Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Semin Cancer Biol ; 64: 13-18, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31128300

RESUMEN

Glioblastoma (GBM) is an aggressive and incurable brain tumor; its malignancy has been associated with the activity of tumor infiltrating myeloid cells. Myeloid cells play important roles in the tumor control by the immune response, but also in tumor progression. Indeed, GBM exploits multiple mechanisms to recruit and modulate myeloid cells. The Aryl Hydrocarbon Receptor (AHR) is a ligand activated transcription factor implicated in the regulation of myeloid cells. In this review, we will summarize current knowledge on the AHR role in the control of myeloid cells and its impact on GBM pathogenesis.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Células Mieloides/inmunología , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Glioblastoma/inmunología , Glioblastoma/metabolismo , Humanos , Transducción de Señal
2.
Brain ; 141(7): 2066-2082, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29873694

RESUMEN

Multiple sclerosis is an inflammatory demyelinating disease in which active demyelination and neurodegeneration are associated with lymphocyte infiltrates in the brain. However, so far little is known regarding the phenotype and function of these infiltrating lymphocyte populations. In this study, we performed an in-depth phenotypic characterization of T and B cell infiltrates in a large set of multiple sclerosis cases with different disease and lesion stages and compared the findings with those seen in inflammatory, non-inflammatory and normal human controls. In multiple sclerosis lesions, we found a dominance of CD8+ T cells and a prominent contribution of CD20+ B cells in all disease courses and lesion stages, including acute multiple sclerosis cases with very short disease duration, while CD4+ T cells were sparse. A dominance of CD8+ T cells was also seen in other inflammatory controls, such as Rasmussen's encephalitis and viral encephalitis, but the contribution of B cells in these diseases was modest. Phenotypic analysis of the CD8+ T cells suggested that part of the infiltrating cells in active lesions proliferate, show an activated cytotoxic phenotype and are in part destroyed by apoptosis. Further characterization of the remaining cells suggest that CD8+ T cells acquire features of tissue-resident memory cells, which may be focally reactivated in active lesions of acute, relapsing and progressive multiple sclerosis, while B cells, at least in part, gradually transform into plasma cells. The loss of surface molecules involved in the egress of leucocytes from inflamed tissue, such as S1P1 or CCR7, and the upregulation of CD103 expression may be responsible for the compartmentalization of the inflammatory response in established lesions. Similar phenotypic changes of tissue-infiltrating CD8+ T cells were also seen in Rasmussen's encephalitis. Our data underline the potential importance of CD8+ T lymphocytes and B cells in the inflammatory response in established multiple sclerosis lesions. Tissue-resident T and B cells may represent guardians of previous inflammatory brain disease, which can be reactivated and sustain the inflammatory response, when they are re-exposed to their specific antigen.


Asunto(s)
Linfocitos/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Adulto , Anciano , Linfocitos B/inmunología , Linfocitos B/fisiología , Encéfalo/patología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/fisiología , Enfermedades Desmielinizantes/patología , Encefalitis/metabolismo , Encefalitis/patología , Femenino , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Linfocitos/fisiología , Masculino , Persona de Mediana Edad , Fenotipo
3.
Mucosal Immunol ; 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38925529

RESUMEN

Dietary proteins are taken up by intestinal dendritic cells (DCs), cleaved into peptides, loaded to major histocompatibility complexes, and presented to T cells to generate an immune response. Amino acid (AA)-diets do not have the same effects because AAs cannot bind to major histocompatibility complex to activate T cells. Here, we show that impairment in regulatory T cell generation and loss of tolerance in mice fed a diet lacking whole protein is associated with major transcriptional changes in intestinal DCs including downregulation of genes related to DC maturation, activation and decreased gene expression of immune checkpoint molecules. Moreover, the AA-diet had a profound effect on microbiome composition, including an increase in Akkermansia muciniphilia and Oscillibacter and a decrease in Lactococcus lactis and Bifidobacterium. Although microbiome transfer experiments showed that AA-driven microbiome modulates intestinal DC gene expression, most of the unique transcriptional change in DC was linked to the absence of whole protein in the diet. Our findings highlight the importance of dietary proteins for intestinal DC function and mucosal tolerance.

4.
Nat Commun ; 14(1): 4286, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37463881

RESUMEN

Traumatic brain injury (TBI) is a leading cause of morbidity and mortality. The innate and adaptive immune responses play an important role in the pathogenesis of TBI. Gamma-delta (γδ) T cells have been shown to affect brain immunopathology in multiple different conditions, however, their role in acute and chronic TBI is largely unknown. Here, we show that γδ T cells affect the pathophysiology of TBI as early as one day and up to one year following injury in a mouse model. TCRδ-/- mice are characterized by reduced inflammation in acute TBI and improved neurocognitive functions in chronic TBI. We find that the Vγ1 and Vγ4 γδ T cell subsets play opposing roles in TBI. Vγ4 γδ T cells infiltrate the brain and secrete IFN-γ and IL-17 that activate microglia and induce neuroinflammation. Vγ1 γδ T cells, however, secrete TGF-ß that maintains microglial homeostasis and dampens TBI upon infiltrating the brain. These findings provide new insights on the role of different γδ T cell subsets after brain injury and lay down the principles for the development of targeted γδ T-cell-based therapy for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Linfocitos Intraepiteliales , Masculino , Ratones , Animales , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Subgrupos de Linfocitos T , Ratones Endogámicos C57BL
5.
J Clin Invest ; 132(10)2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35298438

RESUMEN

A disequilibrium between immunosuppressive Tregs and inflammatory IL-17-producing Th17 cells is a hallmark of autoimmune diseases, including multiple sclerosis (MS). However, the molecular mechanisms underlying the Treg and Th17 imbalance in CNS autoimmunity remain largely unclear. Identifying the factors that drive this imbalance is of high clinical interest. Here, we report a major disease-promoting role for microRNA-92a (miR-92a) in CNS autoimmunity. miR-92a was elevated in experimental autoimmune encephalomyelitis (EAE), and its loss attenuated EAE. Mechanistically, miR-92a mediated EAE susceptibility in a T cell-intrinsic manner by restricting Treg induction and suppressive capacity, while supporting Th17 responses, by directly repressing the transcription factor Foxo1. Although miR-92a did not directly alter Th1 differentiation, it appeared to indirectly promote Th1 cells by inhibiting Treg responses. Correspondingly, miR-92a inhibitor therapy ameliorated EAE by concomitantly boosting Treg responses and dampening inflammatory T cell responses. Analogous to our findings in mice, miR-92a was elevated in CD4+ T cells from patients with MS, and miR-92a silencing in patients' T cells promoted Treg development but limited Th17 differentiation. Together, our results demonstrate that miR-92a drives CNS autoimmunity by sustaining the Treg/Th17 imbalance and implicate miR-92a as a potential therapeutic target for MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , MicroARNs , Esclerosis Múltiple , Linfocitos T Reguladores , Animales , Autoinmunidad , Diferenciación Celular , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Células TH1 , Células Th17
6.
Nat Commun ; 12(1): 2419, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893298

RESUMEN

Chronic inflammation can drive tumor development. Here, we have identified microRNA-146a (miR-146a) as a major negative regulator of colonic inflammation and associated tumorigenesis by modulating IL-17 responses. MiR-146a-deficient mice are susceptible to both colitis-associated and sporadic colorectal cancer (CRC), presenting with enhanced tumorigenic IL-17 signaling. Within myeloid cells, miR-146a targets RIPK2, a NOD2 signaling intermediate, to limit myeloid cell-derived IL-17-inducing cytokines and restrict colonic IL-17. Accordingly, myeloid-specific miR-146a deletion promotes CRC. Moreover, within intestinal epithelial cells (IECs), miR-146a targets TRAF6, an IL-17R signaling intermediate, to restrict IEC responsiveness to IL-17. MiR-146a within IECs further suppresses CRC by targeting PTGES2, a PGE2 synthesis enzyme. IEC-specific miR-146a deletion therefore promotes CRC. Importantly, preclinical administration of miR-146a mimic, or small molecule inhibition of the miR-146a targets, TRAF6 and RIPK2, ameliorates colonic inflammation and CRC. MiR-146a overexpression or miR-146a target inhibition represent therapeutic approaches that limit pathways converging on tumorigenic IL-17 signaling in CRC.


