Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
BMC Cancer ; 24(1): 370, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38528445

RESUMO

BACKGROUND: Relapsed or refractory follicular lymphoma (rrFL) is an incurable disease associated with shorter remissions and survival after each line of standard therapy. Many promising novel, chemotherapy-free therapies are in development, but few are licensed as their role in current treatment pathways is poorly defined. METHODS: The REFRACT trial is an investigator-initiated, UK National Cancer Research Institute, open-label, multi-centre, randomised phase II platform trial aimed at accelerating clinical development of novel therapies by addressing evidence gaps. The first of the three sequential novel therapy arms is epcoritamab plus lenalidomide, to be compared with investigator choice standard therapy (ICT). Patients aged 18 years or older with biopsy proven relapsed or refractory CD20 positive, grade 1-3a follicular lymphoma and assessable disease by PET-CT are eligible. The primary outcome is complete metabolic response by PET-CT at 24 weeks using the Deauville 5-point scale and Lugano 2014 criteria. Secondary outcomes include overall metabolic response, progression-free survival, overall survival, duration of response, and quality of life assessed by EQ-5D-5 L and FACT-Lym. The trial employs an innovative Bayesian design with a target sample size of 284 patients: 95 in the ICT arm and 189 in the novel therapy arms. DISCUSSION: Whilst there are many promising novel drugs in early clinical development for rrFL, understanding the relative efficacy and safety of these agents, and their place in modern treatment pathways, is limited by a lack of randomised trials and dearth of published outcomes for standard regimens to act as historic controls. Therefore, the aim of REFRACT is to provide an efficient platform to evaluate novel agents against standard therapies for rrFL. The adaptive Bayesian power prior methodology design will minimise patient numbers and accelerate trial delivery. TRIAL REGISTRATION: ClinicalTrials.gov: NCT05848765; 08-May-2023. EUDRACT: 2022-000677-75; 10-Feb-2022.


Assuntos
Linfoma Folicular , Humanos , Linfoma Folicular/tratamento farmacológico , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Braço/patologia , Teorema de Bayes , Qualidade de Vida , Resultado do Tratamento , Ensaios Clínicos Controlados Aleatórios como Assunto , Estudos Multicêntricos como Assunto , Ensaios Clínicos Fase II como Assunto
2.
BMC Cancer ; 24(1): 83, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38225549

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most common adult malignant brain tumour, with an incidence of 5 per 100,000 per year in England. Patients with tumours showing O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation represent around 40% of newly diagnosed GBM. Relapse/tumour recurrence is inevitable. There is no agreed standard treatment for patients with GBM, therefore, it is aimed at delaying further tumour progression and maintaining health-related quality of life (HRQoL). Limited clinical trial data exist using cannabinoids in combination with temozolomide (TMZ) in this setting, but early phase data demonstrate prolonged overall survival compared to TMZ alone, with few additional side effects. Jazz Pharmaceuticals (previously GW Pharma Ltd.) have developed nabiximols (trade name Sativex®), an oromucosal spray containing a blend of cannabis plant extracts, that we aim to assess for preliminary efficacy in patients with recurrent GBM. METHODS: ARISTOCRAT is a phase II, multi-centre, double-blind, placebo-controlled, randomised trial to assess cannabinoids in patients with recurrent MGMT methylated GBM who are suitable for treatment with TMZ. Patients who have relapsed ≥ 3 months after completion of initial first-line treatment will be randomised 2:1 to receive either nabiximols or placebo in combination with TMZ. The primary outcome is overall survival time defined as the time in whole days from the date of randomisation to the date of death from any cause. Secondary outcomes include overall survival at 12 months, progression-free survival time, HRQoL (using patient reported outcomes from QLQ-C30, QLQ-BN20 and EQ-5D-5L questionnaires), and adverse events. DISCUSSION: Patients with recurrent MGMT promoter methylated GBM represent a relatively good prognosis sub-group of patients with GBM. However, their median survival remains poor and, therefore, more effective treatments are needed. The phase II design of this trial was chosen, rather than phase III, due to the lack of data currently available on cannabinoid efficacy in this setting. A randomised, double-blind, placebo-controlled trial will ensure an unbiased robust evaluation of the treatment and will allow potential expansion of recruitment into a phase III trial should the emerging phase II results warrant this development. TRIAL REGISTRATION: ISRCTN: 11460478. CLINICALTRIALS: Gov: NCT05629702.


Assuntos
Neoplasias Encefálicas , Canabinoides , Glioblastoma , Adulto , Humanos , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Canabinoides/uso terapêutico , Ensaios Clínicos Fase II como Assunto , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/patologia , Estudos Multicêntricos como Assunto , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/genética , Qualidade de Vida , Ensaios Clínicos Controlados Aleatórios como Assunto , Temozolomida/uso terapêutico
3.
Platelets ; 32(3): 352-367, 2021 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-32129691

RESUMO

C-type lectin-like receptor 2 (CLEC-2) is considered as a potential drug target in settings of wound healing, inflammation, and infection. A potential barrier to this is evidence that CLEC-2 and its ligand podoplanin play a critical role in preventing lymphatic vessel blood filling in mice throughout life. In this study, this aspect of CLEC-2/podoplanin function is investigated in more detail using new and established mouse models of CLEC-2 and podoplanin deficiency, and models of acute and chronic vascular remodeling. We report that CLEC-2 expression on platelets is not required to maintain a barrier between the blood and lymphatic systems in unchallenged mice, post-development. However, under certain conditions of chronic vascular remodeling, such as during tumorigenesis, deficiency in CLEC-2 can lead to lymphatic vessel blood filling. These data provide a new understanding of the function of CLEC-2 in adult mice and confirm the essential nature of CLEC-2-driven platelet activation in vascular developmental programs. This work expands our understanding of how lymphatic blood filling is prevented by CLEC-2-dependent platelet function and provides a context for the development of safe targeting strategies for CLEC-2 and podoplanin.


Assuntos
Lectinas Tipo C/metabolismo , Sistema Linfático/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos
5.
Haematologica ; 104(8): 1648-1660, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30733265

RESUMO

Platelets promote wound healing by forming a vascular plug and by secreting growth factors and cytokines. Glycoprotein (GP)VI and C-type lectin-like receptor (CLEC)-2 signal through a (hem)-immunoreceptor tyrosine-based activation motif, which induces platelet activation. GPVI and CLEC-2 support vascular integrity during inflammation in the skin through regulation of leukocyte migration and function, and by sealing sites of vascular damage. In this study, we investigated the role of impaired vascular integrity due to GPVI and/or CLEC-2 deficiency in wound repair using a full-thickness excisional skin wound model in mice. Transgenic mice deficient in both GPVI and CLEC-2 exhibited accelerated skin wound healing, despite a marked impairment in vascular integrity. The local and temporal bleeding in the skin led to greater plasma protein entry, including fibrinogen and clotting factors, was associated with increased fibrin generation, reduction in wound neutrophils and M1 macrophages, decreased level of tumor necrosis factor (TNF)-α, and enhanced angiogenesis at day 3 after injury. Accelerated wound healing was not due to developmental defects in CLEC-2 and GPVI double-deficient mice as similar results were observed in GPVI-deficient mice treated with a podoplanin-blocking antibody. The rate of wound healing was not altered in mice deficient in either GPVI or CLEC-2. Our results show that, contrary to defects in coagulation, bleeding following a loss of vascular integrity caused by platelet CLEC-2 and GPVI deficiency facilitates wound repair by increasing fibrin(ogen) deposition, reducing inflammation, and promoting angiogenesis.


Assuntos
Lectinas Tipo C/deficiência , Glicoproteínas de Membrana/deficiência , Neovascularização Fisiológica/genética , Glicoproteínas da Membrana de Plaquetas/deficiência , Cicatrização/genética , Animais , Biomarcadores , Feminino , Imunofluorescência , Imuno-Histoquímica , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Glicoproteínas da Membrana de Plaquetas/genética , Glicoproteínas da Membrana de Plaquetas/metabolismo , Pele/metabolismo , Pele/patologia
6.
Nat Commun ; 8(1): 2239, 2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29269852

RESUMO

Platelets play a critical role in vascular inflammation through the podoplanin and collagen/fibrin receptors, C-type-lectin-like-2 (CLEC-2) and glycoprotein VI (GPVI), respectively. Both receptors regulate endothelial permeability and prevent peri-vascular bleeding in inflammation. Here we show that platelet-specific deletion of CLEC-2 but not GPVI leads to enhanced systemic inflammation and accelerated organ injury in two mouse models of sepsis-intra-peritoneal lipopolysaccharide and cecal ligation and puncture. CLEC-2 deficiency is associated with reduced numbers of podoplanin-expressing macrophages despite increased cytokine and chemokine levels in the infected peritoneum. Pharmacological inhibition of the interaction between CLEC-2 and podoplanin regulates immune cell infiltration and the inflammatory reaction during sepsis, suggesting that activation of podoplanin underlies the anti-inflammatory action of platelet CLEC-2. We suggest podoplanin-CLEC-2 as a novel anti-inflammatory axis regulating immune cell recruitment and activation in sepsis.


Assuntos
Plaquetas/imunologia , Inflamação/imunologia , Lectinas Tipo C/imunologia , Macrófagos/imunologia , Glicoproteínas de Membrana/imunologia , Insuficiência de Múltiplos Órgãos/imunologia , Sepse/imunologia , Animais , Ceco/cirurgia , Quimiocinas/imunologia , Citocinas/imunologia , Injeções Intraperitoneais , Rim/imunologia , Rim/patologia , Lectinas Tipo C/genética , Ligadura , Lipopolissacarídeos/toxicidade , Glicoproteínas de Membrana/genética , Camundongos , Fagocitose/imunologia , Glicoproteínas da Membrana de Plaquetas/genética , Glicoproteínas da Membrana de Plaquetas/imunologia , Punções , Sepse/induzido quimicamente
7.
Am J Physiol Lung Cell Mol Physiol ; 313(6): L1016-L1029, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28839100

RESUMO

There is no therapeutic intervention proven to prevent acute respiratory distress syndrome (ARDS). Novel mechanistic insights into the pathophysiology of ARDS are therefore required. Platelets are implicated in regulating many of the pathogenic processes that occur during ARDS; however, the mechanisms remain elusive. The platelet receptor CLEC-2 has been shown to regulate vascular integrity at sites of acute inflammation. Therefore the purpose of this study was to establish the role of CLEC-2 and its ligand podoplanin in a mouse model of ARDS. Platelet-specific CLEC-2-deficient, as well as alveolar epithelial type I cell (AECI)-specific or hematopoietic-specific podoplanin deficient, mice were established using cre-loxP strategies. Combining these with intratracheal (IT) instillations of lipopolysaccharide (LPS), we demonstrate that arterial oxygen saturation decline in response to IT-LPS in platelet-specific CLEC-2-deficient mice is significantly augmented. An increase in bronchoalveolar lavage (BAL) neutrophils and protein was also observed 48 h post-IT-LPS, with significant increases in pro-inflammatory chemokines detected in BAL of platelet-specific CLEC-2-deficient animals. Deletion of podoplanin from hematopoietic cells but not AECIs also reduces lung function and increases pro-inflammatory chemokine expression following IT-LPS. Furthermore, we demonstrate that following IT-LPS, platelets are present in BAL in aggregates with neutrophils, which allows for CLEC-2 interaction with podoplanin expressed on BAL inflammatory alveolar macrophages. Taken together, these data suggest that the platelet CLEC-2-podoplanin signaling axis regulates the severity of lung inflammation in mice and is a possible novel target for therapeutic intervention in patients at risk of developing ARDS.


Assuntos
Plaquetas/imunologia , Lectinas Tipo C/imunologia , Lesão Pulmonar/imunologia , Macrófagos Alveolares/imunologia , Glicoproteínas de Membrana/imunologia , Transdução de Sinais/imunologia , Animais , Plaquetas/patologia , Deleção de Genes , Lectinas Tipo C/genética , Lipopolissacarídeos/toxicidade , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/genética , Lesão Pulmonar/patologia , Macrófagos Alveolares/patologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Síndrome do Desconforto Respiratório/induzido quimicamente , Síndrome do Desconforto Respiratório/genética , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/patologia , Transdução de Sinais/genética
8.
Thorax ; 70(7): 617-24, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25903964

RESUMO

RATIONALE: Vitamin D deficiency has been implicated as a pathogenic factor in sepsis and intensive therapy unit mortality but has not been assessed as a risk factor for acute respiratory distress syndrome (ARDS). Causality of these associations has never been demonstrated. OBJECTIVES: To determine if ARDS is associated with vitamin D deficiency in a clinical setting and to determine if vitamin D deficiency in experimental models of ARDS influences its severity. METHODS: Human, murine and in vitro primary alveolar epithelial cell work were included in this study. FINDINGS: Vitamin D deficiency (plasma 25(OH)D levels <50 nmol/L) was ubiquitous in patients with ARDS and present in the vast majority of patients at risk of developing ARDS following oesophagectomy. In a murine model of intratracheal lipopolysaccharide challenge, dietary-induced vitamin D deficiency resulted in exaggerated alveolar inflammation, epithelial damage and hypoxia. In vitro, vitamin D has trophic effects on primary human alveolar epithelial cells affecting >600 genes. In a clinical setting, pharmacological repletion of vitamin D prior to oesophagectomy reduced the observed changes of in vivo measurements of alveolar capillary damage seen in deficient patients. CONCLUSIONS: Vitamin D deficiency is common in people who develop ARDS. This deficiency of vitamin D appears to contribute to the development of the condition, and approaches to correct vitamin D deficiency in patients at risk of ARDS should be developed. TRIAL REGISTRATION: UKCRN ID 11994.


Assuntos
Síndrome do Desconforto Respiratório/etiologia , Deficiência de Vitamina D/complicações , APACHE , Idoso , Animais , Calcifediol/sangue , Calcifediol/farmacologia , Calcitriol/sangue , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Esofagectomia/efeitos adversos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Unidades de Terapia Intensiva , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/efeitos dos fármacos , Síndrome do Desconforto Respiratório/sangue , Síndrome do Desconforto Respiratório/prevenção & controle , Fatores de Risco , Análise de Sobrevida , Vitamina D/uso terapêutico , Deficiência de Vitamina D/sangue , Deficiência de Vitamina D/tratamento farmacológico
9.
Blood ; 120(7): 1516-27, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22740442

RESUMO

Blood vessel networks form in a 2-step process of sprouting angiogenesis followed by selective branch regression and stabilization of remaining vessels. Pericytes are known to function in stabilizing blood vessels, but their role in vascular sprouting and selective vessel regression is poorly understood. The endosialin (CD248) receptor is expressed by pericytes associated with newly forming but not stable quiescent vessels. In the present study, we used the Endosialin(-/-) mouse as a means to uncover novel roles for pericytes during the process of vascular network formation. We demonstrate in a postnatal retina model that Endosialin(-/-) mice have normal vascular sprouting but are defective in selective vessel regression, leading to increased vessel density. Examination of the Endosialin(-/-) mouse tumor vasculature revealed an equivalent phenotype, indicating that pericytes perform a hitherto unidentified function to promote vessel destabilization and regression in vivo in both physiologic and pathologic angiogenesis. Mechanistically, Endosialin(-/-) mice have no defect in pericyte recruitment. Rather, endosialin binding to an endothelial associated, but not a pericyte associated, basement membrane component induces endothelial cell apoptosis and detachment. The results of the present study advance our understanding of pericyte biology and pericyte/endothelial cell cooperation during vascular patterning and have implications for the design of both pro- and antiangiogenic therapies.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/patologia , Padronização Corporal , Neovascularização Fisiológica , Pericitos/patologia , Animais , Animais Recém-Nascidos , Antígenos CD/metabolismo , Aorta/crescimento & desenvolvimento , Aorta/patologia , Apoptose , Membrana Basal/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/metabolismo , Pericitos/metabolismo , Ratos , Retina/metabolismo , Retina/patologia , Vasos Retinianos/crescimento & desenvolvimento , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Immunology ; 133(3): 288-95, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21466550

RESUMO

CD248 (endosialin) is a transmembrane glycoprotein that is dynamically expressed on pericytes and fibroblasts during tissue development, tumour neovascularization and inflammation. Its role in tissue remodelling is associated with increased stromal cell proliferation and migration. We show that CD248 is also uniquely expressed by human, but not mouse (C57BL/6), CD8(+) naive T cells. CD248 is found only on CD8(+) CCR7(+) CD11a(low) naive T cells and on CD8 single-positive T cells in the thymus. Transfection of the CD248 negative T-cell line MOLT-4 with CD248 cDNA surprisingly reduced cell proliferation. Knock-down of CD248 on naive CD8 T cells increased cell proliferation. These data demonstrate opposing functions for CD248 on haematopoietic (CD8(+)) versus stromal cells and suggests that CD248 helps to maintain naive CD8(+) human T cells in a quiescent state.


Assuntos
Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Estromais/imunologia , Animais , Western Blotting , Proliferação de Células , Citometria de Fluxo , Humanos , Camundongos
11.
Eur J Immunol ; 40(7): 1884-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20432232

RESUMO

CD248 is a cell surface receptor that specifically identifies fibroblasts and pericytes during development and in association with cancer and inflammation. However, its function is poorly defined and its role in lymphoid organs not studied. Here, we used (4-hydroxy-3-nitrophenyl)acetyl chicken gamma-globulin immunisation and mice lacking CD248 to study whether CD248 modulates popliteal LN (pLN) expansion and subsequent immune responses. We have found that CD248 is required for complete pLN expansion but not for co-ordination of B and T cell compartmentalisation or antibody production following (4-hydroxy-3-nitrophenyl)acetyl chicken gamma-globulin immunisation. In vitro, we show that CD248 expression in human MG63 stromal cells and mouse embryonic fibroblasts leads to a pro-proliferative and pro-migratory phenotype. This correlates with a proliferating CD248(+) population observed in vivo during pLN expansion. Taken together, these data highlight a role for CD248 in secondary lymphoid organ remodelling during adaptive immune responses.


Assuntos
Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Fibroblastos/metabolismo , Linfonodos/imunologia , Proteínas de Neoplasias/metabolismo , Pericitos/metabolismo , Células Estromais/metabolismo , Animais , Formação de Anticorpos/genética , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Neoplasias/imunologia , Linfócitos B/imunologia , Biomarcadores/metabolismo , Comunicação Celular/genética , Linhagem Celular , Movimento Celular/genética , Proliferação de Células , Galinhas , Fibroblastos/imunologia , Fibroblastos/patologia , Haptenos/imunologia , Humanos , Imunização , Linfonodos/crescimento & desenvolvimento , Linfonodos/patologia , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Nitrofenóis/imunologia , Pericitos/imunologia , Pericitos/patologia , Fenilacetatos/imunologia , Células Estromais/imunologia , Células Estromais/patologia , Linfócitos T/imunologia , gama-Globulinas/imunologia
12.
PLoS One ; 3(6): e2338, 2008 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-18523558

RESUMO

Although the expression of PECAM-1 (CD31) on vascular and haematopoietic cells within the bone marrow microenvironment has been recognized for some time, its physiological role within this niche remains unexplored. In this study we show that PECAM-1 influences steady state hematopoietic stem cell (HSC) progenitor numbers in the peripheral blood but not the bone marrow compartment. PECAM-1(-/-) mice have higher levels of HSC progenitors in the blood compared to their littermate controls. We show that PECAM-1 is required on both progenitors and bone marrow vascular cells in order for efficient transition between the blood and bone marrow to occur. We have identified key roles for PECAM-1 in both the regulation of HSC migration to the chemokine CXCL12, as well as maintaining levels of the matrix degrading enzyme MMP-9 in the bone marrow vascular niche. Using intravital microscopy and adoptive transfer of either wild type (WT) or PECAM-1(-/-) bone marrow precursors, we demonstrate that the increase in HSC progenitors in the blood is due in part to a reduced ability to migrate from blood to the bone marrow vascular niche. These findings suggest a novel role for PECAM-1 as a regulator of resting homeostatic progenitor cell numbers in the blood.


Assuntos
Células da Medula Óssea/citologia , Compartimento Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/fisiologia , Animais , Quimiocina CXCL12/fisiologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal
13.
Arthritis Res Ther ; 10(2): R47, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18433499

RESUMO

INTRODUCTION: A surprising feature of the inflammatory infiltrate in rheumatoid arthritis is the accumulation of neutrophils within synovial fluid and at the pannus cartilage boundary. Recent findings suggest that a distinct subset of IL-17-secreting T-helper cells (TH17 cells) plays a key role in connecting the adaptive and innate arms of the immune response and in regulating neutrophil homeostasis. We therefore tested the hypothesis that synovial fibroblasts bridge the biological responses that connect TH17 cells to neutrophils by producing neutrophil survival factors following their activation with IL-17. METHODS: IL-17-expressing cells in the rheumatoid synovium, and IL-17-expressing cells in the peripheral blood, and synovial fluid were examined by confocal microscopy and flow cytometry, respectively. Peripheral blood neutrophils were cocultured either with rheumatoid arthritis synovial fibroblasts (RASF) or with conditioned medium from RASF that had been pre-exposed to recombinant human IL-17, TNFalpha or a combination of the two cytokines. Neutrophils were harvested and stained with the vital mitochondrial dye 3,3'-dihexyloxacarbocyanine iodide before being enumerated by flow cytometry. RESULTS: TH17-expressing CD4+ cells were found to accumulate within rheumatoid synovial tissue and in rheumatoid arthritis synovial fluid. RASF treated with IL-17 and TNFalpha (RASFIL-17/TNF) effectively doubled the functional lifespan of neutrophils in coculture. This was entirely due to soluble factors secreted from the fibroblasts. Specific depletion of granulocyte-macrophage colony-stimulating factor from RASFIL-17/TNF-conditioned medium demonstrated that this cytokine accounted for approximately one-half of the neutrophil survival activity. Inhibition of phosphatidylinositol-3-kinase and NF-kappaB pathways showed a requirement for both signalling pathways in RASFIL-17/TNF-mediated neutrophil rescue. CONCLUSION: The increased number of neutrophils with an extended lifespan found in the rheumatoid synovial microenvironment is partly accounted for by IL-17 and TNFalpha activation of synovial fibroblasts. TH17-expressing T cells within the rheumatoid synovium are likely to contribute significantly to this effect.


Assuntos
Artrite Reumatoide/imunologia , Fibroblastos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interleucina-17/metabolismo , Neutrófilos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Idoso , Artrite Reumatoide/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Sobrevivência Celular/fisiologia , Feminino , Citometria de Fluxo , Humanos , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Membrana Sinovial/citologia , Membrana Sinovial/imunologia , Membrana Sinovial/metabolismo , Subpopulações de Linfócitos T/metabolismo
14.
FEBS Lett ; 581(18): 3550-6, 2007 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-17628549

RESUMO

Studies of stromal cell populations in lymphoid tissue (LT) have been hampered by a lack of selective markers. Here, we show that CD248 (Endosialin/TEM1) is a stromal marker that is differentially expressed on fibroblasts and pericytes in the thymus, lymph node and spleen. Expression is high during LT development but largely disappears in the adult. CD248 is re-expressed in a Salmonella-induced model of splenic enlargement; peak expression corresponding to the peak of splenic enlargement. These results suggest that CD248 expression helps define a subset of LT stromal cells which play a role in remodelling during tissue development, infection and repair.


Assuntos
Antígenos CD/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Tecido Linfoide/embriologia , Tecido Linfoide/metabolismo , Proteínas de Neoplasias/metabolismo , Baço/embriologia , Baço/metabolismo , Células Estromais/metabolismo , Animais , Linfócitos B/metabolismo , Biomarcadores , Contagem de Células , Camundongos , Camundongos Endogâmicos C57BL , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Infecções por Salmonella/metabolismo , Infecções por Salmonella/patologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA