Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Infect Dis ; 229(6): 1759-1769, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38557809

RESUMEN

Vγ9Vδ2 T cells play a key role in the innate immune response to viral infections through butyrophilin 3A (BTN3A). Here, we report blood Vγ9Vδ2 T cells decreased in clinically mild COVID-19 compared to healthy volunteers, and this was maintained up to 28 days and in the recovery period. Terminally differentiated Vγ9Vδ2 T cells tended to be enriched on the day of diagnosis, 28 days after, and during the recovery period. These cells showed cytotoxic and inflammatory activities following anti-BTN3A activation. BTN3A upregulation and Vγ9Vδ2 T-cell infiltration were observed in a lung biopsy from a fatal SARS-CoV-2 infection. In vitro, SARS-CoV-2 infection increased BTN3A expression in macrophages and lung cells that enhanced the anti-SARS-CoV-2 Vγ9Vδ2 T-cell cytotoxicity and interferon-γ and tumor necrosis factor-α. Increasing concentrations of anti-BTN3A lead to viral replication inhibition. Altogether, we report Vγ9Vδ2 T cells are important in the immune response against SARS-CoV-2 infection and activation by anti-BTN3A antibody may enhance their response. Clinical Trials Registration. NCT04816760.


Asunto(s)
Butirofilinas , COVID-19 , SARS-CoV-2 , Replicación Viral , Humanos , COVID-19/inmunología , COVID-19/virología , Replicación Viral/efectos de los fármacos , SARS-CoV-2/inmunología , Butirofilinas/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Adulto , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Pulmón/virología , Pulmón/inmunología , Pulmón/patología , Fenotipo , Interferón gamma/metabolismo , Interferón gamma/inmunología , Macrófagos/inmunología , Macrófagos/virología , Antígenos CD
2.
Eur J Immunol ; 45(4): 1092-102, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25545357

RESUMEN

Macrophages orchestrate the immune response via the polarization of CD4(+) T helper (Th) cells. Different subsets of macrophages with distinct phenotypes, and sometimes opposite functions, have been described. M-CSF and IL-34 induce the differentiation of monocytes into IL-10(high) IL-12(low) immunoregulatory macrophages, which are similar to tumor-associated macrophages (TAMs) in ovarian cancer. In this study, we evaluated the capacity of human macrophages induced in the presence of M-CSF (M-CSF macrophages) or IL-34 (IL-34 macrophages) and ovarian cancer TAMs to modulate the phenotype of human CD4(+) T cells. Taken together, our results show that M-CSF-, IL-34 macrophages, and TAMs switch non-Th17 committed memory CD4(+) T cells into conventional CCR4(+) CCR6(+) CD161(+) Th17 cells, expressing or not IFN-gamma. Contrary, the pro-inflammatory GM-CSF macrophages promote Th1 cells. The polarization of memory T cells into Th17 cells is mediated via membrane IL-1α (mIL-1α), which is constitutively expressed by M-CSF-, IL-34 macrophages, and TAMs. This study elucidates a new mechanism that allows macrophages to maintain locally restrained and smoldering inflammation, which is required in angiogenesis and metastasis.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Interleucina-1alfa/inmunología , Interleucinas/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/inmunología , Células Th17/citología , Diferenciación Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Femenino , Humanos , Memoria Inmunológica/inmunología , Interferón gamma/biosíntesis , Interleucina-10/metabolismo , Subunidad p35 de la Interleucina-12/metabolismo , Interleucina-17/biosíntesis , Interleucinas/farmacología , Factor Estimulante de Colonias de Macrófagos/farmacología , Monocitos/inmunología , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias Ováricas/inmunología , Receptores CCR4/metabolismo , Receptores CCR6/metabolismo , Células Th17/inmunología
3.
Hepatology ; 60(6): 1879-90, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25066464

RESUMEN

UNLABELLED: Chronic hepatitis C virus (HCV) infection is characterized by progressive hepatic fibrosis, a process dependent on monocyte recruitment and accumulation into the liver. The mediators expressed in chronically injured liver that control the differentiation of human monocytes into profibrotic macrophages (Mφ) remain poorly defined. We report that chronically HCV-infected patients with high fibrosis stages have higher serum levels of macrophage colony-stimulating factor (M-CSF) and interleukin (IL)-34 than HCV-infected patients with lower fibrosis stages and healthy subjects. Immunohistochemistry reveals an intense expression of IL-34 and M-CSF by hepatocytes around liver lesions. In addition, HCV infection and inflammatory cytokines enhance the in vitro production of IL-34 and M-CSF by hepatocytes. We next analyzed the acquisition of profibrotic properties by Mφ generated with M-CSF (M-CSF-Mφ) or IL-34 (IL-34-Mφ). M-CSF and IL-34 up-regulate the expression, by differentiating monocytes, of chemokine (C-C motif) ligand (CCL)2, CCL4, C-C chemokine receptor (CCR)1, and CCR5, which are involved in monocyte recruitment/Mφ accumulation in liver lesions. M-CSF-Mφ and IL-34-Mφ also express the hepatic stellate cell (HSC) activators, platelet-derived growth factor, transforming growth factor beta, and galectin-3. IL-34-Mφ and M-CSF-Mφ induce type I collagen synthesis by HSCs, the main collagen-producing cells in liver fibrosis. IL-13, whose expression correlates with the fibrosis stage in HCV-infected patients, decreases the expression of the collagenase, matrix metalloproteinase 1, by IL-34-Mφ and M-CSF-Mφ, thereby enhancing collagen synthesis. By inhibiting the production of interferon-gamma (IFN-γ) by activated natural killer cells, IL-34-Mφ and M-CSF-Mφ prevent the IFN-γ-induced killing of HSCs. CONCLUSION: These results identify M-CSF and IL-34 as potent profibrotic factors in HCV liver fibrosis.


Asunto(s)
Hepatitis C Crónica/complicaciones , Interleucinas/sangre , Cirrosis Hepática/inmunología , Factor Estimulante de Colonias de Macrófagos/sangre , Adulto , Anciano , Estudios de Casos y Controles , Línea Celular , Colágeno Tipo I/biosíntesis , Femenino , Células Estrelladas Hepáticas/metabolismo , Hepatitis C Crónica/metabolismo , Hepatocitos/metabolismo , Humanos , Interferón gamma/metabolismo , Interleucina-13/metabolismo , Células Asesinas Naturales/metabolismo , Cirrosis Hepática/metabolismo , Masculino , Metaloproteinasa 1 de la Matriz/metabolismo , Persona de Mediana Edad
4.
Front Immunol ; 13: 915244, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35833118

RESUMEN

Vγ9Vδ2 T cells have been reported to participate to the immune response against infectious diseases such as the Q fever caused by Coxiella burnetii infection. Indeed, the number and proportion of Vγ9Vδ2 T cells are increased during the acute phase of Q fever. Human Vγ9Vδ2 T cell responses are triggered by phosphoantigens (pAgs) produced by pathogens and malignant cells, that are sensed via the membrane receptors butyrophilin-3A1 (BTN3A1) and -2A1 (BTN2A1). Here, by using CRISPR-Cas9 inactivation in THP-1 cells, we show that BTN3A and BTN2A are required to Vγ9Vδ2 T cell response to C. burnetii infection, though not directly involved in the infection process. Furthermore, C. burnetii-infected monocytes display increased BTN3A and BTN2A expression and induce Vγ9Vδ2 T cell activation that can be inhibited by specific antagonist mAb. More importantly, we show that the antimicrobial functions of Vγ9Vδ2 T cells towards C. burnetii are enhanced in the presence of an BTN3A activating antibody. This supports the role of Vγ9Vδ2 T cells in the control of C. burnetii infection and argues in favor of targeting these cells as an alternative treatment strategy for infectious diseases caused by intracellular bacteria.


Asunto(s)
Antígenos CD , Butirofilinas , Enfermedades Transmisibles , Coxiella burnetii , Fiebre Q , Antígenos CD/inmunología , Butirofilinas/inmunología , Humanos , Fiebre Q/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología
5.
Front Immunol ; 12: 663192, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054830

RESUMEN

Objective: Interleukin-26 (IL-26) has a unique ability to activate innate immune cells due to its binding to circulating double-stranded DNA. High levels of IL-26 have been reported in patients with chronic inflammation. We aimed to investigate IL-26 levels in patients with systemic lupus erythematosus (SLE). Methods: IL-26 serum levels were quantified by ELISA for 47 healthy controls and 109 SLE patients previously enrolled in the PLUS study. Performance of IL-26 levels and classical markers (autoantibodies or complement consumption) to identify an active SLE disease (SLE disease activity index (SLEDAI) score > 4) were compared. Results: IL-26 levels were significantly higher in SLE patients than in controls (4.04 ± 11.66 and 0.74 ± 2.02 ng/mL; p = 0.005). IL-26 levels were also significantly higher in patients with active disease than those with inactive disease (33.08 ± 21.06 vs 1.10 ± 3.80 ng/mL, p < 0.0001). IL-26 levels correlated with SLEDAI score and the urine protein to creatinine ratio (uPCR) (p < 0.001). Patients with high IL-26 levels had higher SLEDAI score, anti-DNA antibodies levels, and uPCR (p < 0.05). They presented more frequently with C3 or C4 complement consumption. Lastly, IL-26 showed stronger performance than classical markers (complement consumption or autoantibodies) for active disease identification. Conclusions: Our results suggest that, in addition to classical SLE serological markers, the measurement of IL-26 levels may be a useful biomarker for active disease identification in SLE patients.


Asunto(s)
Biomarcadores/sangre , Interleucinas/sangre , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/diagnóstico , Adulto , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Estudios de Casos y Controles , Complemento C3/inmunología , Complemento C4/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/etiología , Masculino , Persona de Mediana Edad , Pronóstico , Proteinuria/diagnóstico , Proteinuria/etiología , Proteinuria/orina , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad , Adulto Joven
6.
Blood Adv ; 5(7): 1816-1829, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33787861

RESUMEN

In diffuse large B-cell lymphoma (DLBCL), tumor-infiltrating T lymphocytes (TILs) are involved in therapeutic responses. However, tumor-specific TILs can be dysfunctional, with impaired effector functions. Various mechanisms are involved in this exhaustion, and the increased expression of programmed cell death receptor 1 (PD1) and TIM3 on dysfunctional cells suggests their involvement. However, conflicting data have been published regarding their expression or coexpression in DLBCL. We evaluated the presence and phenotype of CD4+ and CD8+ TILs in freshly collected tumor tissues in DLBCL and compared the results with those in follicular lymphoma, classical Hodgkin lymphoma, and nonmalignant reactive lymphadenopathy. We found that TILs expressing both PD1 and TIM3 were expanded in DLBCL, particularly in the activated B cell-like subgroup. Isolated PD1+TIM3+ TILs exhibited a transcriptomic signature related to T-cell exhaustion associated with a reduction in cytokine production, both compromising the antitumor immune response. However, these cells expressed high levels of cytotoxic molecules. In line with this, stimulated PD1+TIM3+ TILs from DLBCL patients exhibited reduced proliferation and impaired secretion of interferon-γ, but these functions were restored by the blockade of PD1 or TIM3. In summary, the PD1+TIM3+ TIL population is expanded and exhausted in DLBCL but can be reinvigorated with appropriate therapies.


Asunto(s)
Receptor 2 Celular del Virus de la Hepatitis A , Linfoma de Células B Grandes Difuso , Linfocitos T CD8-positivos , Receptor 2 Celular del Virus de la Hepatitis A/genética , Humanos , Linfocitos Infiltrantes de Tumor , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Receptor de Muerte Celular Programada 1/genética
7.
Front Immunol ; 9: 1044, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29868007

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal cancers with very few available treatments. For many decades, gemcitabine was the only treatment for patients with PDAC. A recent attempt to improve patient survival by combining this chemotherapy with FOLFIRINOX and nab-paclitaxel failed and instead resulted in increased toxicity. Novel therapies are urgently required to improve PDAC patient survival. New treatments in other cancers such as melanoma, non-small-cell lung cancer, and renal cancer have emerged, based on immunotherapy targeting the immune checkpoints cytotoxic T-lymphocyte-associated antigen 4 or programmed death 1 ligand. However, the first clinical trials using such immune checkpoint inhibitors in PDAC have had limited success. Resistance to immunotherapy in PDAC remains unclear but could be due to tissue components (cancer-associated fibroblasts, desmoplasia, hypoxia) and to the imbalance between immunosuppressive and effector immune populations in the tumor microenvironment. In this review, we analyzed the presence of "good and bad immunological cops" in PDAC and discussed the significance of changes in their balance.


Asunto(s)
Carcinoma Ductal Pancreático/inmunología , Puntos de Control del Ciclo Celular/inmunología , Inmunoterapia , Neoplasias Pancreáticas/inmunología , Microambiente Tumoral/inmunología , Albúminas/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Ductal Pancreático/terapia , Ensayos Clínicos como Asunto , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Fluorouracilo/uso terapéutico , Humanos , Hipoxia/tratamiento farmacológico , Irinotecán/uso terapéutico , Leucovorina/uso terapéutico , Oxaliplatino/uso terapéutico , Paclitaxel/uso terapéutico , Neoplasias Pancreáticas/terapia , Gemcitabina
8.
Blood Adv ; 2(15): 1889-1900, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30087107

RESUMEN

A better characterization of T-cell subsets in the microenvironment of classical Hodgkin lymphoma (cHL) would help to develop immunotherapies. Using multicolor flow cytometry, we identified in 6 of 43 cHL tissue samples a previously unrecognized subset of CD8 T cells coexpressing CXCR5 and inducible T-cell costimulator (ICOS) molecules (CD8CXCR5+ICOS+). These cells shared phenotypic features with follicular helper T (TFH) cells including low CCR7 expression together with high expression of B-cell lymphoma-6, programmed cell death 1, B and T lymphocyte attenuator, CD200, and OX40. They had deficient cytotoxicity, low interferon-γ secretion, and common functional properties with intratumoral CD4+ TFH cells, such as production of interleukin-4 (IL-4), IL-21, CXCL13, and capacity to sustain B cells. Gene profiling analysis showed a significant similarity between the signatures of CD8CXCR5+ICOS+ T cells and CD4+ TFH cells. Benign lymphadenitis tissues (n = 8) were devoid of CD8CXCR5+ICOS+ cells. Among the 35 B-cell lymphoma tissues analyzed, including follicular lymphomas (n = 13), diffuse large cell lymphomas (n = 12), marginal zone lymphomas (MZLs; n = 3), mantle cell lymphomas (n = 3), and chronic lymphocytic leukemias (n = 4), only 1 MZL sample contained CD8CXCR5+ICOS+ cells. Lymphoma tumors with CD8CXCR5+ICOS+ cells shared common histopathological features including residual germinal centers, and contained high amounts of activated CD8CXCR5-ICOS+ cells. These data demonstrate a CD8 T-cell differentiation pathway leading to the acquisition of some TFH similarities. They suggest a particular immunoediting process with global CD8 activation acting mainly, but not exclusively, in HL tumors.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Regulación Neoplásica de la Expresión Génica , Enfermedad de Hodgkin/metabolismo , Proteína Coestimuladora de Linfocitos T Inducibles/biosíntesis , Proteínas de Neoplasias/biosíntesis , Receptores CXCR5/biosíntesis , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Diferenciación Celular , Citocinas/metabolismo , Femenino , Enfermedad de Hodgkin/patología , Humanos , Masculino
9.
Oncoimmunology ; 7(1): e1372080, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29296524

RESUMEN

Vγ9Vδ2 T cells are anti-tumor immune effectors of growing interest in cancer including Pancreatic Ductal Adenocarcinoma (PDAC), an especially aggressive cancer characterized by a hypoxic and nutrient-starved immunosuppressive microenvironment. Since Butyrophilin 3 A (BTN3A) isoforms are critical activating molecules of Vγ9Vδ2 T cells, we set out to study BTN3A expression under both basal and stress conditions in PDAC primary tumors, and in novel patient-derived xenograft and PDAC-derived cell lines. BTN3A2 was shown to be the most abundant isoform in PDAC and was stress-regulated. Vγ9Vδ2 T cells cytolytic functions against PDAC required BTN3A and this activity was strongly enhanced by the agonist anti-BTN3A 20.1 mAb even under conditions of hypoxia. In PDAC primary tumors, we established that BTN3A expression and high plasma levels of soluble BTN3A were strongly associated with a decreased survival. These findings may have important implications in the design of new immunotherapeutic strategies that target BTN3A for treating PDAC.

10.
Cardiovasc Res ; 112(1): 515-25, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27328880

RESUMEN

AIMS: The angiotensin II type 1 receptor (AT1R) through the activation of immune cells plays a key role in arterial inward remodelling and reduced blood flow in cardiovascular disorders. On the other side, flow (shear stress)-mediated outward remodelling (FMR), involved in collateral arteries growth in ischaemic diseases, allows revascularization. We hypothesized that the type 2 receptor (AT2R), described as opposing the effects of AT1R, could be involved in FMR. METHODS AND RESULTS: We studied FMR using a model of ligation of feed arteries supplying collateral pathways in the mouse mesenteric arterial bed in vivo. Seven days after ligation, diameter increased by 30% in high flow (HF) arteries compared with normal flow vessels. FMR was absent in mice lacking AT2R. At Day 2, T lymphocytes expressing AT2R were present preferentially around HF arteries. FMR did not occur in athymic (nude) mice lacking T cells and in mice treated with anti-CD3ε antibodies. AT2R activation induced interleukin-17 production by memory T cells. Treatment of nude mice or AT2R-deficient mice with interleukin-17 restored diameter enlargement in HF arteries. Interleukin-17 increased NO-dependent relaxation and matrix metalloproteinases activity, both important in FMR. Remodelling of feeding arteries in the skin flap model of ischaemia was also absent in AT2R-deficient mice and in anti-interleukin-17-treated mice. Finally, remodelling, absent in 12-month-old mice, was restored by a treatment with the AT2R non-peptidic agonist C21. CONCLUSION: AT2R-dependent interleukin-17 production by T lymphocyte is necessary for collateral artery growth and could represent a new therapeutic target in ischaemic disorders.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Interleucina-17/metabolismo , Isquemia/metabolismo , Mecanotransducción Celular , Arterias Mesentéricas/metabolismo , Mesenterio/irrigación sanguínea , Receptor de Angiotensina Tipo 2/metabolismo , Piel/irrigación sanguínea , Circulación Esplácnica , Remodelación Vascular , Factores de Edad , Animales , Presión Arterial , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Circulación Colateral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Genotipo , Isquemia/genética , Isquemia/inmunología , Isquemia/fisiopatología , Ligadura , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Mecanotransducción Celular/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/inmunología , Arterias Mesentéricas/cirugía , Ratones , Ratones Noqueados , Ratones Desnudos , Fenotipo , Células RAW 264.7 , Receptor de Angiotensina Tipo 2/agonistas , Receptor de Angiotensina Tipo 2/deficiencia , Receptor de Angiotensina Tipo 2/genética , Flujo Sanguíneo Regional , Circulación Esplácnica/efectos de los fármacos , Estrés Mecánico , Factores de Tiempo , Resistencia Vascular , Vasodilatación
11.
PLoS One ; 8(2): e56045, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23409120

RESUMEN

IL-34 is a recently identified cytokine that signals via the M-CSF receptor and promotes monocyte survival. Depending on the environment, monocytes can differentiate into macrophages (Mφ) or dendritic cells (DC). A wide spectrum of Mφ and DC subsets, with distinct phenotypes and functions, has been described. To date, the phenotype of monocytes exposed to IL-34 remains unexplored. We report here that IL-34 induces the differentiation of monocytes into CD14(high) CD163(high) CD1a(-) Mφ (IL-34-Mφ). Upon LPS stimulation, IL-34-Mφ exhibit an IL-10(high) IL-12(low) M2 profile and express low levels of the costimulatory molecules CD80 and CD86. IL-34-Mφ exhibit poor T cell costimulatory properties, and have potent immunosuppressive properties (decrease of TCR-stimulated T cell proliferation). For all the parameters analyzed, IL-34-Mφ are phenotypically and functionally similar to M-CSF-Mφ. IL-34 appears as efficient as M-CSF in inducing the generation of immunosuppressive Mφ. Moreover, the generation of IL-34-Mφ is mediated through the M-CSF receptor, is independent of endogenous M-CSF consumption and is potentiated by IL-6. In an attempt to identify strategies to prevent a deleterious M2 cell accumulation in some pathological situations, we observed that IFNγ and GM-CSF prevent the generation of immunosuppressive Mφ induced by IL-34. IFNγ also switches established IL-34-Mφ into immunostimulatory Mφ. In conclusion, we demonstrate that IL-34 drives the differentiation of monocytes into immunosuppressive M2, in a manner similar to M-CSF, and that IFNγ and GM-CSF prevent this effect.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Interferón gamma/farmacología , Interleucinas/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Monocitos/citología , Monocitos/efectos de los fármacos , Diferenciación Celular/inmunología , Quimiocinas/genética , Análisis por Conglomerados , Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Macrófagos/inmunología , Monocitos/inmunología , Fenotipo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptores de Quimiocina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA