Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Immunol ; 190(12): 6520-32, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23686490

RESUMEN

Liver X receptors (LXRs) exert key functions in lipid homeostasis and in control of inflammation. In this study we have explored the impact of LXR activation on the macrophage response to the endogenous inflammatory cytokine IFN-γ. Transcriptional profiling studies demonstrate that ∼38% of the IFN-γ-induced transcriptional response is repressed by LXR activation in macrophages. LXRs also mediated inhibitory effects on selected IFN-γ-induced genes in primary microglia and in a model of IFN-γ-induced neuroinflammation in vivo. LXR activation resulted in reduced STAT1 recruitment to the promoters tested in this study without affecting STAT1 phosphorylation. A closer look into the mechanism revealed that SUMOylation of LXRs, but not the presence of nuclear receptor corepressor 1, was required for repression of the NO synthase 2 promoter. We have also analyzed whether IFN-γ signaling exerts reciprocal effects on LXR targets. Treatment with IFN-γ inhibited, in a STAT1-dependent manner, the LXR-dependent upregulation of selective targets, including ATP-binding cassette A1 (ABCA1) and sterol response element binding protein 1c. Downregulation of ABCA1 expression correlated with decreased cholesterol efflux to apolipoprotein A1 in macrophages stimulated with IFN-γ. The inhibitory effects of IFN-γ on LXR signaling did not involve reduced binding of LXR/retinoid X receptor heterodimers to target gene promoters. However, overexpression of the coactivator CREB-binding protein/p300 reduced the inhibitory actions of IFN-γ on the Abca1 promoter, suggesting that competition for CREB-binding protein may contribute to STAT1-dependent downregulation of LXR targets. The results from this study suggest an important level of bidirectional negative cross-talk between IFN-γ/STAT1 and LXRs with implications both in the control of IFN-γ-mediated immune responses and in the regulation of lipid metabolism.


Asunto(s)
Interferón gamma/inmunología , Macrófagos/inmunología , Receptores Nucleares Huérfanos/inmunología , Receptor Cross-Talk/inmunología , Factor de Transcripción STAT1/inmunología , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica/inmunología , Inflamación/inmunología , Metabolismo de los Lípidos/fisiología , Receptores X del Hígado , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Nucleares Huérfanos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/fisiología , Transcriptoma
2.
J Immunol ; 186(8): 4656-67, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21398609

RESUMEN

Macrophages serve essential functions as regulators of immunity and homeostasis, and their proliferation contributes to pathogenesis of certain disorders. In this report, we show that induction of macrophage proliferation by the growth factor M-CSF is negatively modulated by agonists that activate the nuclear receptor liver X receptor (LXR), both in vitro and in vivo. Both isoforms LXR α and ß are involved in the antiproliferative actions of LXR ligands in macrophages. In contrast, M-CSF does not exert negative effects on LXR-mediated gene expression. Treatment with LXR agonists results in the accumulation of macrophages in the G(0)/G(1) phase of the cell cycle without affecting ERK-1/2 phosphorylation. The use of small interfering RNA or genetically modified mice revealed that, in contrast to other cellular models, functional expression of either the cyclin-dependent kinase inhibitor p27KIP1 or the cholesterol transporters ATP-binding cassette A1 or ATP-binding cassette G1 was not required for the antiproliferative effects of LXR agonists in macrophages. Western blot analysis revealed that protein expression of key molecules that regulate progression through the cell cycle, such as cyclins D1 and B1 and cyclin-dependent kinases 2 and 4, was downregulated upon LXR activation. These observations suggest a role for LXR agonists in limiting macrophage proliferative responses associated to pathogenic disorders.


Asunto(s)
Proliferación Celular , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Macrófagos/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Animales , Benzoatos/farmacología , Bencilaminas/farmacología , Western Blotting , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Ciclina B1/metabolismo , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Regulación hacia Abajo , Citometría de Flujo , Células HEK293 , Humanos , Hidrocarburos Fluorados/farmacología , Células L , Receptores X del Hígado , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nucleares Huérfanos/agonistas , Receptores Nucleares Huérfanos/genética , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sulfonamidas/farmacología
3.
Cell Stem Cell ; 28(10): 1790-1804.e8, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34010627

RESUMEN

The role of heterochromatin in cell fate specification during development is unclear. We demonstrate that loss of the lysine 9 of histone H3 (H3K9) methyltransferase G9a in the mammary epithelium results in de novo chromatin opening, aberrant formation of the mammary ductal tree, impaired stem cell potential, disrupted intraductal polarity, and loss of tissue function. G9a loss derepresses long terminal repeat (LTR) retroviral sequences (predominantly the ERVK family). Transcriptionally activated endogenous retroviruses generate double-stranded DNA (dsDNA) that triggers an antiviral innate immune response, and knockdown of the cytosolic dsDNA sensor Aim2 in G9a knockout (G9acKO) mammary epithelium rescues mammary ductal invasion. Mammary stem cell transplantation into immunocompromised or G9acKO-conditioned hosts shows partial dependence of the G9acKO mammary morphological defects on the inflammatory milieu of the host mammary fat pad. Thus, altering the chromatin accessibility of retroviral elements disrupts mammary gland development and stem cell activity through both cell-autonomous and non-autonomous mechanisms.


Asunto(s)
Retrovirus Endógenos , N-Metiltransferasa de Histona-Lisina , Glándulas Mamarias Animales/crecimiento & desarrollo , Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo/inmunología , Animales , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Femenino , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Inmunidad , Glándulas Mamarias Animales/inmunología
4.
Sci Rep ; 9(1): 2996, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30816121

RESUMEN

The presence of leukocyte subpopulations in malignant pleural effusions (MPEs) can have a different impact on tumor cell proliferation and vascular leakiness, their analysis can help to understand the metastatic microenvironment. We analyzed the relationship between the leukocyte subpopulation counts per ml of pleural fluid and the tumor cell count, molecular phenotype of lung adenocarcinoma (LAC), time from cancer diagnosis and previous oncologic therapy. We also evaluated the leukocyte composition of MPEs as a biomarker of prognosis. We determined CD4+ T, CD8+ T and CD20+ B cells, monocytes and neutrophils per ml in pleural effusions of 22 LAC and 10 heart failure (HF) patients by flow cytometry. Tumor cells were identified by morphology and CD326 expression. IFNγ, IL-10 and IL-17, and chemokines were determined by ELISAs and migratory response to pleural fluids by transwell assays. MPEs from LAC patients had more CD8+ T lymphocytes and a tendency to more CD4+ T and CD20+ B lymphocytes than HF-related fluids. However, no correlation was found between lymphocytes and tumor cells. In those MPEs which were detected >1 month from LAC diagnosis, there was a negative correlation between pleural tumor cells and CD8+ T lymphocytes. CXCL10 was responsible for the attraction of CD20+ B, CD4+ T and CD8+ T lymphocytes in malignant fluids. Concentrations of IL-17 were higher in MPEs than in HF-related effusions. Survival after MPE diagnosis correlated positively with CD4+ T and CD8+ T lymphocytes, but negatively with neutrophils and IL-17 levels. In conclusion, lymphocyte enrichment in MPEs from LAC patients is mostly due to local migration and increases patient survival.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Movimiento Celular , Neoplasias Pulmonares/patología , Derrame Pleural Maligno/patología , Linfocitos T/metabolismo , Anciano , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Molécula de Adhesión Celular Epitelial/genética , Molécula de Adhesión Celular Epitelial/metabolismo , Femenino , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico , Linfocitos T/fisiología
5.
Nat Commun ; 10(1): 2416, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186412

RESUMEN

Cancer response to immunotherapy depends on the infiltration of CD8+ T cells and the presence of tumor-associated macrophages within tumors. Still, little is known about the determinants of these factors. We show that LIF assumes a crucial role in the regulation of CD8+ T cell tumor infiltration, while promoting the presence of protumoral tumor-associated macrophages. We observe that the blockade of LIF in tumors expressing high levels of LIF decreases CD206, CD163 and CCL2 and induces CXCL9 expression in tumor-associated macrophages. The blockade of LIF releases the epigenetic silencing of CXCL9 triggering CD8+ T cell tumor infiltration. The combination of LIF neutralizing antibodies with the inhibition of the PD1 immune checkpoint promotes tumor regression, immunological memory and an increase in overall survival.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Quimiocina CXCL9/metabolismo , Factor Inhibidor de Leucemia/inmunología , Macrófagos/inmunología , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Anticuerpos Neutralizantes/farmacología , Linfocitos T CD8-positivos/metabolismo , Quimiocina CCL2/metabolismo , Epigénesis Genética , Humanos , Memoria Inmunológica , Factor Inhibidor de Leucemia/antagonistas & inhibidores , Factor Inhibidor de Leucemia/metabolismo , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones SCID , Trasplante de Neoplasias , Neoplasias/inmunología , Neoplasias/patología , Receptor de Muerte Celular Programada 1/inmunología , Microambiente Tumoral/inmunología
6.
Cell Rep ; 18(5): 1241-1255, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28147278

RESUMEN

Macrophages exert potent effector functions against invading microorganisms but constitute, paradoxically, a preferential niche for many bacterial strains to replicate. Using a model of infection by Salmonella Typhimurium, we have identified a molecular mechanism regulated by the nuclear receptor LXR that limits infection of host macrophages through transcriptional activation of the multifunctional enzyme CD38. LXR agonists reduced the intracellular levels of NAD+ in a CD38-dependent manner, counteracting pathogen-induced changes in macrophage morphology and the distribution of the F-actin cytoskeleton and reducing the capability of non-opsonized Salmonella to infect macrophages. Remarkably, pharmacological treatment with an LXR agonist ameliorated clinical signs associated with Salmonella infection in vivo, and these effects were dependent on CD38 expression in bone-marrow-derived cells. Altogether, this work reveals an unappreciated role for CD38 in bacterial-host cell interaction that can be pharmacologically exploited by activation of the LXR pathway.


Asunto(s)
Receptores X del Hígado/metabolismo , Macrófagos/metabolismo , NAD/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/patogenicidad , ADP-Ribosil Ciclasa 1/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Femenino , Masculino , Ratones , Células RAW 264.7
7.
Hum Pathol ; 56: 180-8, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27346574

RESUMEN

Carcinogenesis is a multistep process in which cancer cells and tumor stroma cells play important roles. T lymphocytes are immune constituents of tumor stroma and play a crucial function in anti-tumor response. By immunohistochemistry and flow cytometry, we studied T cytotoxic (CTLs) and T helper lymphocyte distribution and percentage in the tumor microenvironment and peripheral blood from 35 patients with endometrioid endometrial carcinomas (EEC). We also studied 23 healthy donors' blood samples as a control group. Tumor and non-tumoral endometrium samples were obtained. Immunohistochemistry revealed a high number of CTLs and T helper lymphocytes in the tumor stroma of myoinvasive EECs. T lymphocytes were mostly located in the invasive front. By flow cytometry, the percentages of CTLs and T helper lymphocytes were significantly higher in the tumor compared with the non-neoplastic endometrium (P = .0492 and P = .002). The mean fluorescence intensity of CD8 staining was lower in the tumor compared to the non-neoplastic endometrium (P = .001). There was also reduction of the mean fluorescence intensity of CD8 staining on peripheral blood from patients with grade 3 EECs compare to the peripheral blood from healthy donors (P = .0093). No alterations in the expression of granzymes A and B were found in the CTLs from the EEC cases. Finally, in a proteome profiler cytokine array we found that the growth differentiation factor 15 (GDF15) increased in blood in parallel to the tumor grade. EECs are capable of down-regulating CD8 expression of CTLs. Most likely, this effect is mediated by a soluble molecule present in plasma and is not a result of anergy or exhaustion state.


Asunto(s)
Biomarcadores de Tumor/análisis , Antígenos CD8/análisis , Carcinoma Endometrioide/inmunología , Neoplasias Endometriales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T Citotóxicos/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/sangre , Carcinoma Endometrioide/sangre , Carcinoma Endometrioide/patología , Citocinas/sangre , Regulación hacia Abajo , Neoplasias Endometriales/sangre , Neoplasias Endometriales/patología , Femenino , Factor 15 de Diferenciación de Crecimiento/sangre , Humanos , Linfocitos Infiltrantes de Tumor/patología , Persona de Mediana Edad , Clasificación del Tumor , Linfocitos T Citotóxicos/patología , Microambiente Tumoral
8.
Arch Immunol Ther Exp (Warsz) ; 60(4): 235-49, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22696047

RESUMEN

Liver X receptors (LXRs) are members of the nuclear receptor superfamily that are activated by specific oxysterols. LXRs heterodimerize with retinoid X receptors to regulate positively the expression of a variety of target genes, many of which are involved in lipid and glucose metabolism. In the last few years, new targets of LXR activation have been identified with roles in the modulation of immune responses. Moreover, LXRs mediate repression of inflammatory pathways through mechanisms collectively known as transrepression. Here, we revise recent findings on the impact of LXR activation on immune responses, with an emphasis on advances in the understanding of the molecular mechanisms that mediate these effects.


Asunto(s)
Inflamación/metabolismo , Receptores Nucleares Huérfanos/inmunología , Proteínas Represoras/inmunología , Animales , Colesterol/análogos & derivados , Colesterol/metabolismo , Regulación de la Expresión Génica/inmunología , Humanos , Metabolismo de los Lípidos , Receptores X del Hígado , Multimerización de Proteína , Receptores X Retinoide/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA