Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
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.
Cardiovasc Res ; 117(8): 1935-1948, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32647892

RESUMO

AIMS: Inflammation has important roles in atherosclerosis. CD4+CD28null (CD28null) T cells are a specialized T lymphocyte subset that produce inflammatory cytokines and cytotoxic molecules. CD28null T cells expand preferentially in patients with acute coronary syndrome (ACS) rather than stable angina and are barely detectable in healthy subjects. Importantly, ACS patients with CD28null T-cell expansion have increased risk for recurrent acute coronary events and poor prognosis, compared to ACS patients in whom this cell subset does not expand. The mechanisms regulating CD28null T-cell expansion in ACS remain elusive. We therefore investigated the role of cytokines in CD28null T-cell expansion in ACS. METHODS AND RESULTS: High-purity sorted CD4+ T cells from ACS patients were treated with a panel of cytokines (TNF-α, IL-1ß, IL-6, IL-7, and IL-15), and effects on the number, phenotype, and function of CD28null T cells were analysed and compared to the control counterpart CD28+ T-cell subset. IL-7- and IL-15-induced expansion of CD28null T cells from ACS patients, while inflammatory cytokines TNF-α, IL-1ß, and IL-6 did not. The mechanisms underlying CD28null T-cell expansion by IL-7/IL-15 were preferential activation and proliferation of CD28null T cells compared to control CD28+ T cells. Additionally, IL-7/IL-15 markedly augmented CD28null T-cell cytotoxic function and interferon-γ production. Further mechanistic analyses revealed differences in baseline expression of component chains of IL-7/IL-15 receptors (CD127 and CD122) and increased baseline STAT5 phosphorylation in CD28null T cells from ACS patients compared to the control CD28+ T-cell subset. Notably, we demonstrate that CD28null T-cell expansion was significantly inhibited by Tofacitinib, a selective JAK1/JAK3 inhibitor that blocks IL-7/IL-15 signalling. CONCLUSION: Our novel data show that IL-7 and IL-15 drive the expansion and function of CD28null T cells from ACS patients suggesting that IL-7/IL-15 blockade may prevent expansion of these cells and improve patient outcomes.


Assuntos
Síndrome Coronariana Aguda/imunologia , Antígenos CD28/deficiência , Linfócitos T CD4-Positivos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inflamação/imunologia , Interleucina-15/farmacologia , Interleucina-7/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Síndrome Coronariana Aguda/metabolismo , Idoso , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Citotoxicidade Imunológica/efeitos dos fármacos , Feminino , Humanos , Inflamação/metabolismo , Interferon gama/metabolismo , Janus Quinase 1/metabolismo , Janus Quinase 3/metabolismo , Masculino , Pessoa de Meia-Idade , Fenótipo , Fosforilação , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
3.
Front Immunol ; 10: 1644, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31379843

RESUMO

Background: The co-inhibitory receptor PD-1 is expressed in many tumors including head and neck squamous cell carcinoma (HNSCC) and is an important immunotherapy target. However, the role of PD-1 ligands, PD-L1, and particularly PD-L2, in the tumor-stromal cell interactions that cause a tumor-permissive environment in HNSCC is not completely understood and is the focus of our study. Methods: Expression of PD-L1 and PD-L2 was analyzed by immunohistochemistry in situ in HNSCC tumor tissue. Co-cultures were established between stromal cells (fibroblasts and macrophages) and human papilloma virus (HPV)-positive and HPV-negative HNSCC cell lines (HNSCCs) and PD-1 ligands expression was analyzed using flow cytometry. Results: PD-L1 and PD-L2 were expressed both in tumor cells and stroma in HNSCC tissue in situ. In vitro, basal expression of PD-L1 and PD-L2 was low in HNSCCs and high on fibroblasts and macrophages. Interestingly, HPV-positive but not HPV-negative HNSCCs increased the expression of both PD-1 ligands on fibroblasts upon co-culture. This effect was not observed with macrophages. Conversely, both fibroblasts and macrophages increased PD-1 ligands on HPV-positive HNSCCs, whilst this was not observed in HPV-negative HNSCCs. Crucially, we demonstrate that up-regulation of PD-L1 and PD-L2 on fibroblasts by HPV-positive HNSCCs is mediated via TLR9. Conclusions: This work demonstrates in an in vitro model that HPV-positive HNSCCs regulate PD-L1/2 expression on fibroblasts via TLR9. This may open novel avenues to modulate immune checkpoint regulator PD-1 and its ligands by targeting TLR9.


Assuntos
Antígeno B7-H1/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Infecções por Papillomavirus/metabolismo , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Receptor Toll-Like 9/metabolismo , Regulação para Cima/fisiologia , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Feminino , Neoplasias de Cabeça e Pescoço/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Papillomaviridae/patogenicidade , Infecções por Papillomavirus/virologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/virologia , Ativação Transcricional/fisiologia
4.
Cardiovasc Res ; 115(9): 1385-1392, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31228191

RESUMO

Inflammation is an important driver of atherosclerosis, and the favourable outcomes of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial revealed the large potential of anti-inflammatory drugs for the treatment of cardiovascular disease, especially in patients with a pro-inflammatory constitution. However, the complex immune reactions driving inflammation in the vascular wall in response to an atherosclerotic microenvironment are still being unravelled. Novel insights into the cellular processes driving immunity and inflammation revealed that alterations in intracellular metabolic pathways are strong drivers of survival, growth, and function of immune cells. Therefore, this position paper presents a brief overview of the recent developments in the immunometabolism field, focusing on its role in atherosclerosis. We will also highlight the potential impact of immunometabolic markers and targets in clinical cardiovascular medicine.


Assuntos
Artérias/imunologia , Aterosclerose/imunologia , Metabolismo Energético/imunologia , Sistema Imunitário/imunologia , Imunomodulação , Inflamação/imunologia , Animais , Artérias/metabolismo , Artérias/fisiopatologia , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Consenso , Humanos , Sistema Imunitário/metabolismo , Sistema Imunitário/fisiopatologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Placa Aterosclerótica , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/metabolismo
5.
Immunology ; 146(2): 185-93, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26190355

RESUMO

Inflammation contributes to the development and perpetuation of several disorders and T lymphocytes orchestrate the inflammatory immune response. Although the role of T cells in inflammation is widely recognized, specific therapies that tackle inflammatory networks in disease are yet to be developed. CD4(+) CD28(null) T cells are a unique subset of helper T lymphocytes that recently shot back into the limelight as potential catalysts of inflammation in several inflammatory disorders such as autoimmunity, atherosclerosis and chronic viral infections. In contrast to conventional helper T cells, CD4(+) CD28(null) T cells have an inbuilt ability to release inflammatory cytokines and cytotoxic molecules that can damage tissues and amplify inflammatory pathways. It comes as no surprise that patients who have high numbers of these cells have more severe disease and poor prognosis. In this review, I provide an overview on the latest advances in the biology of CD4(+) CD28(null) T cells. Understanding the complex functions and dynamics of CD4(+) CD28(null) T cells may open new avenues for therapeutic intervention to prevent progression of inflammatory diseases.


Assuntos
Antígenos CD28/imunologia , Citotoxicidade Imunológica , Inflamação/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Anti-Inflamatórios/uso terapêutico , Antígenos CD28/deficiência , Morte Celular , Proliferação de Células , Citocinas/imunologia , Citocinas/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Ativação Linfocitária , Fenótipo , Prognóstico , Índice de Gravidade de Doença , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/patologia
6.
Curr Biol ; 25(5): 577-88, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25702581

RESUMO

BACKGROUND: Cells undergoing apoptosis are known to modulate their tissue microenvironments. By acting on phagocytes, notably macrophages, apoptotic cells inhibit immunological and inflammatory responses and promote trophic signaling pathways. Paradoxically, because of their potential to cause death of tumor cells and thereby militate against malignant disease progression, both apoptosis and tumor-associated macrophages (TAMs) are often associated with poor prognosis in cancer. We hypothesized that, in progression of malignant disease, constitutive loss of a fraction of the tumor cell population through apoptosis could yield tumor-promoting effects. RESULTS: Here, we demonstrate that apoptotic tumor cells promote coordinated tumor growth, angiogenesis, and accumulation of TAMs in aggressive B cell lymphomas. Through unbiased "in situ transcriptomics" analysis-gene expression profiling of laser-captured TAMs to establish their activation signature in situ-we show that these cells are activated to signal via multiple tumor-promoting reparatory, trophic, angiogenic, tissue remodeling, and anti-inflammatory pathways. Our results also suggest that apoptotic lymphoma cells help drive this signature. Furthermore, we demonstrate that, upon induction of apoptosis, lymphoma cells not only activate expression of the tumor-promoting matrix metalloproteinases MMP2 and MMP12 in macrophages but also express and process these MMPs directly. Finally, using a model of malignant melanoma, we show that the oncogenic potential of apoptotic tumor cells extends beyond lymphoma. CONCLUSIONS: In addition to its profound tumor-suppressive role, apoptosis can potentiate cancer progression. These results have important implications for understanding the fundamental biology of cell death, its roles in malignant disease, and the broader consequences of apoptosis-inducing anti-cancer therapy.


Assuntos
Apoptose/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Linfoma de Células B/fisiopatologia , Fagócitos/fisiologia , Transdução de Sinais/fisiologia , Microambiente Tumoral/fisiologia , Análise de Variância , Proliferação de Células/fisiologia , Fluorescência , Perfilação da Expressão Gênica , Técnicas Histológicas , Humanos , Estimativa de Kaplan-Meier , Macrófagos/fisiologia , Metaloproteinases da Matriz/metabolismo , Melanoma Experimental/fisiopatologia , Neovascularização Patológica/fisiopatologia
7.
Circulation ; 131(8): 709-20, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25527700

RESUMO

BACKGROUND: The number of CD4(+)CD28(null) (CD28(null)) T cells, a unique subset of T lymphocytes with proinflammatory and cell-lytic phenotype, increases markedly in patients with acute coronary syndrome (ACS). ACS patients harboring high numbers of CD28(null) T cells have increased risk of recurrent severe acute coronary events and unfavorable prognosis. The mechanisms that govern the increase in CD28(null) T cells in ACS remain elusive. We investigated whether apoptosis pathways regulating T-cell homeostasis are perturbed in CD28(null) T cells in ACS. METHODS AND RESULTS: We found that CD28(null) T cells in ACS were resistant to apoptosis induction via Fas-ligation or ceramide. This was attributable to a dramatic reduction in proapoptotic molecules Bim, Bax, and Fas in CD28(null) T cells, whereas antiapoptotic molecules Bcl-2 and Bcl-xL were similar in CD28(null) and CD28(+) T cells. We also show that Bim is phosphorylated in CD28(null) T cells and degraded by the proteasome. Moreover, we demonstrate for the first time that proteasomal inhibition restores the apoptosis sensitivity of CD28(null) T cells in ACS. CONCLUSIONS: We show that CD28(null) T cells in ACS harbor marked defects in molecules that regulate T-cell apoptosis, which tips the balance in favor of antiapoptotic signals and endows these cells with resistance to apoptosis. We demonstrate that the inhibition of proteasomal activity allows CD28(null) T cells to regain sensitivity to apoptosis. A better understanding of the molecular switches that control the apoptosis sensitivity of CD28(null) T cells may reveal novel strategies for targeted elimination of these T cells in ACS patients.


Assuntos
Síndrome Coronariana Aguda/metabolismo , Síndrome Coronariana Aguda/patologia , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/fisiologia , Antígenos CD28/deficiência , Linfócitos T CD4-Positivos/patologia , Proteínas de Membrana/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Síndrome Coronariana Aguda/epidemiologia , Anticorpos Anti-Idiotípicos/farmacologia , Apoptose/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2 , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células/fisiologia , Células Cultivadas , Proteína de Domínio de Morte Associada a Fas/metabolismo , Homeostase/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Fenótipo , Recidiva , Fatores de Risco , Proteína X Associada a bcl-2/metabolismo
8.
J Biomed Biotechnol ; 2012: 464532, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22235167

RESUMO

Atherosclerosis is a chronic inflammatory disease that is mediated by both the innate and adaptive immune responses. T lymphocytes, that together with B cells are the cellular effectors of the adaptive immune system, are currently endowed with crucial roles in the development and progression of atherosclerosis. Costimulatory receptors are a class of molecules expressed by T lymphocytes that regulate the activation of T cells and the generation of effector T-cell responses. In this review we present the roles of costimulatory receptors of the tumour necrosis factor receptor (TNFR) superfamily in atherosclerosis and discuss the implications for future therapies that could be used to specifically modulate the immune response of pathogenic T cells in this disease.


Assuntos
Antígenos CD/imunologia , Aterosclerose/imunologia , Ativação Linfocitária/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Linfócitos T/imunologia , Aterosclerose/patologia , Linfócitos B/imunologia , Humanos , Sistema Imunitário/imunologia
9.
Circ Res ; 110(6): 857-69, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22282196

RESUMO

RATIONALE: Patients with acute coronary syndrome (ACS) predisposed to recurrent coronary events have an expansion of a distinctive T-cell subset, the CD4(+)CD28(null) T cells. These cells are highly inflammatory and cytotoxic in spite of lacking the costimulatory receptor CD28, which is crucial for optimal T cell function. The mechanisms that govern CD4(+)CD28(null) T cell function are unknown. OBJECTIVE: Our aim was to investigate the expression and role of alternative costimulatory receptors in CD4(+)CD28(null) T cells in ACS. METHODS AND RESULTS: Expression of alternative costimulatory receptors (inducible costimulator, OX40, 4-1BB, cytotoxic T lymphocyte associated antigen-4, programmed death-1) was quantified in CD4(+)CD28(null) T cells from circulation of ACS and stable angina patients. Strikingly, in ACS, levels of OX40 and 4-1BB were significantly higher in circulating CD4(+)CD28(null) T cells compared to classical CD4(+)CD28(+) T lymphocytes. This was not observed in stable angina patients. Furthermore, CD4(+)CD28(null) T cells constituted an important proportion of CD4(+) T lymphocytes in human atherosclerotic plaques and exhibited high levels of OX40 and 4-1BB. In addition, the ligands for OX40 and 4-1BB were present in plaques and also expressed on monocytes in circulation. Importantly, blockade of OX40 and 4-1BB reduced the ability of CD4(+)CD28(null) T cells to produce interferon-γ and tumor necrosis factor-α and release perforin. CONCLUSIONS: Costimulatory pathways are altered in CD4(+)CD28(null) T cells in ACS. We show that the inflammatory and cytotoxic function of CD4(+)CD28(null) T cells can be inhibited by blocking OX40 and 4-1BB costimulatory receptors. Modulation of costimulatory receptors may allow specific targeting of this cell subset and may improve the survival of ACS patients.


Assuntos
Síndrome Coronariana Aguda/imunologia , Linfócitos T CD4-Positivos/imunologia , Receptores OX40/imunologia , Transdução de Sinais/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Síndrome Coronariana Aguda/metabolismo , Idoso , Idoso de 80 Anos ou mais , Antígenos CD28/genética , Antígenos CD28/imunologia , Antígenos CD4/genética , Antígenos CD4/imunologia , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Degranulação Celular/imunologia , Doença da Artéria Coronariana/imunologia , Doença da Artéria Coronariana/metabolismo , Feminino , Granzimas/metabolismo , Humanos , Ligantes , Masculino , Pessoa de Meia-Idade , Perforina/metabolismo , Receptores OX40/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
10.
Immunobiology ; 217(7): 669-75, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22204816

RESUMO

BACKGROUND AND AIM: Head and neck cancers (HNC) are aggressive tumours. Tumour-specific T cells are frequently identified in patients with cancer, although they fail to control tumour progression. A family of proteins called co-stimulatory receptors regulate the function of T cells and may account for T cell dysfunction in cancer. Our aim was to characterise co-stimulatory receptors on T cells in HNC patients to identify novel targets for immunotherapy. METHODS: Peripheral blood mononuclear cells were isolated from HNC patients and healthy controls and the expression of co-stimulatory (OX40, 4-1BB, ICOS) and co-inhibitory (CTLA-4, PD1) receptors was analysed on CD4(+) and CD8(+) T cells using flow cytometry. RESULTS: We found that the levels of co-stimulatory receptors OX40 and 4-1BB were significantly lower on CD4(+) T cells from HNC patients. This was more pronounced in locally advanced tumours (T3/T4) compared to early carcinomas (T1/T2). PD-1 levels were higher on CD8(+) T cells in HNC patients compared to controls. Human papilloma virus (HPV)-specific CD8(+) T cells appeared to be more affected than Influenza-specific T cells. CONCLUSIONS: Our results indicate that expression of co-stimulatory receptors on T cells from HNC patients is imbalanced with a preponderance of inhibitory signals, and reduction of stimulatory signals, especially in advanced disease. Restoring this balance could improve T cell therapy outcomes in HNC.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Carcinoma/genética , Neoplasias de Cabeça e Pescoço/genética , Receptores OX40/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Carcinoma/imunologia , Carcinoma/patologia , Estudos de Casos e Controles , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Influenza Humana/genética , Influenza Humana/imunologia , Influenza Humana/patologia , Estadiamento de Neoplasias , Orthomyxoviridae/imunologia , Papillomaviridae/imunologia , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores OX40/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
11.
J Immunol ; 182(5): 2795-807, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19234174

RESUMO

Dendritic cells (DCs) have a central role in the development of adaptive immune responses, including antitumor immunity. Factors present in the tumor milieu can alter the maturation of DCs and inhibit their capacity to activate T cells. Using gene expression analysis, we found that human DCs increased the expression of TGF-beta1 transcripts following culture with human lung carcinoma cells (LCCs). These DCs produced increased amounts of TGF-beta1 protein compared with DCs not exposed to tumor cells. LCCs also decreased the expression of CD86 and HLA-DR by immature DCs. Furthermore, LCCs decreased CD86 expression and the production of TNF-alpha and IL-12 p70 by mature DCs. Moreover, LCCs also converted mature DCs into cells producing TGF-beta1. These TGF-beta1-producing DCs were poor at eliciting the activation of naive CD4(+) T cells and sustaining their proliferation and differentiation into Th1 (IFN-gamma(+)) effectors. Instead, TGF-beta1-producing DCs demonstrated an increased ability to generate CD4(+)CD25(+)Foxp3(+) regulatory T cells that suppress the proliferation of T lymphocytes. These results identify a novel mechanism by which the function of human DCs is altered by tumor cells and contributes to the evasion of the immune response.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Fatores de Transcrição Forkhead/biossíntese , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Neoplasias Pulmonares/imunologia , Linfócitos T Reguladores/imunologia , Antígeno B7-2/biossíntese , Antígeno B7-2/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Regulação para Baixo/imunologia , Antígenos HLA-DR/biossíntese , Antígenos HLA-DR/metabolismo , Humanos , Interleucina-12/antagonistas & inibidores , Interleucina-12/biossíntese , Neoplasias Pulmonares/metabolismo , Ativação Linfocitária/imunologia , Monócitos/imunologia , Monócitos/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/biossíntese
12.
Blood ; 113(15): 3485-93, 2009 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-19171874

RESUMO

Dendritic cells (DCs) are known to produce C1q, the initiator of the classical complement pathway. We demonstrate that murine DCs deficient in C1q (C1qa(-/-)) are poorer than wild-type (WT) DCs at eliciting the proliferation and Th1 differentiation of antigen-specific T cells. These defects result from decreased production of IL-12p70 by C1qa(-/-) DCs and impaired expression of costimulatory molecules CD80 and CD86 in response to CD40 ligation. The defective production of IL-12p70 and the reduced expression of CD80 and CD86 by C1qa(-/-) DCs were specifically mediated via CD40 ligation, as normal levels of IL-12p70 and CD80/86 were observed after ligation of Toll-like receptors (TLRs) on C1qa(-/-) DCs. CD40 ligation on C1qa(-/-) DCs, but not TLR ligation, results in decreased phosphorylation of p38 and ERK1/2 kinases. A strong colocalization of CD40 and C1q was observed by confocal microscopy upon CD40 ligation (but not TLR ligation) on DCs. Furthermore, human DCs from 2 C1q-deficient patients were found to have impaired IL-12p70 production in response to CD40L stimulation. Our novel data suggest that C1q augments the production of IL-12p70 by mouse and human DCs after CD40 triggering and plays important roles in sustaining the maturation of DCs and guiding the activation of T cells.


Assuntos
Antígenos CD40/metabolismo , Complemento C1q/metabolismo , Células Dendríticas/metabolismo , Interferon gama/metabolismo , Células Th1/citologia , Animais , Apresentação de Antígeno/imunologia , Apoptose/imunologia , Calreticulina/metabolismo , Comunicação Celular/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Complemento C1q/genética , Complemento C1q/farmacologia , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Feminino , Humanos , Interleucina-12/metabolismo , Transfusão de Linfócitos , Masculino , Camundongos , Camundongos Mutantes , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fagocitose/imunologia , Baço/citologia , Células Th1/metabolismo , Receptores Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Blood ; 112(13): 5026-36, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18799722

RESUMO

Cells undergoing apoptosis are efficiently located and engulfed by phagocytes. The mechanisms by which macrophages, the professional scavenging phagocytes of apoptotic cells, are attracted to sites of apoptosis are poorly defined. Here we show that CX3CL1/fractalkine, a chemokine and intercellular adhesion molecule, is released rapidly from apoptotic lymphocytes, via caspase- and Bcl-2-regulated mechanisms, to attract macrophages. Effective chemotaxis of macrophages to apoptotic lymphocytes is dependent on macrophage fractalkine receptor, CX3CR1. CX3CR1 deficiency caused diminished recruitment of macrophages to germinal centers of lymphoid follicles, sites of high-rate B-cell apoptosis. These results provide the first demonstration of chemokine/chemokine-receptor activity in the navigation of macrophages toward apoptotic cells and identify a mechanism by which macrophage infiltration of tissues containing apoptotic lymphocytes is achieved.


Assuntos
Apoptose , Quimiocina CX3CL1/metabolismo , Quimiotaxia , Linfócitos/metabolismo , Macrófagos/fisiologia , Animais , Linfoma de Burkitt , Caspases , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Linfonodos , Linfócitos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas c-bcl-2
14.
Am J Rhinol ; 21(2): 224-30, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17424885

RESUMO

BACKGROUND: Pentraxin 3 (PTX3) and complement component C1q are humoral factors of innate immunity, produced at sites of inflammation, and are essential in immune defense against several microbes such as Aspergillus, which is commonly implicated in nasal polyposis. METHODS: PTX3 and C1q were measured in nasal polyp tissue, normal nasal mucosa, and serum of patients and healthy subjects. Immunohistochemistry for the two proteins was done on normal nasal mucosa and nasal polyps. In addition, PTX3 and C1q production from mononuclear cells from patients and healthy subjects was assessed. RESULTS: Normal nasal mucosa was found to have 100-fold higher levels of PTX3 compared with serum. No measurable local increase of PTX3 was observed in polyps compared with normal mucosa. Immunohistochemistry revealed PTX3 expression in the lining of blood vessels both within normal mucosa and nasal polyps. PTX3 also was present in mononuclear cells infiltrating nasal polyps. C1q levels were higher in polyps than in normal nasal mucosa. CONCLUSION: High levels of PTX3 are present in normal nasal mucosa, suggesting a role in the maintenance of tissue homeostasis. Elevated C1q levels in nasal polyps might be indicative of an ongoing inflammatory response in the nasal mucosa in these patients.


Assuntos
Aspergillus , Proteína C-Reativa/metabolismo , Complemento C1q/metabolismo , Imunidade Inata , Mucosa Nasal/imunologia , Pólipos Nasais/imunologia , Pólipos Nasais/microbiologia , Componente Amiloide P Sérico/metabolismo , Aspergillus/patogenicidade , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Leucócitos Mononucleares/metabolismo , Mucosa Nasal/metabolismo , Pólipos Nasais/metabolismo
15.
J Leukoc Biol ; 80(1): 87-95, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16617159

RESUMO

Pentraxins (PTX) and complement belong to the humoral arm of the innate immune system and have essential functions in immune defense to microbes and in scavenging cellular debris. The prototypic long PTX, PTX3, and the first component of the classical complement pathway, C1q, are innate opsonins involved in the disposal of dying cells by phagocytes. Whether the interaction between various innate opsonins impacts on their function is not fully understood. We show here that characterized Toll-like receptor (TLR) ligands elicit the production of C1q and PTX3 by immature dendritic cells (DC). Moreover, these molecules bind to dying cells with similar kinetics, although they recognize different domains on the cell membranes. PTX3 binds in the fluid phase to C1q, decreasing C1q deposition and subsequent complement activation on apoptotic cells. C1q increases the phagocytosis of apoptotic cells by DC and the release of interleukin-12 in the presence of TLR4 ligands and apoptotic cells; PTX3 inhibits both events. Moreover, PTX3 inhibited the cross-presentation of the MELAN-A/melanoma antigen-reactive T cell 1 (MART-1) tumor antigen expressed by dying cells, even in the presence of C1q. These results suggest that interaction of C1q and PTX3 influences the clearance of apoptotic cells by DC. The coordinated induction by primary, proinflammatory signals of C1q and PTX3 and their reciprocal regulation during inflammation influences the clearance of apoptotic cells by antigen-presenting cells and possibly plays a role in immune homeostasis.


Assuntos
Apoptose/imunologia , Proteína C-Reativa/imunologia , Ativação do Complemento/imunologia , Complemento C1q/imunologia , Células Dendríticas/imunologia , Fagócitos/imunologia , Componente Amiloide P Sérico/imunologia , Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/biossíntese , Proteína C-Reativa/biossíntese , Complemento C1q/biossíntese , Humanos , Antígeno MART-1 , Proteínas de Neoplasias/biossíntese , Fagocitose/imunologia , Componente Amiloide P Sérico/biossíntese
16.
Blood ; 107(1): 151-8, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16166594

RESUMO

Pentraxins are soluble pattern recognition receptors with a dual role: protection against extracellular microbes and autoimmunity. The mechanisms by which they accomplish these tasks are not yet fully understood. Here we show that the prototypic long pentraxin PTX3 is specifically recruited at both sides of the phagocytic synapse between dendritic cells (DCs) and dying cells and remains stably bound to the apoptotic membranes (estimated half-time > 36 hours). Apoptotic cells per se influence the production of PTX3 by maturing DCs. When both microbial stimuli and dying cells are present, PTX3 behaves as a flexible adaptor of DC function, regulating the maturation program and the secretion of soluble factors. Moreover a key event associated with autoimmunity (ie, the cross-presentation of epitopes expressed by apoptotic cells to T cells) abates in the presence of PTX3, as evaluated using self, viral, and tumor-associated model antigens (vinculin, NS3, and MelanA/MART1). In contrast, PTX3 did not influence the presentation of exogenous soluble antigens, an event required for immunity against extracellular pathogens. These data suggest that PTX3 acts as a third-party agent between microbial stimuli and dying cells, contributing to limit tissue damage under inflammatory conditions and the activation of autoreactive T cells.


Assuntos
Apresentação de Antígeno , Apoptose/imunologia , Proteína C-Reativa/metabolismo , Proteína C-Reativa/fisiologia , Apresentação Cruzada , Células Dendríticas/imunologia , Componente Amiloide P Sérico/metabolismo , Componente Amiloide P Sérico/fisiologia , Células 3T3 , Animais , Anticorpos Antivirais , Antígenos de Neoplasias , Autoantígenos , Autoimunidade , Proteína C-Reativa/biossíntese , Proteína C-Reativa/genética , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/metabolismo , Células HeLa , Humanos , Camundongos , Componente Amiloide P Sérico/biossíntese , Componente Amiloide P Sérico/genética , Transfecção
17.
J Exp Med ; 200(9): 1157-65, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15504819

RESUMO

Strategies to enhance the immunogenicity of tumors are urgently needed. Although vaccination with irradiated dying lymphoma cells recruits a tumor-specific immune response, its efficiency as immunogen is poor. Annexin V (AxV) binds with high affinity to phosphatidylserine on the surface of apoptotic and necrotic cells and thereby impairs their uptake by macrophages. Here, we report that AxV preferentially targets irradiated lymphoma cells to CD8+ dendritic cells for in vivo clearance, elicits the release of proinflammatory cytokines and dramatically enhances the protection elicited against the tumor. The response was endowed with both memory, because protected animals rejected living lymphoma cells after 72 d, and specificity, because vaccinated animals failed to reject unrelated neoplasms. Finally, AxV-coupled irradiated cells induced the regression of growing tumors. These data indicate that endogenous adjuvants that bind to dying tumor cells can be exploited to target tumors for immune rejection.


Assuntos
Anexina A5/imunologia , Imunização , Linfoma/imunologia , Linfoma/terapia , Receptores de Superfície Celular/imunologia , Raios Ultravioleta , Animais , Anexina A5/metabolismo , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose/imunologia , Receptores de Superfície Celular/metabolismo , Células Tumorais Cultivadas
18.
EMBO Rep ; 5(8): 825-30, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15272298

RESUMO

Immune responses against pathogens require that microbial components promote the activation of antigen-presenting cells (APCs). Autoimmune diseases and graft rejections occur in the absence of pathogens; in these conditions, endogenous molecules, the so-called 'innate adjuvants', activate APCs. Necrotic cells contain and release innate adjuvants; necrotic cells also release high-mobility group B1 protein (HMGB1), an abundant and conserved constituent of vertebrate nuclei. Here, we show that necrotic HMGB1(-/-) cells have a reduced ability to activate APCs, and HMGB1 blockade reduces the activation induced by necrotic wild-type cell supernatants. In vivo, HMGB1 enhances the primary antibody responses to soluble antigens and transforms poorly immunogenic apoptotic lymphoma cells into efficient vaccines.


Assuntos
Adjuvantes Imunológicos , Proteína HMGB1/imunologia , Necrose , Animais , Antígenos CD/imunologia , Linhagem Celular , Células Dendríticas/imunologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Proteína HMGB1/genética , Proteína HMGB1/isolamento & purificação , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA