Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Immunol ; 8(88): eadf8838, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37889984

RESUMEN

In chronic infections and cancer, T cells are exposed to prolonged antigen stimulation, resulting in loss of function (or exhaustion) and impairment of effective immunological protection. Exhausted T cells are heterogeneous and include early progenitors (Tpex) and terminally exhausted cells (Tex). Here, we used bulk and single-cell transcriptomics to analyze expression of transposable elements (TEs) in subpopulations of mouse and human CD8+ tumor-infiltrating T lymphocytes (TILs). We show that in mice, members of the virus-like murine VL30 TE family (mostly intact, evolutionary young ERV1s) are strongly repressed in terminally exhausted CD8+ T cells in both tumor and viral models of exhaustion. Tpex expression of these VL30s, which are mainly intergenic and transcribed independently of their closest gene neighbors, was driven by Fli1, a transcription factor involved in progression from Tpex to Tex. Immune checkpoint blockade (ICB) in both mice and patients with cancer increased TE expression (including VL30 in mice), demonstrating that TEs may be applicable as ICB response biomarkers. We conclude that expression of TEs is tightly regulated in TILs during establishment of exhaustion and reprogramming by ICB. Analyses of TE expression on single cells and bulk populations open opportunities for understanding immune cell identity and heterogeneity, as well as for defining cellular gene expression signatures and disease biomarkers.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Animales , Ratones , Humanos , Elementos Transponibles de ADN/genética , Agotamiento de Células T , Biomarcadores
3.
Cell Rep ; 39(10): 110916, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35675780

RESUMEN

We analyze transposable elements (TEs) in glioblastoma (GBM) patients using a proteogenomic pipeline that combines single-cell transcriptomics, bulk RNA sequencing (RNA-seq) samples from tumors and healthy-tissue cohorts, and immunopeptidomic samples. We thus identify 370 human leukocyte antigen (HLA)-I-bound peptides encoded by TEs differentially expressed in GBM. Some of the peptides are encoded by repeat sequences from intact open reading frames (ORFs) present in up to several hundred TEs from recent long interspersed nuclear element (LINE)-1, long terminal repeat (LTR), and SVA subfamilies. Other HLA-I-bound peptides are encoded by single copies of TEs from old subfamilies that are expressed recurrently in GBM tumors and not expressed, or very infrequently and at low levels, in healthy tissues (including brain). These peptide-coding, GBM-specific, highly recurrent TEs represent potential tumor-specific targets for cancer immunotherapies.


Asunto(s)
Glioblastoma , Antígenos de Histocompatibilidad Clase I , Proteogenómica , Elementos Transponibles de ADN , Glioblastoma/genética , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Péptidos/genética , RNA-Seq
4.
Nat Commun ; 13(1): 3739, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35768432

RESUMEN

Tumor-infiltrating CD8 + T cells progressively lose functionality and fail to reject tumors. The underlying mechanism and re-programing induced by checkpoint blockers are incompletely understood. We show here that genetic ablation or pharmacological inhibition of histone lysine methyltransferase Suv39h1 delays tumor growth and potentiates tumor rejection by anti-PD-1. In the absence of Suv39h1, anti-PD-1 induces alternative activation pathways allowing survival and differentiation of IFNγ and Granzyme B producing effector cells that express negative checkpoint molecules, but do not reach final exhaustion. Their transcriptional program correlates with that of melanoma patients responding to immune-checkpoint blockade and identifies the emergence of cytolytic-effector tumor-infiltrating lymphocytes as a biomarker of clinical response. Anti-PD-1 favors chromatin opening in loci linked to T-cell activation, memory and pluripotency, but in the absence of Suv39h1, cells acquire accessibility in cytolytic effector loci. Overall, Suv39h1 inhibition enhances anti-tumor immune responses, alone or combined with anti-PD-1, suggesting that Suv39h1 is an "epigenetic checkpoint" for tumor immunity.


Asunto(s)
Linfocitos T CD8-positivos , Melanoma , Metiltransferasas , Receptor de Muerte Celular Programada 1 , Proteínas Represoras , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Epigénesis Genética , Humanos , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma/genética , Melanoma/inmunología , Melanoma/terapia , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/genética , Metiltransferasas/inmunología , Metiltransferasas/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(14): 7905-7916, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32193341

RESUMEN

Transposable elements (TEs) compose nearly half of mammalian genomes and provide building blocks for cis-regulatory elements. Using high-throughput sequencing, we show that 84 TE subfamilies are overrepresented, and distributed in a lineage-specific fashion in core and boundary domains of CD8+ T cell enhancers. Endogenous retroviruses are most significantly enriched in core domains with accessible chromatin, and bear recognition motifs for immune-related transcription factors. In contrast, short interspersed elements (SINEs) are preferentially overrepresented in nucleosome-containing boundaries. A substantial proportion of these SINEs harbor a high density of the enhancer-specific histone mark H3K4me1 and carry sequences that match enhancer boundary nucleotide composition. Motifs with regulatory features are better preserved within enhancer-enriched TE copies compared to their subfamily equivalents located in gene deserts. TE-rich and TE-poor enhancers associate with both shared and unique gene groups and are enriched in overlapping functions related to lymphocyte and leukocyte biology. The majority of T cell enhancers are shared with other immune lineages and are accessible in common hematopoietic progenitors. A higher proportion of immune tissue-specific enhancers are TE-rich compared to enhancers specific to other tissues, correlating with higher TE occurrence in immune gene-associated genomic regions. Our results suggest that during evolution, TEs abundant in these regions and carrying motifs potentially beneficial for enhancer architecture and immune functions were particularly frequently incorporated by evolving enhancers. Their putative selection and regulatory cooption may have accelerated the evolution of immune regulatory networks.


Asunto(s)
Elementos Transponibles de ADN/genética , Elementos de Facilitación Genéticos/genética , Evolución Molecular , Linfocitos T/inmunología , Animales , Cromatina/genética , Cromatina/inmunología , Elementos Transponibles de ADN/inmunología , Retrovirus Endógenos/genética , Retrovirus Endógenos/inmunología , Elementos de Facilitación Genéticos/inmunología , Redes Reguladoras de Genes/genética , Genoma Humano/genética , Genoma Humano/inmunología , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Elementos de Nucleótido Esparcido Corto/genética , Elementos de Nucleótido Esparcido Corto/inmunología
6.
Proc Natl Acad Sci U S A ; 116(51): 25839-25849, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31776254

RESUMEN

Naive CD4+ T lymphocytes differentiate into different effector types, including helper and regulatory cells (Th and Treg, respectively). Heritable gene expression programs that define these effector types are established during differentiation, but little is known about the epigenetic mechanisms that install and maintain these programs. Here, we use mice defective for different components of heterochromatin-dependent gene silencing to investigate the epigenetic control of CD4+ T cell plasticity. We show that, upon T cell receptor (TCR) engagement, naive and regulatory T cells defective for TRIM28 (an epigenetic adaptor for histone binding modules) or for heterochromatin protein 1 ß and γ isoforms (HP1ß/γ, 2 histone-binding factors involved in gene silencing) fail to effectively signal through the PI3K-AKT-mTOR axis and switch to glycolysis. While differentiation of naive TRIM28-/- T cells into cytokine-producing effector T cells is impaired, resulting in reduced induction of autoimmune colitis, TRIM28-/- regulatory T cells also fail to expand in vivo and to suppress autoimmunity effectively. Using a combination of transcriptome and chromatin immunoprecipitation-sequencing (ChIP-seq) analyses for H3K9me3, H3K9Ac, and RNA polymerase II, we show that reduced effector differentiation correlates with impaired transcriptional silencing at distal regulatory regions of a defined set of Treg-associated genes, including, for example, NRP1 or Snai3. We conclude that TRIM28 and HP1ß/γ control metabolic reprograming through epigenetic silencing of a defined set of Treg-characteristic genes, thus allowing effective T cell expansion and differentiation into helper and regulatory phenotypes.


Asunto(s)
Diferenciación Celular/fisiología , Reprogramación Celular/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Epigénesis Genética/fisiología , Linfocitos T/metabolismo , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Animales , Autoinmunidad/fisiología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Plasticidad de la Célula/fisiología , Reprogramación Celular/genética , Homólogo de la Proteína Chromobox 5 , Colon/patología , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Silenciador del Gen , Histonas/metabolismo , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Transcriptoma , Proteína 28 que Contiene Motivos Tripartito/genética
8.
Cell Rep ; 26(9): 2377-2393.e13, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30811988

RESUMEN

Cytosolic DNA activates cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS), an innate immune sensor pivotal in anti-microbial defense, senescence, auto-immunity, and cancer. cGAS is considered to be a sequence-independent DNA sensor with limited access to nuclear DNA because of compartmentalization. However, the nuclear envelope is a dynamic barrier, and cGAS is present in the nucleus. Here, we identify determinants of nuclear cGAS localization and activation. We show that nuclear-localized cGAS synthesizes cGAMP and induces innate immune activation of dendritic cells, although cGAMP levels are 200-fold lower than following transfection with exogenous DNA. Using cGAS ChIP-seq and a GFP-cGAS knockin mouse, we find nuclear cGAS enrichment on centromeric satellite DNA, confirmed by imaging, and to a lesser extent on LINE elements. The non-enzymatic N-terminal domain of cGAS determines nucleo-cytoplasmic localization, enrichment on centromeres, and activation of nuclear-localized cGAS. These results reveal a preferential functional association of nuclear cGAS with centromeres.


Asunto(s)
Centrómero/enzimología , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Inmunidad Innata/genética , Nucleotidiltransferasas/metabolismo , Adulto , Animales , Línea Celular , Núcleo Celular/enzimología , ADN , ADN Satélite , Femenino , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Nucleotidiltransferasas/química , Dominios Proteicos
9.
Cell ; 175(2): 488-501.e22, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30270045

RESUMEN

Detection of viruses by innate immune sensors induces protective antiviral immunity. The viral DNA sensor cyclic GMP-AMP synthase (cGAS) is necessary for detection of HIV by human dendritic cells and macrophages. However, synthesis of HIV DNA during infection is not sufficient for immune activation. The capsid protein, which associates with viral DNA, has a pivotal role in enabling cGAS-mediated immune activation. We now find that NONO is an essential sensor of the HIV capsid in the nucleus. NONO protein directly binds capsid with higher affinity for weakly pathogenic HIV-2 than highly pathogenic HIV-1. Upon infection, NONO is essential for cGAS activation by HIV and cGAS association with HIV DNA in the nucleus. NONO recognizes a conserved region in HIV capsid with limited tolerance for escape mutations. Detection of nuclear viral capsid by NONO to promote DNA sensing by cGAS reveals an innate strategy to achieve distinction of viruses from self in the nucleus.


Asunto(s)
Proteínas de la Cápside/inmunología , Proteínas Asociadas a Matriz Nuclear/inmunología , Proteínas Asociadas a Matriz Nuclear/fisiología , Factores de Transcripción de Octámeros/inmunología , Factores de Transcripción de Octámeros/fisiología , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/fisiología , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/fisiología , Núcleo Celular/metabolismo , ADN Viral/genética , ADN Viral/inmunología , Proteínas de Unión al ADN , Células Dendríticas/inmunología , Infecciones por VIH/inmunología , VIH-1/genética , VIH-1/inmunología , VIH-2/genética , VIH-2/inmunología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata/inmunología , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/fisiología , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/inmunología
10.
Science ; 359(6372): 177-186, 2018 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-29326266

RESUMEN

After priming, naïve CD8+ T lymphocytes establish specific heritable transcription programs that define progression to long-lasting memory cells or to short-lived effector cells. Although lineage specification is critical for protection, it remains unclear how chromatin dynamics contributes to the control of gene expression programs. We explored the role of gene silencing by the histone methyltransferase Suv39h1. In murine CD8+ T cells activated after Listeria monocytogenes infection, Suv39h1-dependent trimethylation of histone H3 lysine 9 controls the expression of a set of stem cell-related memory genes. Single-cell RNA sequencing revealed a defect in silencing of stem/memory genes selectively in Suv39h1-defective T cell effectors. As a result, Suv39h1-defective CD8+ T cells show sustained survival and increased long-term memory reprogramming capacity. Thus, Suv39h1 plays a critical role in marking chromatin to silence stem/memory genes during CD8+ T effector terminal differentiation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/metabolismo , Memoria Inmunológica , Listeriosis/inmunología , Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Cromatina/metabolismo , Epigénesis Genética , Femenino , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Listeria monocytogenes/inmunología , Masculino , Metilación , Metiltransferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética
11.
Cell Cycle ; 13(10): 1530-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24626200

RESUMEN

CRABP1 (cellular retinoic acid binding protein 1) belongs to the family of fatty acid binding proteins. Retinoic acid binding is the only known functional activity of this protein. The role of CRABP1 in human carcinogenesis remains poorly understood. Here, for the first time we demonstrated pro-metastatic and pro-tumorigenic activity of CRABP1 in mesenchymal tumors. Further functional analysis revealed that the pro-tumorigenic effect of CRABP1 does not depend on retinoic acid binding activity. These results suggest that CRABP1 could have an alternative intracellular functional activity that contributes to the high malignancy of transformed mesenchymal cells. Microarray analysis detected CRABP1-mediated alterations in the expression of about 100 genes, including those encoding key regulatory proteins. CRABP1 is ubiquitously expressed in monophasic synovial sarcomas, while in biphasic synovial sarcomas it is expressed uniquely by the spindle cells of the aggressive mesenchymal component. High level of CRABP1 expression is associated with lymph node metastasis and poor differentiation/high grade of pancreatic neuroendocrine tumors (pNETs). Presented data suggest CRABP1 as a promising biomarker of pNETs' clinical behavior. Our results give the first evidence of pro-tumorigenic and pro-metastatic activity of CRABP1 in mesenchymal and neuroendocrine tumors.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Receptores de Ácido Retinoico/metabolismo , Sarcoma Sinovial/patología , Adulto , Anciano , Línea Celular Transformada , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Masculino , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Ácido Retinoico/genética , Sarcoma Sinovial/metabolismo , Tretinoina/metabolismo
12.
Nature ; 487(7406): 249-53, 2012 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-22763435

RESUMEN

During immune responses, naive CD4+ T cells differentiate into several T helper (TH) cell subsets under the control of lineage-specifying genes. These subsets (TH1, TH2 and TH17 cells and regulatory T cells) secrete distinct cytokines and are involved in protection against different types of infection. Epigenetic mechanisms are involved in the regulation of these developmental programs, and correlations have been drawn between the levels of particular epigenetic marks and the activity or silencing of specifying genes during differentiation. Nevertheless, the functional relevance of the epigenetic pathways involved in TH cell subset differentiation and commitment is still unclear. Here we explore the role of the SUV39H1­H3K9me3­HP1α silencing pathway in the control of TH2 lineage stability. This pathway involves the histone methylase SUV39H1, which participates in the trimethylation of histone H3 on lysine 9 (H3K9me3), a modification that provides binding sites for heterochromatin protein 1α (HP1α) and promotes transcriptional silencing. This pathway was initially associated with heterochromatin formation and maintenance but can also contribute to the regulation of euchromatic genes. We now propose that the SUV39H1­H3K9me3­HP1α pathway participates in maintaining the silencing of TH1 loci, ensuring TH2 lineage stability. In TH2 cells that are deficient in SUV39H1, the ratio between trimethylated and acetylated H3K9 is impaired, and the binding of HP1α at the promoters of silenced TH1 genes is reduced. Despite showing normal differentiation, both SUV39H1-deficient TH2 cells and HP1α-deficient TH2 cells, in contrast to wild-type cells, expressed TH1 genes when recultured under conditions that drive differentiation into TH1 cells. In a mouse model of TH2-driven allergic asthma, the chemical inhibition or loss of SUV39H1 skewed T-cell responses towards TH1 responses and decreased the lung pathology. These results establish a link between the SUV39H1­H3K9me3­HP1α pathway and the stability of TH2 cells, and they identify potential targets for therapeutic intervention in TH2-cell-mediated inflammatory diseases.


Asunto(s)
Epigénesis Genética , Células Th2/citología , Células Th2/inmunología , Animales , Asma/enzimología , Asma/inmunología , Asma/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Silenciador del Gen , Histonas/metabolismo , Masculino , Metiltransferasas/deficiencia , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Proteínas Represoras/deficiencia , Proteínas Represoras/metabolismo , Células TH1/metabolismo , Células Th2/enzimología
14.
Int J Cancer ; 128(4): 797-804, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20473891

RESUMEN

Metastasis, the capacity of tumour cells to disseminate and grow at distant sites, is the main factor in cancer mortality. Compounds inhibiting migration and invasion of cancer cells are promising candidates for anticancer therapy strategies. We have generated nuclease-resistant RNA ligands (aptamers) recognizing highly metastatic cells with high affinity and specificity, and inhibiting their migratory and invasive potentials. Aptamers were generated by a cell-based subtractive SELEX technology using isogenic cell lines with similar tumorigenic potentials but opposite metastatic aggressiveness. Two aptamers, E37 and E10, bound specifically to the metastatically aggressive cell line and altered the phosphorylation of several tyrosine kinases. Fluorescent microscopy showed intracellular uptake of E37, in contrast to membrane binding of E10. Both aptamers inhibited migration of tumour cells in culture (50 and 85% inhibition with respect to control pool for E10 and E37, respectively) while only E10 inhibited cell invasion (-75% with respect to control pool). This proof-of-concept study demonstrates the potential of cell-based SELEX to yield ligands that selectively recognize aggressive metastatic cells and inhibit phenotypes linked to metastatic potential.


Asunto(s)
Aptámeros de Nucleótidos/uso terapéutico , Neoplasias de la Mama/patología , Neoplasias de la Mama/prevención & control , Movimiento Celular , Técnica SELEX de Producción de Aptámeros , Animales , Western Blotting , Neoplasias de la Mama/genética , Adhesión Celular , Línea Celular Transformada , Cricetinae , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Mesocricetus , Ratones , Células 3T3 NIH , Invasividad Neoplásica , Fosforilación , Cicatrización de Heridas
15.
Clin Exp Metastasis ; 27(6): 409-18, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19904617

RESUMEN

We compared gene expression patterns related to cancer progression, cell adhesion and cytokine expression in v-Src-transformed isogenic cell lines with different levels of metastatic aggressiveness in syngenic animals: a high (HM) and a low metastatic (LM) cell lines, both predetermined by in vitro oncogenic transformation, and a high metastatic (NM) cell line resulting from in vivo selection of grafted LM cells. While LM and NM shared a common v-Src transforming variant different from that of HM, gene expression patterns were similar in HM and NM but different from that of LM. In this model, the induction of the metastatic phenotype by an oncogene variant or by the host environment led to highly similar expression profiles. If confirmed in other models, these results would be relevant for the diagnosis and treatment of metastasis.


Asunto(s)
Perfilación de la Expresión Génica , Metástasis de la Neoplasia/genética , Línea Celular Tumoral , Citocinas/genética , Genes src , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA