Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Cell Biol ; 25(9): 1332-1345, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37605008

RESUMEN

MacroH2A has established tumour suppressive functions in melanoma and other cancers, but an unappreciated role in the tumour microenvironment. Using an autochthonous, immunocompetent mouse model of melanoma, we demonstrate that mice devoid of macroH2A variants exhibit increased tumour burden compared with wild-type counterparts. MacroH2A-deficient tumours accumulate immunosuppressive monocytes and are depleted of functional cytotoxic T cells, characteristics consistent with a compromised anti-tumour response. Single cell and spatial transcriptomics identify increased dedifferentiation along the neural crest lineage of the tumour compartment and increased frequency and activation of cancer-associated fibroblasts following macroH2A loss. Mechanistically, macroH2A-deficient cancer-associated fibroblasts display increased myeloid chemoattractant activity as a consequence of hyperinducible expression of inflammatory genes, which is enforced by increased chromatin looping of their promoters to enhancers that gain H3K27ac. In summary, we reveal a tumour suppressive role for macroH2A variants through the regulation of chromatin architecture in the tumour stroma with potential implications for human melanoma.


Asunto(s)
Fibroblastos Asociados al Cáncer , Histonas , Melanoma , Animales , Ratones , Cromatina/genética , Expresión Génica , Histonas/genética , Melanoma/genética , Microambiente Tumoral/genética
2.
Cancer Discov ; 12(11): 2606-2625, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36027053

RESUMEN

It is currently accepted that cancer-associated fibroblasts (CAF) participate in T-cell exclusion from tumor nests. To unbiasedly test this, we used single-cell RNA sequencing coupled with multiplex imaging on a large cohort of lung tumors. We identified four main CAF populations, two of which are associated with T-cell exclusion: (i) MYH11+αSMA+ CAF, which are present in early-stage tumors and form a single cell layer lining cancer aggregates, and (ii) FAP+αSMA+ CAF, which appear in more advanced tumors and organize in patches within the stroma or in multiple layers around tumor nests. Both populations orchestrate a particular structural tissue organization through dense and aligned fiber deposition compared with T cell-permissive CAF. Yet they produce distinct matrix molecules, including collagen IV (MYH11+αSMA+ CAF) and collagen XI/XII (FAP+αSMA+ CAF). Hereby, we uncovered unique molecular programs of CAF driving T-cell marginalization, whose targeting should increase immunotherapy efficacy in patients bearing T cell-excluded tumors. SIGNIFICANCE: The cellular and molecular programs driving T-cell marginalization in solid tumors remain unclear. Here, we describe two CAF populations associated with T-cell exclusion in human lung tumors. We demonstrate the importance of pairing molecular and spatial analysis of the tumor microenvironment, a prerequisite to developing new strategies targeting T cell-excluding CAF. See related commentary by Sherman, p. 2501. This article is highlighted in the In This Issue feature, p. 2483.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias Pulmonares , Humanos , Fibroblastos Asociados al Cáncer/patología , Linfocitos T , Microambiente Tumoral , Inmunoterapia/métodos , Neoplasias Pulmonares/patología , Fibroblastos
4.
Nature ; 580(7802): 257-262, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32269339

RESUMEN

Checkpoint blockade therapies have improved cancer treatment, but such immunotherapy regimens fail in a large subset of patients. Conventional type 1 dendritic cells (DC1s) control the response to checkpoint blockade in preclinical models and are associated with better overall survival in patients with cancer, reflecting the specialized ability of these cells to prime the responses of CD8+ T cells1-3. Paradoxically, however, DC1s can be found in tumours that resist checkpoint blockade, suggesting that the functions of these cells may be altered in some lesions. Here, using single-cell RNA sequencing in human and mouse non-small-cell lung cancers, we identify a cluster of dendritic cells (DCs) that we name 'mature DCs enriched in immunoregulatory molecules' (mregDCs), owing to their coexpression of immunoregulatory genes (Cd274, Pdcd1lg2 and Cd200) and maturation genes (Cd40, Ccr7 and Il12b). We find that the mregDC program is expressed by canonical DC1s and DC2s upon uptake of tumour antigens. We further find that upregulation of the programmed death ligand 1 protein-a key checkpoint molecule-in mregDCs is induced by the receptor tyrosine kinase AXL, while upregulation of interleukin (IL)-12 depends strictly on interferon-γ and is controlled negatively by IL-4 signalling. Blocking IL-4 enhances IL-12 production by tumour-antigen-bearing mregDC1s, expands the pool of tumour-infiltrating effector T cells and reduces tumour burden. We have therefore uncovered a regulatory module associated with tumour-antigen uptake that reduces DC1 functionality in human and mouse cancers.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/patología , Neoplasias Pulmonares/inmunología , Animales , Antígenos de Neoplasias/inmunología , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Humanos , Inmunoterapia , Interferón gamma/inmunología , Interleucina-12/inmunología , Interleucina-4/antagonistas & inhibidores , Interleucina-4/inmunología , Interleucina-4/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Masculino , Ratones , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
5.
Cell ; 178(5): 1102-1114.e17, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442403

RESUMEN

Caloric restriction is known to improve inflammatory and autoimmune diseases. However, the mechanisms by which reduced caloric intake modulates inflammation are poorly understood. Here we show that short-term fasting reduced monocyte metabolic and inflammatory activity and drastically reduced the number of circulating monocytes. Regulation of peripheral monocyte numbers was dependent on dietary glucose and protein levels. Specifically, we found that activation of the low-energy sensor 5'-AMP-activated protein kinase (AMPK) in hepatocytes and suppression of systemic CCL2 production by peroxisome proliferator-activator receptor alpha (PPARα) reduced monocyte mobilization from the bone marrow. Importantly, we show that fasting improves chronic inflammatory diseases without compromising monocyte emergency mobilization during acute infectious inflammation and tissue repair. These results reveal that caloric intake and liver energy sensors dictate the blood and tissue immune tone and link dietary habits to inflammatory disease outcome.


Asunto(s)
Restricción Calórica , Monocitos/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Adulto , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Quimiocina CCL2/deficiencia , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Femenino , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , PPAR alfa/deficiencia , PPAR alfa/genética , PPAR alfa/metabolismo
6.
J Exp Med ; 216(10): 2265-2281, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31350310

RESUMEN

Microglia, the brain resident macrophages, critically shape forebrain neuronal circuits. However, their precise function in the cerebellum is unknown. Here we show that human and mouse cerebellar microglia express a unique molecular program distinct from forebrain microglia. Cerebellar microglial identity was driven by the CSF-1R ligand CSF-1, independently of the alternate CSF-1R ligand, IL-34. Accordingly, CSF-1 depletion from Nestin+ cells led to severe depletion and transcriptional alterations of cerebellar microglia, while microglia in the forebrain remained intact. Strikingly, CSF-1 deficiency and alteration of cerebellar microglia were associated with reduced Purkinje cells, altered neuronal function, and defects in motor learning and social novelty interactions. These findings reveal a novel CSF-1-CSF-1R signaling-mediated mechanism that contributes to motor function and social behavior.


Asunto(s)
Conducta Animal/fisiología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Microglía/metabolismo , Actividad Motora/fisiología , Células de Purkinje/metabolismo , Transducción de Señal/fisiología , Conducta Social , Animales , Humanos , Factor Estimulante de Colonias de Macrófagos/genética , Ratones , Ratones Transgénicos , Células de Purkinje/citología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo
7.
Immunity ; 44(4): 924-38, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27096321

RESUMEN

Large numbers of melanoma lesions develop resistance to targeted inhibition of mutant BRAF or fail to respond to checkpoint blockade. We explored whether modulation of intratumoral antigen-presenting cells (APCs) could increase responses to these therapies. Using mouse melanoma models, we found that CD103(+) dendritic cells (DCs) were the only APCs transporting intact antigens to the lymph nodes and priming tumor-specific CD8(+) T cells. CD103(+) DCs were required to promote anti-tumoral effects upon blockade of the checkpoint ligand PD-L1; however, PD-L1 inhibition only led to partial responses. Systemic administration of the growth factor FLT3L followed by intratumoral poly I:C injections expanded and activated CD103(+) DC progenitors in the tumor, enhancing responses to BRAF and PD-L1 blockade and protecting mice from tumor rechallenge. Thus, the paucity of activated CD103(+) DCs in tumors limits checkpoint-blockade efficacy and combined FLT3L and poly I:C therapy can enhance tumor responses to checkpoint and BRAF blockade.


Asunto(s)
Antígenos CD/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Cadenas alfa de Integrinas/metabolismo , Melanoma Experimental/inmunología , Poli I-C/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/farmacología , Animales , Presentación de Antígeno/inmunología , Línea Celular Tumoral , Células Dendríticas/citología , Ratones Endogámicos C57BL , Ratones Noqueados
8.
Cell Rep ; 9(5): 1856-1870, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25466255

RESUMEN

Matrix metalloproteinase-2 (MMP-2) is involved in several physiological mechanisms, including wound healing and tumor progression. We show that MMP-2 directly stimulates dendritic cells (DCs) to both upregulate OX40L on the cell surface and secrete inflammatory cytokines. The mechanism underlying DC activation includes physical association with Toll-like receptor-2 (TLR2), leading to NF-κB activation, OX40L upregulation on DCs, and ensuing TH2 differentiation. Significantly, MMP-2 polarizes T cells toward type 2 responses in vivo, in a TLR2-dependent manner. MMP-2-dependent type 2 polarization may represent a key immune regulatory mechanism for protection against a broad array of disorders, such as inflammatory, infectious, and autoimmune diseases, which can be hijacked by tumors to evade immunity.


Asunto(s)
Células Dendríticas/inmunología , Metaloproteinasa 2 de la Matriz/fisiología , Receptor Toll-Like 2/metabolismo , Animales , Células Dendríticas/enzimología , Células HEK293 , Humanos , Lipopolisacáridos/farmacología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Ligando OX40/metabolismo , Unión Proteica , Transducción de Señal , Linfocitos T/enzimología , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/fisiología
9.
Cell ; 158(3): 506-21, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25083866

RESUMEN

Adaptation of the endoplasmic reticulum (ER) pathway for MHC class I (MHC-I) presentation in dendritic cells enables cross-presentation of peptides derived from phagocytosed microbes, infected cells, or tumor cells to CD8 T cells. How these peptides intersect with MHC-I molecules remains poorly understood. Here, we show that MHC-I selectively accumulate within phagosomes carrying microbial components, which engage Toll-like receptor (TLR) signaling. Although cross-presentation requires Sec22b-mediated phagosomal recruitment of the peptide loading complex from the ER-Golgi intermediate compartment (ERGIC), this step is independent of TLR signaling and does not deliver MHC-I. Instead, MHC-I are recruited from an endosomal recycling compartment (ERC), which is marked by Rab11a, VAMP3/cellubrevin, and VAMP8/endobrevin and holds large reserves of MHC-I. While Rab11a activity stocks ERC stores with MHC-I, MyD88-dependent TLR signals drive IκB-kinase (IKK)2-mediated phosphorylation of phagosome-associated SNAP23. Phospho-SNAP23 stabilizes SNARE complexes orchestrating ERC-phagosome fusion, enrichment of phagosomes with ERC-derived MHC-I, and subsequent cross-presentation during infection.


Asunto(s)
Presentación de Antígeno , Endosomas/metabolismo , Fagosomas/metabolismo , Receptores Toll-Like/metabolismo , Animales , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Tejido Linfoide , Ratones , Ovalbúmina/inmunología , Fagocitosis , Fosforilación , Transporte de Proteínas , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Receptores Toll-Like/inmunología , Proteínas de Unión al GTP rab/metabolismo
10.
Nat Immunol ; 15(1): 54-62, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24270517

RESUMEN

miR-126 is a microRNA expressed predominately by endothelial cells and controls angiogenesis. We found miR-126 was required for the innate response to pathogen-associated nucleic acids and that miR-126-deficient mice had greater susceptibility to infection with pseudotyped HIV. Profiling of miRNA indicated that miR-126 had high and specific expression by plasmacytoid dendritic cells (pDCs). Moreover, miR-126 controlled the survival and function of pDCs and regulated the expression of genes encoding molecules involved in the innate response, including Tlr7, Tlr9 and Nfkb1, as well as Kdr, which encodes the growth factor receptor VEGFR2. Deletion of Kdr in DCs resulted in reduced production of type I interferon, which supports the proposal of a role for VEGFR2 in miR-126 regulation of pDCs. Our studies identify the miR-126-VEGFR2 axis as an important regulator of the innate response that operates through multiscale control of pDCs.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Innata/inmunología , MicroARNs/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Animales , Células Dendríticas/metabolismo , Citometría de Flujo , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Inmunidad Innata/genética , Immunoblotting , Interferón-alfa/sangre , Interferón-alfa/inmunología , Interferón-alfa/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , MicroARNs/genética , MicroARNs/metabolismo , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/inmunología , Subunidad p50 de NF-kappa B/metabolismo , Ácidos Nucleicos/inmunología , Ácidos Nucleicos/metabolismo , Oligodesoxirribonucleótidos/genética , Oligodesoxirribonucleótidos/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología , Receptor Toll-Like 9/metabolismo , Transcriptoma/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Nat Methods ; 9(8): 840-6, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22751203

RESUMEN

We introduce two large-scale resources for functional analysis of microRNA (miRNA): a decoy library for inhibiting miRNA function and a sensor library for monitoring microRNA activity. To take advantage of the sensor library, we developed a high-throughput assay called Sensor-seq to simultaneously quantify the activity of hundreds of miRNAs. Using this approach, we show that only the most abundant miRNAs in a cell mediate target suppression. Over 60% of detected miRNAs had no discernible activity, which indicated that the functional 'miRNome' of a cell is considerably smaller than currently inferred from profiling studies. Moreover, some highly expressed miRNAs exhibited relatively weak activity, which in some cases correlated with a high target-to-miRNA ratio or increased nuclear localization of the miRNA. Finally, we show that the miRNA decoy library can be used for pooled loss-of-function studies. These tools are valuable resources for studying miRNA biology and for miRNA-based therapeutics.


Asunto(s)
Técnicas Biosensibles , Biblioteca de Genes , Vectores Genéticos/genética , Ensayos Analíticos de Alto Rendimiento , MicroARNs/genética , MicroARNs/metabolismo , Animales , Humanos , MicroARNs/antagonistas & inhibidores
12.
Prostate ; 72(12): 1339-50, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22228025

RESUMEN

BACKGROUND: Elevated TGF-ß levels are associated with prostate cancer progression. Although TGF-ß is a tumor suppressor for normal epithelial and early-stage cancer cells, it may act paradoxically as a tumor promoter in more advanced cancers, although its effects are largely cell and context dependent. This study analyzed prostate cancer responses to TGF-ß signaling in an isogenic model of androgen-sensitive and castration-resistant prostate cancer cells. METHODS: Phosphorylation and nuclear translocation of Smad2 and Smad3 were analyzed using immunoblotting. Proliferation and cell cycle responses to TGF-ß1 (5 ng/ml) were assessed using growth assays and flow cytometry for DNA content, as well as Western blot and immunoprecipitation of cell cycle proteins. RESULTS: Both androgen-sensitive (LNCaP) and castration-resistant (C4-2 and C4-2B) prostate cancer cell lines demonstrated TGF-ß1-induced phosphorylation and nuclear translocation of Smad2/3 that was robust in metastatic lines. Smad phosphorylation was completely abrogated with inhibition of ALK-5 kinase activity using the kinase inhibitor, SB-431542. Increased sensitivity to TGF-ß1-mediated growth inhibition was observed in C4-2 and C4-2B cells, as compared to LNCaP cells. This was paralleled with downregulation of Cyclin D and increased association of p15(Ink4b) or p27(Kip) with CDK's. Additionally, TGF-ß1 inhibited motility and invasion of metastatic cell lines. CONCLUSIONS: TGF-ß-mediated suppression of growth and motility is enhanced in metastatic, castration-resistant prostate cancer cells. Enhanced TGF-ß1-induced Smad2 and -3 signaling in prostate cancer cells may correlate with tumor suppressive activity. Therefore, the direct effects of TGF-ß1 on prostate cancer cells post-castration may be anti-tumorigenic and growth-suppressive.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular , Inhibidores de Crecimiento/fisiología , Neoplasias de la Próstata/patología , Transducción de Señal/fisiología , Proteína Smad2/fisiología , Proteína smad3/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Transporte Activo de Núcleo Celular/fisiología , Línea Celular Tumoral , Humanos , Masculino , Orquiectomía , Fosforilación/fisiología , Neoplasias de la Próstata/terapia , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...