Asunto(s)
Carcinogénesis/genética , Neoplasias Colorrectales/genética , Inflamación/genética , MicroARNs/genética , Animales , Células Cultivadas , Colitis/genética , Colitis/metabolismo , Colitis/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-17/genética , Interleucina-17/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Transducción de Señal/genética , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo
7.
Nat Commun ; 12(1): 4907, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34389726

RESUMEN

The intestinal mucosa constitutes an environment of closely regulated immune cells. Dendritic cells (DC) interact with the gut microbiome and antigens and are important in maintaining gut homeostasis. Here, we investigate DC transcriptome, phenotype and function in five anatomical locations of the gut lamina propria (LP) which constitute different antigenic environments. We show that DC from distinct gut LP compartments induce distinct T cell differentiation and cytokine secretion. We also find that PD-L1+ DC in the duodenal LP and XCR1+ DC in the colonic LP comprise distinct tolerogenic DC subsets that are crucial for gut homeostasis. Mice lacking PD-L1+ and XCR1+ DC have a proinflammatory gut milieu associated with an increase in Th1/Th17 cells and a decrease in Treg cells and have exacerbated disease in the models of 5-FU-induced mucositis and DSS-induced colitis. Our findings identify PD-L1+ and XCR1+ DC as region-specific physiologic regulators of intestinal homeostasis.


Asunto(s)
Antígeno B7-H1/inmunología , Células Dendríticas/inmunología , Homeostasis/inmunología , Mucosa Intestinal/inmunología , Receptores de Quimiocina/inmunología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/metabolismo , Heces/microbiología , Femenino , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Homeostasis/genética , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transcriptoma/genética , Transcriptoma/inmunología
8.
iScience ; 24(11): 103347, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34820606

RESUMEN

Myeloid suppressor cells promote tumor growth by a variety of mechanisms which are not fully characterized. We identified myeloid cells (MCs) expressing the latency-associated peptide (LAP) of TGF-ß on their surface and LAPHi MCs that stimulate Foxp3+ Tregs while inhibiting effector T cell proliferation and function. Blocking TGF-ß inhibits the tolerogenic ability of LAPHi MCs. Furthermore, adoptive transfer of LAPHi MCs promotes Treg accumulation and tumor growth in vivo. Conversely, anti-LAP antibody, which reduces LAPHi MCs, slows cancer progression. Single-cell RNA-Seq analysis on tumor-derived immune cells revealed LAPHi dominated cell subsets with distinct immunosuppressive signatures, including those with high levels of MHCII and PD-L1 genes. Analogous to mice, LAP is expressed on myeloid suppressor cells in humans, and these cells are increased in glioma patients. Thus, our results identify a previously unknown function by which LAPHi MCs promote tumor growth and offer therapeutic intervention to target these cells in cancer.

9.
Mol Cell Biol ; 27(2): 526-40, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17101773

RESUMEN

Although COPI function on the early secretory pathway in eukaryotes is well established, earlier studies also proposed a nonconventional role for this coat complex in endocytosis in mammalian cells. Here we present results that suggest an involvement for specific COPI subunits in the late steps of endosomal protein sorting in Saccharomyces cerevisiae. First, we found that carboxypeptidase Y (CPY) was partially missorted to the cell surface in certain mutants of the COPIB subcomplex (COPIb; Sec27, Sec28, and possibly Sec33), which indicates an impairment in endosomal transport. Second, integral membrane proteins destined for the vacuolar lumen (i.e., carboxypeptidase S [CPS1]; Fur4, Ste2, and Ste3) accumulated at an aberrant late endosomal compartment in these mutants. The observed phenotypes for COPIb mutants resemble those of class E vacuolar protein sorting (vps) mutants that are impaired in multivesicular body (MVB) protein sorting and biogenesis. Third, we observed physical interactions and colocalization between COPIb subunits and an MVB-associated protein, Vps27. Together, our findings suggest that certain COPI subunits could have a direct role in vacuolar protein sorting to the MVB compartment.


Asunto(s)
Proteína Coat de Complejo I/metabolismo , Endosomas/metabolismo , Saccharomyces cerevisiae/metabolismo , Vacuolas/fisiología , Catepsina A/metabolismo , Proteína Coat de Complejo I/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte , Hidrolasas/metabolismo , Proteínas de la Membrana/metabolismo , Mutación , Unión Proteica , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Transporte de Proteínas , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Transporte Vesicular/metabolismo
10.
J Cell Mol Med ; 13(1): 39-53, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19175699

RESUMEN

More than 1000 microRNAs (miRNAs) are expressed in human cells, some tissue or cell type specific, others considered as house-keeping molecules. Functions and direct mRNA targets for some miRNAs have been relatively well studied over the last years. Every miRNA potentially regulates the expression of numerous protein-coding genes (tens to hundreds), but it has become increasingly clear that not all miRNAs are equally important; diverse high-throughput screenings of various systems have identified a limited number of key functional miRNAs over and over again. Particular miRNAs emerge as principal regulators that control major cell functions in various physiological and pathophysiological settings. Since its identification 3 years ago as the miRNA most commonly and strongly up-regulated in human brain tumour glioblastoma [1], miR-21 has attracted the attention of researchers in various fields, such as development, oncology, stem cell biology and aging, becoming one of the most studied miRNAs, along with let-7, miR-17-92 cluster ('oncomir-1'), miR-155 and a few others. However, an miR-21 knockout mouse has not yet been generated, and the data about miR-21 functions in normal cells are still very limited. In this review, we summarise the current knowledge of miR-21 functions in human disease, with an emphasis on its regulation, oncogenic role, targets in human cancers, potential as a disease biomarker and novel therapeutic target in oncology.


Asunto(s)
MicroARNs , Neoplasias/genética , Neoplasias/fisiopatología , Animales , Apoptosis/fisiología , Secuencia de Bases , Biomarcadores de Tumor , Ciclo Celular/fisiología , Enfermedad/genética , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Datos de Secuencia Molecular , Neoplasias/patología , Neoplasias/terapia , Procesamiento Postranscripcional del ARN , Transcripción Genética
11.
Mol Biol Cell ; 17(4): 1845-58, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16452633

RESUMEN

Gcs1 is an Arf GTPase-activating protein (Arf-GAP) that mediates Golgi-ER and post-Golgi vesicle transport in yeast. Here we show that the Snc1,2 v-SNAREs, which mediate endocytosis and exocytosis, interact physically and genetically with Gcs1. Moreover, Gcs1 and the Snc v-SNAREs colocalize to subcellular structures that correspond to the trans-Golgi and endosomal compartments. Studies performed in vitro demonstrate that the Snc-Gcs1 interaction results in the efficient binding of recombinant Arf1Delta17N-Q71L to the v-SNARE and the recruitment of purified coatomer. In contrast, the presence of Snc had no effect on Gcs1 Arf-GAP activity in vitro, suggesting that v-SNARE binding does not attenuate Arf1 function. Disruption of both the SNC and GCS1 genes results in synthetic lethality, whereas overexpression of either SNC gene inhibits the growth of a distinct subset of COPI mutants. We show that GFP-Snc1 recycling to the trans-Golgi is impaired in gcs1Delta cells and these COPI mutants. Together, these results suggest that Gcs1 facilitates the incorporation of the Snc v-SNAREs into COPI recycling vesicles and subsequent endosome-Golgi sorting in yeast.


Asunto(s)
Proteínas Fúngicas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Aparato de Golgi/metabolismo , Proteínas R-SNARE/metabolismo , Levaduras/metabolismo , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Endocitosis , Endosomas , Proteínas Fúngicas/análisis , Proteínas Fúngicas/genética , Proteínas Activadoras de GTPasa/genética , Genes Fúngicos , Genes Letales , Inmunoprecipitación , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Proteínas R-SNARE/análisis , Proteínas R-SNARE/genética , Eliminación de Secuencia , Técnicas del Sistema de Dos Híbridos , Levaduras/química
12.
Cell Rep ; 28(13): 3353-3366.e5, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31553906

RESUMEN

Smad7, a negative regulator of TGF-ß signaling, has been implicated in the pathogenesis and treatment of inflammatory bowel diseases (IBDs), including Crohn's disease (CD) and ulcerative colitis (UC). Here, we found that Smad7 mediates intestinal inflammation by limiting the PDL2/1-PD1 axis in dendritic cells (DCs) and CD4+T cells. Smad7 deficiency in DCs promotes TGF-ß responsiveness and the co-inhibitory molecules PDL2/1 on DCs, and it further imprints T cell-PD1 signaling to promote Treg differentiation. DC-specific Smad7 deletion mitigates DSS-induced colitis by inducing CD103+PDL2/1+DCs and Tregs. In addition, Smad7 deficiency in CD4+T cells promotes PD1 and PD1-induced Tregs in vitro. The transfer of Smad7-deficient CD4+T cells enhances Tregs in vivo and protects against T cell-mediated colitis. Furthermore, Smad7 antisense ameliorates DSS-induced UC, increasing TGF-ß and PDL2/1-PD1 signaling. Enhancing PD1 signaling directly via Fc-fused PDL2/1 is also beneficial. Our results identify how Smad7 mediates intestinal inflammation and leverages these pathways therapeutically, providing additional strategies for IBD intervention.


Asunto(s)
Autoinmunidad/genética , Inflamación/genética , Intestinos/patología , Proteína smad7/genética , Humanos , Transducción de Señal
13.
Nat Neurosci ; 22(5): 729-740, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30962630

RESUMEN

Tumor-associated macrophages (TAMs) play an important role in the immune response to cancer, but the mechanisms by which the tumor microenvironment controls TAMs and T cell immunity are not completely understood. Here we report that kynurenine produced by glioblastoma cells activates aryl hydrocarbon receptor (AHR) in TAMs to modulate their function and T cell immunity. AHR promotes CCR2 expression, driving TAM recruitment in response to CCL2. AHR also drives the expression of KLF4 and suppresses NF-κB activation in TAMs. Finally, AHR drives the expression of the ectonucleotidase CD39 in TAMs, which promotes CD8+ T cell dysfunction by producing adenosine in cooperation with CD73. In humans, the expression of AHR and CD39 was highest in grade 4 glioma, and high AHR expression was associated with poor prognosis. In summary, AHR and CD39 expressed in TAMs participate in the regulation of the immune response in glioblastoma and constitute potential targets for immunotherapy.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Quinurenina/metabolismo , Macrófagos/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Glioblastoma/metabolismo , Humanos , Factor 4 Similar a Kruppel , Receptores de Lipopolisacáridos/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , Factor de Transcripción STAT1 , Factor de Transcripción STAT3/metabolismo , Linfocitos T/inmunología , Microambiente Tumoral
16.
Nat Commun ; 9(1): 3151, 2018 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-30089795

RESUMEN

γδ T cells have many known functions, including the regulation of antibody responses. However, how γδ T cells control humoral immunity remains elusive. Here we show that complete Freund's adjuvant (CFA), but not alum, immunization induces a subpopulation of CXCR5-expressing γδ T cells in the draining lymph nodes. TCRγδ+CXCR5+ cells present antigens to, and induce CXCR5 on, CD4 T cells by releasing Wnt ligands to initiate the T follicular helper (Tfh) cell program. Accordingly, TCRδ-/- mice have impaired germinal center formation, inefficient Tfh cell differentiation, and reduced serum levels of chicken ovalbumin (OVA)-specific antibodies after CFA/OVA immunization. In a mouse model of lupus, TCRδ-/- mice develop milder glomerulonephritis, consistent with decreased serum levels of lupus-related autoantibodies, when compared with wild type mice. Thus, modulation of the γδ T cell-dependent humoral immune response may provide a novel therapy approach for the treatment of antibody-mediated autoimmunity.


Asunto(s)
Diferenciación Celular , Inmunidad Humoral/inmunología , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/fisiología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/fisiología , Compuestos de Alumbre , Animales , Formación de Anticuerpos , Autoanticuerpos/sangre , Pollos , Femenino , Adyuvante de Freund/inmunología , Glomerulonefritis , Inmunización , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes/inmunología , Modelos Animales , Modelos Inmunológicos , Ovalbúmina/sangre , Ovalbúmina/inmunología , Receptores CXCR5/metabolismo
17.
Trends Endocrinol Metab ; 28(6): 428-436, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28318896

RESUMEN

Glioblastoma (GBM) progression is associated with metabolic remodeling in both glioma and immune cells, resulting in the use of aerobic glycolysis as the main source of energy and biosynthetic molecules. The transcription factor hypoxia-inducible factor (HIF)-1α drives this metabolic reorganization. Oxygen levels, as well as other factors, control the activity of HIF-1α. In addition, the ligand-activated transcription factor aryl hydrocarbon receptor (AHR) modulates tumor-specific immunity and can also participate in metabolic remodeling. AHR activity is regulated by tryptophan derivatives present in the tumor microenvironment. Thus, the tumor microenvironment and signaling via HIF-1α and AHR regulate the metabolism of gliomas and immune cells, modulating tumor-specific immunity and, consequently, tumor growth. Here, we review the roles of HIF-1α and AHR in cancer and immune cell metabolism in GBM.


Asunto(s)
Glioblastoma/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Regulación de la Expresión Génica , Glioblastoma/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Oxígeno/metabolismo , Receptores de Hidrocarburo de Aril/genética
18.
Sci Immunol ; 2(11)2017 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-28763794

RESUMEN

Regulatory T cells (Tregs) promote cancer by suppressing antitumor immune responses. We found that anti-LAP antibody, which targets the latency-associated peptide (LAP)/transforming growth factor-ß (TGF-ß) complex on Tregs and other cells, enhances antitumor immune responses and reduces tumor growth in models of melanoma, colorectal carcinoma, and glioblastoma. Anti-LAP decreases LAP+ Tregs, tolerogenic dendritic cells, and TGF-ß secretion and is associated with CD8+ T cell activation. Anti-LAP increases infiltration of tumors by cytotoxic CD8+ T cells and reduces CD103+ CD8 T cells in draining lymph nodes and the spleen. We identified a role for CD103+ CD8 T cells in cancer. Tumor-associated CD103+ CD8 T cells have a tolerogenic phenotype with increased expression of CTLA-4 and interleukin-10 and decreased expression of interferon-γ, tumor necrosis factor-α, and granzymes. Adoptive transfer of CD103+ CD8 T cells promotes tumor growth, whereas CD103 blockade limits tumorigenesis. Thus, anti-LAP targets multiple immunoregulatory pathways and represents a potential approach for cancer immunotherapy.

19.
EMBO Mol Med ; 8(3): 268-87, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26881967

RESUMEN

MicroRNA-10b (miR-10b) is a unique oncogenic miRNA that is highly expressed in all GBM subtypes, while absent in normal neuroglial cells of the brain. miR-10b inhibition strongly impairs proliferation and survival of cultured glioma cells, including glioma-initiating stem-like cells (GSC). Although several miR-10b targets have been identified previously, the common mechanism conferring the miR-10b-sustained viability of GSC is unknown. Here, we demonstrate that in heterogeneous GSC, miR-10b regulates cell cycle and alternative splicing, often through the non-canonical targeting via 5'UTRs of its target genes, including MBNL1-3, SART3, and RSRC1. We have further assessed the inhibition of miR-10b in intracranial human GSC-derived xenograft and murine GL261 allograft models in athymic and immunocompetent mice. Three delivery routes for the miR-10b antisense oligonucleotide inhibitors (ASO), direct intratumoral injections, continuous osmotic delivery, and systemic intravenous injections, have been explored. In all cases, the treatment with miR-10b ASO led to targets' derepression, and attenuated growth and progression of established intracranial GBM. No significant systemic toxicity was observed upon ASO administration by local or systemic routes. Our results indicate that miR-10b is a promising candidate for the development of targeted therapies against all GBM subtypes.


Asunto(s)
Antineoplásicos/administración & dosificación , Glioblastoma/tratamiento farmacológico , MicroARNs/antagonistas & inhibidores , Oligonucleótidos Antisentido/administración & dosificación , Aloinjertos , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ratones , Resultado del Tratamiento
20.
Oncotarget ; 6(6): 3770-83, 2015 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-25738367

RESUMEN

MicroRNA-10b (miR-10b) is commonly elevated in glioblastoma (GBM), while not expressed in normal brain tissues. Targeted inhibition of miR-10b has pleiotropic effects on GBM derived cell lines, it reduces GBM growth in animal models, but does not affect normal neurons and astrocytes. This data raises the possibility of developing miR-10b-targeting GBM therapy. However, the mechanisms contributing to miR-10b-mediated glioma cell survival and proliferation are unexplored. We found that inhibition of miR-10b has distinct effects on specific glioma cell lines. In cells expressing high levels of tumor suppressor p21WAF1/Cip1, it represses E2F1-mediated transcription, leading to down-regulation of multiple E2F1 target genes encoding for S-phase specific proteins, epigenetic modulators, and miRNAs (e.g. miR-15/16), and thereby stalling progression through the S-phase of cell cycle. Subsequently, miR-15/16 activities are reduced and many of their direct targets are de-repressed, including ubiquitin ligase FBXW7 that destabilizes Cyclin E. Conversely, GBM cells expressing low p21 level, or after p21 knock-down, exhibit weaker or no E2F1 response to miR-10b inhibition. Comparative analysis of The Cancer Genome Atlas revealed a strong correlation between miR-10b and multiple E2F target genes in GBM and low-grade glioma. Taken together, these findings indicate that miR-10b regulates E2F1-mediated transcription in GBM, in a p21-dependent fashion.


Asunto(s)
Neoplasias Encefálicas/genética , Factor de Transcripción E2F1/genética , Glioblastoma/genética , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Factor de Transcripción E2F1/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , MicroARNs/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA