Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Genomics ; 18(1): 243, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28327095

RESUMEN

BACKGROUND: Although tumor-associated macrophages (TAMs) are essential for cancer progression, connections between different clinical outcomes and transcriptional networks have not been reported. We have addressed this issue by analyzing global expression patterns of TAMs isolated from the ascites of ovarian cancer patients. RESULTS: TAMs isolated from different ovarian cancer patients can be stratified by coexpression or principal component analysis into subgroups with specific biological features and associated with distinct clinical outcomes. A hallmark of subgroup A is a high expression of clinically unfavorable markers, including (i) high CD163 expression, a surface receptor characteristic of an anti-inflammatory activation state, (ii) increased PCOLCE2 expression, indicative of enhanced extracellular matrix organization, and (iii) elevated ascites levels of IL-6 and IL-10, linked to the aggressiveness of ovarian cancer and immune suppression. In contrast, subgroup B TAMs are characterized by the upregulation of genes linked to immune defense mechanisms and interferon (IFN) signaling. Intriguingly, analysis of published data for 1763 ovarian cancer patients revealed a strong association of this transcriptional signature with a longer overall survival. Consistent with these results, IFNγ was able to abrogate the suppressive effect of ovarian cancer ascites on the inducibility of IL12B expression and IL-12 secretion, a key determinant of a cytotoxic immune response. CONCLUSIONS: The survival of ovarian cancer patients is linked to the presence of TAMs with a transcriptional signature that is characterized by a low expression of protumorigenic and immunosuppressive markers and an upregulation of genes linked to interferon signaling. The observed IFNγ-mediated restoration of the inducibility of IL-12 in the presence of ascites provides a possible explanation for the association of an interferon signaling-associated signature with a favorable clinical outcome.


Asunto(s)
Ascitis/patología , Interferones/metabolismo , Macrófagos/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/mortalidad , Transducción de Señal , Biomarcadores , Análisis por Conglomerados , Citocinas , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunofenotipificación , Macrófagos/patología , Neoplasias Ováricas/patología , Pronóstico , Reproducibilidad de los Resultados , Transcriptoma , Microambiente Tumoral
2.
Oncotarget ; 7(46): 75339-75352, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27659538

RESUMEN

Macrophages occur as resident cells of fetal origin or as infiltrating blood monocyte-derived cells. Despite the critical role of tumor-associated macrophages (TAMs) in tumor progression, the contribution of these developmentally and functionally distinct macrophage subsets and their alteration by the tumor microenvironment are poorly understood. We have addressed this question by comparing TAMs from human ovarian carcinoma ascites, resident peritoneal macrophages (pMPHs) and monocyte-derived macrophages (MDMs). Our study revealed striking a similarity between TAMs and pMPHs, which was considerably greater that the resemblance of TAMs and MDMs, including their transcriptomes, their inflammation-related activation state, the presence of receptors mediating immune functions and the expression of tumor-promoting mediators. Consistent with these results, TAMs phagocytized bacteria, presented peptide antigens and activated cytotoxic T cells within their pathophysiological environment. These observations support the notion that tumor-promoting properties of TAMs may reflect, at least to some extent, normal features of resident macrophages rather than functions induced by the tumor microenvironment. In spite of these surprising similarities between TAMs and pMPHs, bioinformatic analyses identified a TAM-selective signature of 30 genes that are upregulated relative to both pMPHs and MDMs. The majority of these genes is linked to extracellular matrix (ECM) remodeling, supporting a role for TAMs in cancer cell invasion and ovarian cancer progression.


Asunto(s)
Matriz Extracelular/metabolismo , Macrófagos/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Transcriptoma , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Macrófagos/patología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Fagocitosis , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
3.
Oncotarget ; 6(15): 13416-33, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-25968567

RESUMEN

The nuclear receptor peroxisome proliferator-activated receptor ß/δ (PPARß/δ) is a lipid ligand-inducible transcription factor associated with macrophage polarization. However, its function in tumor-associated macrophages (TAMs) has not been investigated to date. Here, we report the PPARß/δ-regulated transcriptome and cistrome for TAMs from ovarian carcinoma patients. Comparison with monocyte-derived macrophages shows that the vast majority of direct PPARß/δ target genes are upregulated in TAMs and largely refractory to synthetic agonists, but repressible by inverse agonists. Besides genes with metabolic functions, these include cell type-selective genes associated with immune regulation and tumor progression, e.g., LRP5, CD300A, MAP3K8 and ANGPTL4. This deregulation is not due to increased expression of PPARß/δ or its enhanced recruitment to target genes. Instead, lipidomic analysis of malignancy-associated ascites revealed high concentrations of polyunsaturated fatty acids, in particular linoleic acid, acting as potent PPARß/δ agonists in macrophages. These fatty acid ligands accumulate in lipid droplets in TAMs, thereby providing a reservoir of PPARß/δ ligands. These observations suggest that the deregulation of PPARß/δ target genes by ligands of the tumor microenvironment contributes to the pro-tumorigenic polarization of ovarian carcinoma TAMs. This conclusion is supported by the association of high ANGPTL4 expression with a shorter relapse-free survival in serous ovarian carcinoma.


Asunto(s)
Ácido Linoleico/genética , Macrófagos/patología , Neoplasias Ováricas/sangre , Neoplasias Ováricas/genética , PPAR delta/genética , PPAR-beta/genética , Microambiente Tumoral/genética , Animales , Estudios de Casos y Controles , Ácidos Grasos , Femenino , Humanos , Ligandos , Ácido Linoleico/sangre , Macrófagos/metabolismo , Ratones , Recurrencia Local de Neoplasia/sangre , Recurrencia Local de Neoplasia/genética , PPAR delta/sangre , PPAR-beta/sangre
4.
Nucleic Acids Res ; 43(10): 5033-51, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25934804

RESUMEN

Peroxisome proliferator-activated receptor ß/δ (PPARß/δ) is a lipid ligand-inducible transcription factor with established metabolic functions, whereas its anti-inflammatory function is poorly understood. To address this issue, we determined the global PPARß/δ-regulated signaling network in human monocyte-derived macrophages. Besides cell type-independent, canonical target genes with metabolic and immune regulatory functions we identified a large number of inflammation-associated NFκB and STAT1 target genes that are repressed by agonists. Accordingly, PPARß/δ agonists inhibited the expression of multiple pro-inflammatory mediators and induced an anti-inflammatory, IL-4-like morphological phenotype. Surprisingly, bioinformatic analyses also identified immune stimulatory effects. Consistent with this prediction, PPARß/δ agonists enhanced macrophage survival under hypoxic stress and stimulated CD8(+) T cell activation, concomitantly with the repression of immune suppressive target genes and their encoded products CD274 (PD-1 ligand), CD32B (inhibitory Fcγ receptor IIB) and indoleamine 2,3-dioxygenase 1 (IDO-1), as well as a diminished release of the immune suppressive IDO-1 metabolite kynurenine. Comparison with published data revealed a significant overlap of the PPARß/δ transcriptome with coexpression modules characteristic of both anti-inflammatory and pro-inflammatory cytokines. Our findings indicate that PPARß/δ agonists induce a unique macrophage activation state with strong anti-inflammatory but also specific immune stimulatory components, pointing to a context-dependent function of PPARß/δ in immune regulation.


Asunto(s)
Redes Reguladoras de Genes , Activación de Macrófagos , Macrófagos/inmunología , PPAR delta/metabolismo , PPAR-beta/metabolismo , Diferenciación Celular , Línea Celular , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Macrófagos/metabolismo , Monocitos/citología , Monocitos/metabolismo , PPAR delta/agonistas , PPAR-beta/agonistas , Transcriptoma
5.
Int J Cancer ; 134(1): 32-42, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23784932

RESUMEN

Ovarian cancer is typically accompanied by the occurrence of malignant ascites containing large number of macrophages. It has been suggested that these tumor-associated macrophages (TAMs) are skewed to alternative polarization (M2) and thereby play an essential role in therapy resistance and metastatic spread. In our study, we have investigated the nature, regulation and clinical correlations of TAM polarization in serous ovarian cancer. Macrophage polarization markers on TAMs and ascites cytokine levels were analyzed for 30 patients and associated with relapse-free survival (RFS) in a prospective study with 20 evaluable patients. Surface expression of the M2 marker CD163 on TAMs was inversely associated with RFS (p < 0.01). However, global gene expression profiles determined for 17 of these patients revealed a mixed-polarization phenotype unrelated to the M1/M2 classification. CD163 surface expression also correlated with the ascites levels of IL-6 and IL-10 (p < 0.05), both cytokines induced CD163 expression, and their ascites levels showed a clear inverse association with RFS (p < 0.01). These findings define a subgroup of patients with high CD163 expression, high IL-6 and/or IL-10 levels and poor clinical outcome.


Asunto(s)
Ascitis/inmunología , Cistadenocarcinoma Seroso/inmunología , Macrófagos/inmunología , Neoplasias Ováricas/inmunología , Antígenos CD/biosíntesis , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/biosíntesis , Antígenos de Diferenciación Mielomonocítica/inmunología , Polaridad Celular , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patología , Citocinas/biosíntesis , Citocinas/inmunología , Supervivencia sin Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Estimación de Kaplan-Meier , Macrófagos/metabolismo , Macrófagos/patología , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fenotipo , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/inmunología , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA