 staining showed a significant negative correlation to lung function parameters FEV1, FVC, TLC, and TLCO (r2 > 0 · 35, p < 0 · 01). CD248 protein levels were significantly greater in IPF-derived lung fibroblasts vs normal lung fibroblasts (p < 0 · 01) and CD248 silencing significantly reduced the proliferation of lung fibroblasts, but did not affected myofibroblast differentiation. CONCLUSION: We conclude that CD248 overexpression is possibly involved in the pathogenesis of IPF and it has potential as a disease severity marker. Given that CD248 ligands are collagen type I, IV and fibronectin, we hypothesise that CD248 signalling represents a novel matrix-fibroblast interaction that may be a potential therapeutic target in IPF.


Assuntos
Antígenos CD/genética , Antígenos de Neoplasias/genética , Regulação da Expressão Gênica , Fibrose Pulmonar Idiopática/genética , Pulmão/patologia , RNA/genética , Adulto , Idoso , Antígenos CD/biossíntese , Antígenos de Neoplasias/biossíntese , Biomarcadores/metabolismo , Biópsia , Proliferação de Células , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Citometria de Fluxo , Humanos , Fibrose Pulmonar Idiopática/diagnóstico , Fibrose Pulmonar Idiopática/metabolismo , Imuno-Histoquímica , Pulmão/metabolismo , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Estudos Retrospectivos , Índice de Gravidade de Doença , Transdução de Sinais
10.
Gut ; 65(7): 1175-85, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26078290

RESUMO

INTRODUCTION: CD248 (endosialin) is a stromal cell marker expressed on fibroblasts and pericytes. During liver injury, myofibroblasts are the main source of fibrotic matrix. OBJECTIVE: To determine the role of CD248 in the development of liver fibrosis in the rodent and human setting. DESIGN: CD248 expression was studied by immunostaining and quantitative PCR in both normal and diseased human and murine liver tissue and isolated hepatic stellate cells (HSCs). Hepatic fibrosis was induced in CD248(-/-) and wild-type controls with carbon tetrachloride (CCl4) treatment. RESULTS: Expression of CD248 was seen in normal liver of humans and mice but was significantly increased in liver injury using both immunostaining and gene expression assays. CD248 was co-expressed with a range of fibroblast/HSC markers including desmin, vimentin and α-smooth muscle actin (α-SMA) in murine and human liver sections. CD248 expression was restricted to isolated primary murine and human HSC. Collagen deposition and α-SMA expression, but not inflammation and neoangiogenesis, was reduced in CD248(-/-) mice compared with wild-type mice after CCl4 treatment. Isolated HSC from wild-type and CD248(-/-) mice expressed platelet-derived growth factor receptor α (PDGFR-α) and PDGFR-ß at similar levels. As expected, PDGF-BB stimulation induced proliferation of wild-type HSC, whereas CD248(-/-) HSC did not demonstrate a proliferative response to PDGF-BB. Abrogated PDGF signalling in CD248(-/-) HSC was confirmed by significantly reduced c-fos expression in CD248(-/-) HSC compared with wild-type HSC. CONCLUSIONS: Our data show that deletion of CD248 reduces susceptibility to liver fibrosis via an effect on PDGF signalling, making it an attractive clinical target for the treatment of liver injury.


Assuntos
Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Células Estreladas do Fígado/fisiologia , Cirrose Hepática/metabolismo , Fígado/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Actinas/análise , Indutores da Angiogênese/farmacologia , Animais , Antígenos CD/análise , Antígenos de Neoplasias/análise , Becaplermina , Tetracloreto de Carbono , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Doença Crônica , Colágeno/metabolismo , Desmina/análise , Fibrose , Expressão Gênica , Células Estreladas do Fígado/química , Humanos , Inflamação/genética , Fígado/química , Cirrose Hepática/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , RNA Mensageiro/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Vimentina/análise
11.
Kidney Int ; 87(4): 807-11, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25272233

RESUMO

Pregnancy in patients with anti-neutrophil cytoplasm antibody-associated vasculitis is reportedly associated with a high risk of fetal and maternal complications. Here we describe the outcome of pregnancies in patients with granulomatosis with polyangiitis and microscopic polyangiitis at five centers in the United Kingdom using a retrospective case review of all women who became pregnant following diagnosis. We report 15 pregnancies in 13 women resulting in 15 live births including one twin pregnancy and 13 singleton pregnancies. One patient had an unplanned pregnancy and a first trimester miscarriage while taking methotrexate. All other pregnancies were planned following a minimum of 6 months clinical remission. Eleven successful pregnancies were delivered vaginally at full term, whereas three were delivered by cesarean section. All infants were healthy with no neonatal complications on their initial health check within the first 24 h of delivery and no evidence of neonatal vasculitis. One relapse occurred during pregnancy and was successfully treated with an increased dose of azathioprine and corticosteroids, intravenous immunoglobulin, and plasma exchange therapy. One patient developed tracheal crusting and subglottic stenosis of infective etiology in the third trimester requiring tracheal debridement post delivery. No patient had a relapse in the first 12 months postpartum. Thus, successful pregnancy outcomes can occur following planned pregnancy in women in sustained remission on non-teratogenic therapies.


Assuntos
Granulomatose com Poliangiite/tratamento farmacológico , Poliangiite Microscópica/tratamento farmacológico , Complicações na Gravidez/tratamento farmacológico , Aborto Espontâneo/induzido quimicamente , Adolescente , Corticosteroides/uso terapêutico , Adulto , Anti-Inflamatórios/uso terapêutico , Azatioprina/uso terapêutico , Cesárea , Ciclofosfamida/uso terapêutico , Feminino , Granulomatose com Poliangiite/terapia , Humanos , Imunossupressores/uso terapêutico , Nascido Vivo , Metotrexato/efeitos adversos , Poliangiite Microscópica/terapia , Troca Plasmática , Cuidado Pré-Concepcional , Gravidez , Complicações na Gravidez/terapia , Gravidez de Alto Risco , Gravidez não Planejada , Recidiva , Estudos Retrospectivos , Nascimento a Termo , Reino Unido , Adulto Jovem
12.
PLoS One ; 9(9): e107146, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25243742

RESUMO

CD248 (Endosialin) is a type 1 membrane protein involved in developmental and pathological angiogenesis through its expression on pericytes and regulation of PDGFRß signalling. Here we explore the function of CD248 in skeletal muscle angiogenesis. Two distinct forms of capillary growth (splitting and sprouting) can be induced separately by increasing microcirculatory shear stress (chronic vasodilator treatment) or by inducing functional overload (extirpation of a synergistic muscle). We show that CD248 is present on pericytes in muscle and that CD248-/- mice have a specific defect in capillary sprouting. In contrast, splitting angiogenesis is independent of CD248 expression. Endothelial cells respond to pro-sprouting angiogenic stimulus by up-regulating gene expression for HIF1α, angiopoietin 2 and its receptor TEK, PDGF-B and its receptor PDGFRß; this response did not occur following a pro-splitting angiogenic stimulus. In wildtype mice, defective sprouting angiogenesis could be mimicked by blocking PDGFRß signalling using the tyrosine kinase inhibitor Imatinib mesylate. We conclude that CD248 is required for PDGFRß-dependant capillary sprouting but not splitting angiogenesis, and identify a new role for CD248 expressed on pericytes in the early stages of physiological angiogenesis during muscle remodelling.


Assuntos
Antígenos CD/metabolismo , Capilares/metabolismo , Músculo Esquelético/metabolismo , Proteínas de Neoplasias/metabolismo , Neovascularização Fisiológica/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Angiopoietina-2/genética , Angiopoietina-2/metabolismo , Animais , Antígenos CD/genética , Benzamidas/farmacologia , Capilares/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mesilato de Imatinib , Camundongos , Camundongos Knockout , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Proteínas de Neoplasias/genética , Neovascularização Fisiológica/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Piperazinas/farmacologia , Prazosina/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Receptores do Fator de Crescimento Derivado de Plaquetas/genética , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
13.
Clin Rheumatol ; 31(6): 1013-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22249374

RESUMO

Chronic progressive multisystem granulomatous disease is seen in 10-30% of patients with sarcoidosis and can result in end organ damage. Corticosteroids are the mainstay of treatment with the addition of cytotoxic agents in severe cases. Some patients are refractory to such treatment and, therefore, management is a challenge. There is currently limited evidence for biological agents such as infliximab, a monoclonal anti-tumor necrosis factor-α antibody in the treatment of multisystem sarcoidosis. We report outcomes of three patients with extensive multisystem sarcoidosis refractory to conventional treatment and treated at our center. Clinical assessment and radiographic imaging were used to assess the response to infliximab treatment. Infliximab therapy induced clinical remission in all three patients, and this clinical response correlated with radiographic evidence of the resolution of granulomatous disease. Serum ACE level was reduced in all cases, and daily steroid dosage was reduced. We propose that infliximab can be an effective treatment in patients with multisystem complex sarcoidosis refractory to conventional drug therapy and can result in sustained clinical remission. Our experience supports the urgent need for randomized controlled clinical trials of anti-TNF therapy in refractory systemic sarcoidosis.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Sarcoidose/tratamento farmacológico , Sarcoidose/imunologia , Corticosteroides/uso terapêutico , Adulto , Antirreumáticos/uso terapêutico , Doença Crônica , Feminino , Fluordesoxiglucose F18/farmacologia , Humanos , Inflamação , Infliximab , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons/métodos , Prednisolona/uso terapêutico , Indução de Remissão , Tomografia Computadorizada por Raios X
14.
Kidney Int ; 80(2): 199-207, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21490589

RESUMO

Stromal fibroblasts are the primary cells of the kidney that produce fibrotic matrix. CD248 is a stromal marker expressed on fibroblasts and pericytes within the human kidney. Here, we tested whether CD248 expression in the kidney colocalizes with fibrosis and if it is associated with known determinants of chronic kidney disease (CKD). CD248 expression was located and quantified in situ by immunohistochemistry in kidney biopsies from 93 patients with IgA nephropathy and compared with 22 archived biopsies encompassing normal kidney tissue as control. In normal kidney tissue, CD248 was expressed by resident pericytes, stromal fibroblasts, and was upregulated in human CKD. The expression was linked to known determinants of renal progression. This relationship was maintained in a multivariate analysis with CD248 expression linked to renal survival. CD248 was expressed by a population of α-smooth muscle actin (SMA)(+) myofibroblasts and α-SMA(-) stromal cells but not expressed on CD45(+) leukocytes. Thus, CD248 defines a subset of stromal cells, including but not limited to some myofibroblasts, linked to albuminuria and tubulointerstitial damage during tissue remodeling in CKD.


Assuntos
Antígenos CD/análise , Antígenos de Neoplasias/análise , Insuficiência Renal Crônica/patologia , Células Estromais/química , Células Estromais/patologia , Animais , Biomarcadores , Estudos de Casos e Controles , Progressão da Doença , Expressão Gênica , Humanos , Camundongos , Miofibroblastos/metabolismo , Insuficiência Renal Crônica/genética , Distribuição Tecidual
15.
Int J Cancer ; 125(10): 2400-5, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19610069

RESUMO

Second primary neoplasms (SPNs) are a recognised late effect of treatment for childhood cancer. Thyroid SPNs can develop after exposure to low-dose radiation, due to the radio-sensitivity of the thyroid gland. The British Childhood Cancer Survivor Study (BCCSS) was set up to directly monitor the late effects of treatment, including risk of SPNs, in childhood cancer survivors and includes 17,980 5-year survivors. We carried out a cohort analysis to determine the risk of thyroid SPNs in the BCCSS, and estimated risk using standardised incidence ratios (SIRs), relative risk (RR) using multivariate Poisson regression and cumulative incidence curves. There were 340,202 person years at risk subsequent to a 5-year survival, median follow-up 17.4 years per survivor. We identified 50 thyroid SPNs including 31 (62%) papillary carcinomas, 15 (30%) follicular carcinomas and 4 (8%) other types. 88% of thyroid SPNs developed after exposure to radiotherapy in or around the thyroid gland. SIR overall was 18.0 (95% confidence interval 13.4-23.8). Risk of thyroid cancer was highest after Hodgkin's disease: RR 3.3 (1.1-10.1) and Non Hodgkin's Lymphoma: RR 3.4 (1.1-10.7) relative to leukaemia (RR 1.0) (p < 0.001). Survivors treated with radiotherapy in childhood had a RR of 4.6 (1.4-15.1) relative to survivors not treated with radiotherapy (RR 1.0), p = 0003. In conclusion, the risk of thyroid cancer in childhood cancer survivors is relatively high in this cohort of childhood cancer survivors. These results will be of use in counselling survivors of childhood cancer exposed to radiation in or around the thyroid area.


Assuntos
Neoplasias Induzidas por Radiação/epidemiologia , Segunda Neoplasia Primária/epidemiologia , Glândula Tireoide/efeitos da radiação , Neoplasias da Glândula Tireoide/epidemiologia , Adenocarcinoma/epidemiologia , Adenocarcinoma/etiologia , Adenocarcinoma Folicular/epidemiologia , Adenocarcinoma Folicular/etiologia , Adolescente , Adulto , Carcinoma Papilar/epidemiologia , Carcinoma Papilar/etiologia , Criança , Estudos de Coortes , Relação Dose-Resposta à Radiação , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Induzidas por Radiação/etiologia , Segunda Neoplasia Primária/etiologia , Prognóstico , Medição de Risco , Fatores de Risco , Taxa de Sobrevida , Sobreviventes , Neoplasias da Glândula Tireoide/etiologia , Reino Unido/epidemiologia , Adulto Jovem
16.
Exp Neurol ; 216(2): 329-41, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19159625

RESUMO

The neurogenic response to injury in the postnatal brain is limited and insufficient for restoration of function. Recent evidence suggests that transplantation of mesenchymal stem cells (MSCs) into the injured brain is associated with improved functional recovery, mediated in part through amplification in the endogenous neurogenic response to injury. In the current study we investigate the interactions between bone marrow-derived MSCs and embryonic neural stem cells (NSCs) plus their differentiated progeny using an in vitro co-culture system. Two populations of MSCs were used, MSCs induced to express neural antigens (nestin+, Tuj-1+, GFAP+) and neural antigen negative MSCs. Following co-culture of induced MSCs with differentiating NSC/progenitor cells a significant increase in Tuj-1+ neurons was detected compared to co-cultures of non-induced MSCs in which an increase in astrocyte (GFAP+) differentiation was observed. The effect was mediated by soluble interactions between the two cell populations and was independent of any effect on cell death and proliferation. Induced and non-induced MSCs also promoted the survival of Tuj-1+ cell progeny in long-term cultures and both promoted axonal growth, an effect also seen in differentiating neuroblastoma cells. Therefore, MSCs provide instructive signals that are able to direct the differentiation of NSCs and promote axonal development in neuronal progeny. The data indicates that the nature of MSC derived signals is dependent not only on their microenvironment but on the developmental status of the MSCs. Pre-manipulation of MSCs prior to transplantation in vivo may be an effective means of enhancing the endogenous neurogenic response to injury.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/imunologia , Neurônios/fisiologia , Análise de Variância , Animais , Bromodesoxiuridina/metabolismo , Morte Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura/métodos , Meios de Cultivo Condicionados/farmacologia , Embrião de Mamíferos , Citometria de Fluxo , Proteína Glial Fibrilar Ácida/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas de Filamentos Intermediários/imunologia , Proteínas de Filamentos Intermediários/metabolismo , Células-Tronco Mesenquimais/química , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neuritos/fisiologia , Neurogênese/fisiologia , Neurônios/citologia , Neurônios/imunologia , Propídio/metabolismo , Ratos , Ratos Wistar , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Fatores de Tempo , Tubulina (Proteína)/metabolismo
17.
Stem Cells ; 24(8): 1841-51, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16868208

RESUMO

Trans-differentiation is a mechanism proposed to explain how tissue-specific stem cells could generate cells of other organs, thus supporting the emerging concept of enhanced adult stem cell plasticity. Although spontaneous cell fusion rather than trans-differentiation may explain some unexpected cell fate changes in vivo, such a mechanism does not explain potential trans-differentiation events in vitro, including the generation of neural cell types from cultured bone marrow-derived stem cells. Here we present evidence that shows that cultured bone marrow-derived stem cells express neural proteins and form structures resembling neurons under defined growth conditions. We demonstrate that these changes in cell structure and neural protein expression are not consistent with typical neural development. Furthermore, the ability of bone marrow-derived stem cells to adopt a neural phenotype in vitro may occur as a result of cellular stress in response to removing cells from their niche and their growth in alternative environmental conditions. These findings suggest a potential explanation for the growth behavior of cultured bone marrow-derived stem cells and highlight the need to carefully validate the plasticity of stem cell differentiation.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Neurônios/citologia , Actinas/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Artefatos , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura/farmacologia , Citoesqueleto/fisiologia , Citoesqueleto/ultraestrutura , Inibidores Enzimáticos/farmacologia , Proteínas de Filamentos Intermediários/biossíntese , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Nestina , Neurônios/metabolismo , Ratos , Ratos Wistar , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA