Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Br J Cancer ; 130(10): 1659-1669, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38480935

RESUMEN

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.


Asunto(s)
Macrófagos , Neuroma Acústico , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Humanos , Neuroma Acústico/genética , Neuroma Acústico/patología , Neuroma Acústico/metabolismo , Análisis de la Célula Individual/métodos , Macrófagos/metabolismo , Macrófagos/patología , Microambiente Tumoral/genética , Femenino , Masculino , Persona de Mediana Edad , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo
2.
Eur J Neurol ; 28(6): 1910-1921, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33768607

RESUMEN

BACKGROUND: Evidence supports that neurodevelopmental diseases, such as Tourette syndrome (TS), may involve dysfunctional neural-immune crosstalk. This could lead to altered brain maturation and differences in immune and stress responses. Dendritic cells (DCs) play a major role in immunity as professional antigen-presenting cells; changes in their frequency have been observed in several autoimmune conditions. METHODS: In 18 TS patients (15 on stable pharmacological treatment, three unmedicated) and 18 age-matched healthy volunteers (HVs), we explored circulating blood-derived DCs and their relationship with clinical variables and brain metabolites, measured via proton magnetic resonance spectroscopy (1H-MRS). DC subsets, including plasmacytoid and myeloid type 1 and 2 dendritic cells (MDC1, MDC2), were studied with flow cytometry. 1H-MRS was used to measure total choline, glutamate plus glutamine, total creatine (tCr), and total N-acetylaspartate and N-acetylaspartyl-glutamate levels in frontal white matter (FWM) and the putamen. RESULTS: We did not observe differences in absolute concentrations of DC subsets or brain inflammatory metabolites between patients and HVs. However, TS patients manifesting anxiety showed a significant increase in MDC1s compared to TS patients without anxiety (p = 0.01). We also found a strong negative correlation between MDC1 frequency and tCr in the FWM of patients with TS (p = 0.0015), but not of HVs. CONCLUSION: Elevated frequencies of the MDC1 subset in TS patients manifesting anxiety may reflect a proinflammatory status, potentially facilitating altered neuro-immune crosstalk. Furthermore, the strong inverse correlation between brain tCr levels and MDC1 subset frequency in TS patients suggests a potential association between proinflammatory status and metabolic changes in sensitive brain regions.


Asunto(s)
Síndrome de Tourette , Encéfalo/diagnóstico por imagen , Células Dendríticas , Humanos , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética
3.
Cardiovasc Res ; 117(8): 1935-1948, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32647892

RESUMEN

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.


Asunto(s)
Síndrome Coronario Agudo/inmunología , Antígenos CD28/deficiencia , Linfocitos T CD4-Positivos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inflamación/inmunología , Interleucina-15/farmacología , Interleucina-7/farmacología , Activación de Linfocitos/efectos de los fármacos , Síndrome Coronario Agudo/metabolismo , Anciano , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Femenino , Humanos , Inflamación/metabolismo , Interferón gamma/metabolismo , Janus Quinasa 1/metabolismo , Janus Quinasa 3/metabolismo , Masculino , Persona de Mediana Edad , Fenotipo , Fosforilación , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
4.
Front Immunol ; 10: 1644, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31379843

RESUMEN

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.


Asunto(s)
Antígeno B7-H1/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Infecciones por Papillomavirus/metabolismo , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Receptor Toll-Like 9/metabolismo , Regulación hacia Arriba/fisiología , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Femenino , Neoplasias de Cabeza y Cuello/virología , Humanos , Masculino , Persona de Mediana Edad , Papillomaviridae/patogenicidad , Infecciones por Papillomavirus/virología , Carcinoma de Células Escamosas de Cabeza y Cuello/virología , Activación Transcripcional/fisiología
5.
Cardiovasc Res ; 115(9): 1385-1392, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31228191

RESUMEN

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.


Asunto(s)
Arterias/inmunología , Aterosclerosis/inmunología , Metabolismo Energético/inmunología , Sistema Inmunológico/inmunología , Inmunomodulación , Inflamación/inmunología , Animales , Arterias/metabolismo , Arterias/fisiopatología , Aterosclerosis/metabolismo , Aterosclerosis/fisiopatología , Consenso , Humanos , Sistema Inmunológico/metabolismo , Sistema Inmunológico/fisiopatología , Inflamación/metabolismo , Inflamación/fisiopatología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Placa Aterosclerótica , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo
6.
Artículo en Inglés | MEDLINE | ID: mdl-27418972

RESUMEN

More than 150 years from the initial description of inflammation in atherosclerotic plaques, randomized clinical trials to test anti-inflammatory therapies in atherosclerosis have recently been initiated. Lymphocytes and macrophages are main participants in the inflammatory response in atherosclerosis. T lymphocytes operate mainly by exerting strong influences on the function of many cells in the immune system and beyond, and co-ordinating their interactions. Importantly, T lymphocytes are not a homogenous population, but include several subsets with specialized functions that can either promote or suppress inflammation. The interactions between these T-lymphocyte subsets have critical consequences on the course and outcome of inflammation. The complexity of the inflammatory response in atherosclerosis poses significant challenges on translating experimental findings into clinical therapies and makes the journey from bench to bedside an arduous one. Here, we summarize recent advances on the role of CD4(+) T cells in the inflammatory process in atherosclerosis and discuss potential therapies to modulate these lymphocytes that may provide future breakthroughs in the treatment of atherosclerosis.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Linfocitos T/efectos de los fármacos , Animales , Aterosclerosis/sangre , Linfocitos T CD4-Positivos/efectos de los fármacos , Humanos , Linfocitos T Reguladores/efectos de los fármacos
7.
Blood ; 127(16): 1976-86, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-26903549

RESUMEN

Self-tolerance and immune homeostasis are orchestrated by FOXP3(+)regulatory T cells (Tregs). Recent data have revealed that upon stimulation, Tregs may exhibit plasticity toward a proinflammatory phenotype, producing interleukin 17 (IL-17) and/or interferon γ (IFN-γ). Such deregulation of Tregs may contribute to the perpetuation of inflammatory processes, including graft-versus-host disease. Thus, it is important to identify immunomodulatory factors influencing Treg stability. Platelet-derived microparticles (PMPs) are involved in hemostasis and vascular health and have recently been shown to be intimately involved in (pathogenic) immune responses. Therefore, we investigated whether PMPs have the ability to affect Treg plasticity. PMPs were cocultured with healthy donor peripheral blood-derived Tregs that were stimulated with anti-CD3/CD28 monoclonal antibodies in the presence of IL-2, IL-15, and IL-1ß. PMPs prevented the differentiation of peripheral blood-derived Tregs into IL-17- and IFN-γ-producing cells, even in the presence of the IL-17-driving proinflammatory cytokine IL-1ß. The mechanism of action by which PMPs prevent Treg plasticity consisted of rapid and selective P-selectin-dependent binding of PMPs to a CCR6(+)HLA-DR(+)memory-like Treg subset and their ability to inhibit Treg proliferation, in part through CXCR3 engagement. The findings that ~8% of Tregs in the circulation of healthy individuals are CD41(+)P-selectin(+)and that distinct binding of patient plasma PMPs to Tregs was observed support in vivo relevance. These findings open the exciting possibility that PMPs actively regulate the immune response at sites of (vascular) inflammation, where they are known to accumulate and interact with leukocytes, consolidating the (vascular) healing process.


Asunto(s)
Plaquetas/ultraestructura , Micropartículas Derivadas de Células/patología , Micropartículas Derivadas de Células/fisiología , Interleucina-17/metabolismo , Selectina-P/fisiología , Linfocitos T Reguladores/metabolismo , Adulto , Plaquetas/patología , Diferenciación Celular/inmunología , Células Cultivadas , Regulación hacia Abajo/inmunología , Factores de Transcripción Forkhead/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Activación de Linfocitos , Linfopoyesis/fisiología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/fisiología
8.
Immunology ; 146(2): 185-93, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26190355

RESUMEN

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.


Asunto(s)
Antígenos CD28/inmunología , Citotoxicidad Inmunológica , Inflamación/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Antiinflamatorios/uso terapéutico , Antígenos CD28/deficiencia , Muerte Celular , Proliferación Celular , Citocinas/inmunología , Citocinas/metabolismo , Citotoxicidad Inmunológica/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Fenotipo , Pronóstico , Índice de Severidad de la Enfermedad , Transducción de Señal , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Colaboradores-Inductores/patología
9.
Curr Biol ; 25(5): 577-88, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25702581

RESUMEN

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.


Asunto(s)
Apoptosis/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Linfoma de Células B/fisiopatología , Fagocitos/fisiología , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología , Análisis de Varianza , Proliferación Celular/fisiología , Fluorescencia , Perfilación de la Expresión Génica , Técnicas Histológicas , Humanos , Estimación de Kaplan-Meier , Macrófagos/fisiología , Metaloproteinasas de la Matriz/metabolismo , Melanoma Experimental/fisiopatología , Neovascularización Patológica/fisiopatología
10.
Circulation ; 131(8): 709-20, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25527700

RESUMEN

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.


Asunto(s)
Síndrome Coronario Agudo/metabolismo , Síndrome Coronario Agudo/patología , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/fisiología , Antígenos CD28/deficiencia , Linfocitos T CD4-Positivos/patología , Proteínas de la Membrana/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Síndrome Coronario Agudo/epidemiología , Anticuerpos Antiidiotipos/farmacología , Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2 , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular/fisiología , Células Cultivadas , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Homeostasis/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Fenotipo , Recurrencia , Factores de Riesgo , Proteína X Asociada a bcl-2/metabolismo
11.
J Biomed Biotechnol ; 2012: 464532, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22235167

RESUMEN

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.


Asunto(s)
Antígenos CD/inmunología , Aterosclerosis/inmunología , Activación de Linfocitos/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Linfocitos T/inmunología , Aterosclerosis/patología , Linfocitos B/inmunología , Humanos , Sistema Inmunológico/inmunología
12.
Circ Res ; 110(6): 857-69, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22282196

RESUMEN

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.


Asunto(s)
Síndrome Coronario Agudo/inmunología , Linfocitos T CD4-Positivos/inmunología , Receptores OX40/inmunología , Transducción de Señal/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Síndrome Coronario Agudo/metabolismo , Anciano , Anciano de 80 o más Años , Antígenos CD28/genética , Antígenos CD28/inmunología , Antígenos CD4/genética , Antígenos CD4/inmunología , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Degranulación de la Célula/inmunología , Enfermedad de la Arteria Coronaria/inmunología , Enfermedad de la Arteria Coronaria/metabolismo , Femenino , Granzimas/metabolismo , Humanos , Ligandos , Masculino , Persona de Mediana Edad , Perforina/metabolismo , Receptores OX40/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
13.
Immunobiology ; 217(7): 669-75, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22204816

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Carcinoma/genética , Neoplasias de Cabeza y Cuello/genética , Receptores OX40/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Carcinoma/inmunología , Carcinoma/patología , Estudios de Casos y Controles , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Gripe Humana/genética , Gripe Humana/inmunología , Gripe Humana/patología , Estadificación de Neoplasias , Orthomyxoviridae/inmunología , Papillomaviridae/inmunología , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores OX40/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
14.
J Immunol ; 184(11): 6256-65, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20435933

RESUMEN

The 129-derived Sle16 is a susceptibility locus for systemic autoimmunity when present on the C57BL/6 (B6) background. Genetic analysis of a (129xB6)F2 cross identified a region from the B6 chromosome 3 (Sle18) with positive linkage to antinuclear Abs. In this study, we have generated a B6 congenic strain harboring the 129 allele of Sle18 and intercrossed this line with the lupus-prone B6.129-Sle16 strain. The presence of the 129-Sle18 allele in the B6.129-Sle16Sle18 double congenic mice suppressed the development of Sle16-mediated autoantibody production and ameliorated the renal pathology. The 129-Sle18 locus rectified the B cell abnormalities detected in the B6.129-Sle16 mice, such as the reduction in the percentage of marginal zone B and B1a cells and the increased number of germinal centers. The B6.129-Sle16Sle18 spleens still displayed an increased percentage of activated T and B cells. However, in the B6.129-Sle16Sle18 strain the percentage of naive T cells was equivalent to that in B6.129-Sle18 and B6 mice and these cells showed a reduced proliferative response to anti-CD3 stimulation compared with B6.129-Sle16 T cells. There was a significant increase in the percentage of CD4(+)FoxP3(+)regulatory T cells in all congenic strains. These cells had normal regulatory function when tested in vitro. Thus, 129-Sle18 represents a novel, non-MHC lupus-suppressor locus probably operating as a functional modifier of B cells that, in combination with other factors, leads to lupus resistance. Further characterization of this locus will help to uncover the immune mechanism(s) conferring protection against lupus.


Asunto(s)
Predisposición Genética a la Enfermedad , Lupus Eritematoso Sistémico/genética , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Separación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Congénicos , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología
15.
Eur J Immunol ; 40(6): 1758-67, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20213737

RESUMEN

Complement activation is known to have deleterious effects on organ transplantation. On the other hand, the complement system is also known to have an important role in regulating immune responses. The balance between these two opposing effects is critical in the context of transplantation. Here, we report that female mice deficient in C1q (C1qa(-/-)) or C3 (C3(-/-)) reject male syngeneic grafts (HY incompatible) at an accelerated rate compared with WT mice. Intranasal HY peptide administration, which induces tolerance to syngeneic male grafts in WT mice, fails to induce tolerance in C1qa(-/-) or C3(-/-) mice. The rejection of the male grafts correlated with the presence of HY D(b)Uty-specific CD8(+) T cells. Consistent with this, peptide-treated C1qa(-/-) and C3(-/-) female mice rejecting male grafts exhibited more antigen-specific CD8(+)IFN-gamma(+) and CD8(+)IL-10(+) cells compared with WT females. This suggests that accumulation of IFN-gamma- and IL-10-producing T cells may play a key role in mediating the ongoing inflammatory process and graft rejection. Interestingly, within the tolerized male skin grafts of peptide-treated WT mice, IFN-gamma, C1q and C3 mRNA levels were higher compared to control female grafts. These results suggest that C1q and C3 facilitate the induction of intranasal tolerance.


Asunto(s)
Complemento C1q/inmunología , Complemento C3/inmunología , Rechazo de Injerto/inmunología , Antígeno H-Y/inmunología , Trasplante Homólogo/inmunología , Administración Intranasal , Animales , Linfocitos T CD8-positivos/inmunología , Complemento C1q/deficiencia , Complemento C3/deficiencia , Citocinas/biosíntesis , Citocinas/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Antígeno H-Y/administración & dosificación , Interferón gamma/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante de Piel/inmunología
16.
J Immunol ; 182(5): 2795-807, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19234174

RESUMEN

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.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Neoplasias Pulmonares/inmunología , Linfocitos T Reguladores/inmunología , Antígeno B7-2/biosíntesis , Antígeno B7-2/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Células Dendríticas/patología , Regulación hacia Abajo/inmunología , Antígenos HLA-DR/biosíntesis , Antígenos HLA-DR/metabolismo , Humanos , Interleucina-12/antagonistas & inhibidores , Interleucina-12/biosíntesis , Neoplasias Pulmonares/metabolismo , Activación de Linfocitos/inmunología , Monocitos/inmunología , Monocitos/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Blood ; 113(15): 3485-93, 2009 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-19171874

RESUMEN

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.


Asunto(s)
Antígenos CD40/metabolismo , Complemento C1q/metabolismo , Células Dendríticas/metabolismo , Interferón gamma/metabolismo , Células TH1/citología , Animales , Presentación de Antígeno/inmunología , Apoptosis/inmunología , Calreticulina/metabolismo , Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Células Cultivadas , Complemento C1q/genética , Complemento C1q/farmacología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Femenino , Humanos , Interleucina-12/metabolismo , Transfusión de Linfocitos , Masculino , Ratones , Ratones Mutantes , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fagocitosis/inmunología , Bazo/citología , Células TH1/metabolismo , Receptores Toll-Like/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Cardiovasc Res ; 81(1): 11-9, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18818214

RESUMEN

The crucial role of T cells in atherosclerosis and coronary artery disease (CAD) has been highlighted by recent observations. Helper CD4(+) T cells can both aggravate or attenuate the atherogenic process and the development of CAD. CD4(+)CD28(null) T cells are an unusual subset of helper cells which expand and have deleterious effects in CAD. In this review, we discuss the current issues on the generation of CD4(+)CD28(null) T cells and focus on their phenotypic and functional characteristics relevant to the development of cardiovascular events. The possible effects of the present day therapies for CAD on the CD4(+)CD28(null) T cells are also explored. Targeting the CD4(+)CD28(null) T cell subset in CAD could provide novel therapeutic strategies to prevent acute life-threatening coronary events.


Asunto(s)
Antígenos CD28/metabolismo , Enfermedad de la Arteria Coronaria/patología , Linfocitos T Colaboradores-Inductores/patología , Animales , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/fisiopatología , Antígenos CD28/genética , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/fisiopatología , Vasos Coronarios/patología , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Humanos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/fisiología , Trombosis/patología , Trombosis/fisiopatología
19.
Blood ; 112(13): 5026-36, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18799722

RESUMEN

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.


Asunto(s)
Apoptosis , Quimiocina CX3CL1/metabolismo , Quimiotaxis , Linfocitos/metabolismo , Macrófagos/fisiología , Animales , Linfoma de Burkitt , Caspasas , Línea Celular Tumoral , Células Cultivadas , Humanos , Ganglios Linfáticos , Linfocitos/citología , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-bcl-2
20.
Am J Rhinol ; 21(2): 224-30, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17424885

RESUMEN

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.


Asunto(s)
Aspergillus , Proteína C-Reactiva/metabolismo , Complemento C1q/metabolismo , Inmunidad Innata , Mucosa Nasal/inmunología , Pólipos Nasales/inmunología , Pólipos Nasales/microbiología , Componente Amiloide P Sérico/metabolismo , Aspergillus/patogenicidad , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Leucocitos Mononucleares/metabolismo , Mucosa Nasal/metabolismo , Pólipos Nasales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...