Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1146699, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37275914

RESUMEN

Introduction: Transcriptional activation depends on the interplay of chromatin modifiers to establish a permissive epigenetic landscape. While histone 3 lysine 9 (H3K9) methylation has long been associated with gene repression, there is limited evidence to support a role for H3K9 demethylases in gene activation. Methods: We leveraged knockdown and overexpression of JMJD2d / Kdm4d in mouse embryonic fibroblasts, coupled with extensive epigenomic analysesm to decipher the role of histone 3 lysine 9 demethylases in the innate immune response. Results: Here we describe the H3K9 demethylase Kdm4d/JMJD2d as a positive regulator of type I interferon responses. In mouse embryonic fibroblasts (MEFs), depletion of JMJD2d attenuates the transcriptional response, conferring increased viral susceptibility, while overexpression of the demethylase results in more robust IFN activation. We find that the underlying mechanism of JMJD2d in type I interferon responses consists of an effect both on the transcription of enhancer RNAs (eRNAs) and on dynamic H3K9me2 at associated promoters. In support of these findings, we establish that JMJD2d is associated with enhancer regions throughout the genome prior to stimulation but is redistributed to inducible promoters in conjunction with transcriptional activation. Discussion: Taken together, our data reveal JMJD2d as a chromatin modifier that connects enhancer transcription with promoter demethylation to modulate transcriptional responses.


Asunto(s)
Histona Demetilasas , Interferón Tipo I , Animales , Ratones , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histonas/genética , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Interferón Tipo I/genética , Lisina/genética , Fibroblastos/metabolismo , Cromatina/genética
2.
FEBS Lett ; 594(20): 3324-3337, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32862441

RESUMEN

Humoral immunity in mammals relies on the function of two developmentally and functionally distinct B-cell subsets-B1 and B2 cells. While B2 cells are responsible for the adaptive response to environmental antigens, B1 cells regulate the production of polyreactive and low-affinity antibodies for innate humoral immunity. The molecular mechanism of B-cell specification into different subsets is understudied. In this study, we identified lysine methyltransferase NSD2 (MMSET/WHSC1) as a critical regulator of B1 cell development. In contrast to its minor impact on B2 cells, deletion of the catalytic domain of NSD2 in primary B cells impairs the generation of B1 lineage. Thus, NSD2, a histone H3 K36 dimethylase, is the first-in-class epigenetic regulator of a B-cell lineage in mice.


Asunto(s)
Linfocitos B/metabolismo , Dominio Catalítico , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , Animales Recién Nacidos , Centro Germinal/metabolismo , Histonas/metabolismo , Inmunidad Humoral , Cambio de Clase de Inmunoglobulina , Lisina/metabolismo , Metilación , Ratones Endogámicos C57BL , Relación Estructura-Actividad , Análisis de Supervivencia
3.
Blood ; 133(4): 331-343, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30429161

RESUMEN

Conditional knockout (KO) mouse models are invaluable for elucidating the physiological roles of platelets. The Platelet factor 4-Cre recombinase (Pf4-Cre) transgenic mouse is the current model of choice for generating megakaryocyte/platelet-specific KO mice. Platelets and leukocytes work closely together in a wide range of disease settings, yet the specific contribution of platelets to these processes remains unclear. This is partially a result of the Pf4-Cre transgene being expressed in a variety of leukocyte populations. To overcome this issue, we developed a Gp1ba-Cre transgenic mouse strain in which Cre expression is driven by the endogenous Gp1ba locus. By crossing Gp1ba-Cre and Pf4-Cre mice to the mT/mG dual-fluorescence reporter mouse and performing a head-to-head comparison, we demonstrate more stringent megakaryocyte lineage-specific expression of the Gp1ba-Cre transgene. Broader tissue expression was observed with the Pf4-Cre transgene, leading to recombination in many hematopoietic lineages, including monocytes, macrophages, granulocytes, and dendritic and B and T cells. Direct comparison of phenotypes of Csk, Shp1, or CD148 conditional KO mice generated using either the Gp1ba-Cre or Pf4-Cre strains revealed similar platelet phenotypes. However, additional inflammatory and immunological anomalies were observed in Pf4-Cre-generated KO mice as a result of nonspecific deletion in other hematopoietic lineages. By excluding leukocyte contributions to phenotypes, the Gp1ba-Cre mouse will advance our understanding of the role of platelets in inflammation and other pathophysiological processes in which platelet-leukocyte interactions are involved.


Asunto(s)
Plaquetas/metabolismo , Integrasas/metabolismo , Leucocitos/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Aglutinación , Animales , Células de la Médula Ósea/citología , Proteína Tirosina Quinasa CSK , Linaje de la Célula , Tamaño de la Célula , Marcación de Gen , Homeostasis , Recuento de Linfocitos , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Fenotipo , Agregación Plaquetaria , Factor Plaquetario 4/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Recombinación Genética/genética , Bazo/citología , Familia-src Quinasas/metabolismo
4.
Nat Struct Mol Biol ; 25(9): 885-893, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30177761

RESUMEN

Viral infection perturbs host cells and can be used to uncover regulatory mechanisms controlling cellular responses and susceptibility to infections. Using cell biological, biochemical, and genetic tools, we reveal that influenza A virus (IAV) infection induces global transcriptional defects at the 3' ends of active host genes and RNA polymerase II (RNAPII) run-through into extragenic regions. Deregulated RNAPII leads to expression of aberrant RNAs (3' extensions and host-gene fusions) that ultimately cause global transcriptional downregulation of physiological transcripts, an effect influencing antiviral response and virulence. This phenomenon occurs with multiple strains of IAV, is dependent on influenza NS1 protein, and can be modulated by SUMOylation of an intrinsically disordered region (IDR) of NS1 expressed by the 1918 pandemic IAV strain. Our data identify a strategy used by IAV to suppress host gene expression and indicate that polymorphisms in IDRs of viral proteins can affect the outcome of an infection.


Asunto(s)
Gripe Humana/genética , ARN Polimerasa II/genética , Regiones Terminadoras Genéticas/genética , Humanos , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/fisiología , Virulencia
5.
J Exp Med ; 215(7): 1777-1787, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29934321

RESUMEN

Humans carry trillions of viruses that thrive because of their ability to exploit the host. In this exploitation, viruses promote their own replication by suppressing the host antiviral response and by inducing changes in host biosynthetic processes, often with extremely small genomes of their own. In the review, we discuss the phenomenon of histone mimicry by viral proteins and how this mimicry allows the virus to dial in to the cell's transcriptional processes and establish a cell state that promotes infection. We suggest that histone mimicry is part of a broader viral strategy to use intrinsic protein disorder as a means to overcome the size limitations of its own genome and to maximize its impact on host protein networks. In particular, we discuss how intrinsic protein disorder may enable viral proteins to interfere with phase-separated host protein condensates, including those that contribute to chromatin-mediated control of gene expression.


Asunto(s)
Histonas/metabolismo , Virus/metabolismo , Secuencia de Aminoácidos , Animales , Histonas/química , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Virosis/virología , Replicación Viral
6.
J Exp Med ; 215(4): 1101-1113, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29523590

RESUMEN

Differentiation and activation of T cells require the activity of numerous histone lysine methyltransferases (HMT) that control the transcriptional T cell output. One of the most potent regulators of T cell differentiation is the HMT Ezh2. Ezh2 is a key enzymatic component of polycomb repressive complex 2 (PRC2), which silences gene expression by histone H3 di/tri-methylation at lysine 27. Surprisingly, in many cell types, including T cells, Ezh2 is localized in both the nucleus and the cytosol. Here we show the presence of a nuclear-like PRC2 complex in T cell cytosol and demonstrate a role of cytosolic PRC2 in T cell antigen receptor (TCR)-mediated signaling. We show that short-term suppression of PRC2 precludes TCR-driven T cell activation in vitro. We also demonstrate that pharmacological inhibition of PRC2 in vivo greatly attenuates the severe T cell-driven autoimmunity caused by regulatory T cell depletion. Our data reveal cytoplasmic PRC2 is one of the most potent regulators of T cell activation and point toward the therapeutic potential of PRC2 inhibitors for the treatment of T cell-driven autoimmune diseases.


Asunto(s)
Complejo Represivo Polycomb 2/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Animales , Autoinmunidad , Proliferación Celular , Citoplasma/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Interleucina-2/genética , Subgrupos Linfocitarios/inmunología , Lisina/metabolismo , Metilación , Ratones , Fosforilación , Receptores de Interleucina-2/genética , Transcripción Genética
7.
Blood ; 131(10): 1122-1144, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29301754

RESUMEN

Src family kinases (SFKs) coordinate the initiating and propagating activation signals in platelets, but it remains unclear how they are regulated. Here, we show that ablation of C-terminal Src kinase (Csk) and receptor-like protein tyrosine-phosphatase CD148 in mice results in a dramatic increase in platelet SFK activity, demonstrating that these proteins are essential regulators of platelet reactivity. Paradoxically, Csk/CD148-deficient mice exhibit reduced in vivo and ex vivo thrombus formation and increased bleeding following injury rather than a prothrombotic phenotype. This is a consequence of multiple negative feedback mechanisms, including downregulation of the immunoreceptor tyrosine-based activation motif (ITAM)- and hemi-ITAM-containing receptors glycoprotein VI (GPVI)-Fc receptor (FcR) γ-chain and CLEC-2, respectively and upregulation of the immunoreceptor tyrosine-based inhibition motif (ITIM)-containing receptor G6b-B and its interaction with the tyrosine phosphatases Shp1 and Shp2. Results from an analog-sensitive Csk mouse model demonstrate the unconventional role of SFKs in activating ITIM signaling. This study establishes Csk and CD148 as critical molecular switches controlling the thrombotic and hemostatic capacity of platelets and reveals cell-intrinsic mechanisms that prevent pathological thrombosis from occurring.


Asunto(s)
Plaquetas/metabolismo , Homeostasis , Trombosis/metabolismo , Familia-src Quinasas/metabolismo , Secuencias de Aminoácidos , Animales , Plaquetas/patología , Proteína Tirosina Quinasa CSK , Ratones , Ratones Noqueados , Glicoproteínas de Membrana Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Trombosis/genética , Familia-src Quinasas/genética
8.
Genes (Basel) ; 8(8)2017 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-28825667

RESUMEN

Viral infection leads to a robust cellular response whereby the infected cell produces hundreds of molecular regulators to combat infection. Currently, non-canonical components, e.g., long noncoding RNAs (lncRNAs) have been added to the repertoire of immune regulators involved in the antiviral program. Interestingly, studies utilizing next-generation sequencing technologies show that a subset of the >10,000 lncRNAs in the mammalian genome contain small open reading frames (smORFs) associated with active translation, i.e., many lncRNAs are not noncoding. Here, we use genome-wide high-throughput methods to identify potential micropeptides in smORF-containing lncRNAs involved in the immune response. Using influenza as a viral infection model, we performed RNA-seq and ribosome profiling to track expression and translation of putative lncRNAs that may encode for peptides and identify tens of potential candidates. Interestingly, many of these peptides are highly conserved at the protein level, strongly suggesting biological relevance and activity. By perusing publicly available data sets, four potential peptides of interest seem common to stress induction and/or are highly conserved; potential peptides from the MMP24-AS1, ZFAS1, RP11-622K12.1, and MIR22HG genes. Interestingly, using an antibody against the potential peptide encoded by MIR22HG RNA, we show that the peptide is stably expressed in the absence of infection, and upregulated in response to infection, corroborating the prediction of the ribosome profiling results. These data show the utility of perturbation approaches in identifying potentially relevant novel molecules encoded in the genome.

9.
Nat Rev Drug Discov ; 15(12): 835-853, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27765940

RESUMEN

Immune-mediated diseases are clinically heterogeneous but they share genetic and pathogenic mechanisms. These diseases may develop from the interplay of genetic factors and environmental or lifestyle factors. Exposure to such factors, including infectious agents, is associated with coordinated changes in gene transcription owing to epigenetic alterations. A growing understanding of how epigenetic mechanisms control gene expression patterns and cell function has been aided by the development of small-molecule inhibitors that target these processes. These chemical tools have helped to reveal the importance of epigenetics in guiding cell fate decisions during immune responses and have also highlighted the potential for targeting epigenetic mechanisms for the treatment of inflammation and immune-mediated diseases. In this Review, we discuss the most advanced areas of epigenetic drug development for autoimmune and inflammatory diseases and summarize the promising preclinical data in this exciting and evolving field. These agents will inevitably begin to move into clinical trials for use in patients with immune-mediated diseases.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Descubrimiento de Drogas , Epigénesis Genética , Inflamación/tratamiento farmacológico , Animales , Humanos
10.
Nat Neurosci ; 19(10): 1321-30, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27526204

RESUMEN

Normal brain function depends on the interaction between highly specialized neurons that operate within anatomically and functionally distinct brain regions. Neuronal specification is driven by transcriptional programs that are established during early neuronal development and remain in place in the adult brain. The fidelity of neuronal specification depends on the robustness of the transcriptional program that supports the neuron type-specific gene expression patterns. Here we show that polycomb repressive complex 2 (PRC2), which supports neuron specification during differentiation, contributes to the suppression of a transcriptional program that is detrimental to adult neuron function and survival. We show that PRC2 deficiency in striatal neurons leads to the de-repression of selected, predominantly bivalent PRC2 target genes that are dominated by self-regulating transcription factors normally suppressed in these neurons. The transcriptional changes in PRC2-deficient neurons lead to progressive and fatal neurodegeneration in mice. Our results point to a key role of PRC2 in protecting neurons against degeneration.


Asunto(s)
Silenciador del Gen , Degeneración Nerviosa/genética , Complejo Represivo Polycomb 2/metabolismo , Animales , Muerte Celular/genética , Supervivencia Celular/genética , Regulación hacia Abajo , Femenino , N-Metiltransferasa de Histona-Lisina/metabolismo , Masculino , Ratones , Ratones Noqueados , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Complejo Represivo Polycomb 2/deficiencia , Complejo Represivo Polycomb 2/genética
11.
Nat Immunol ; 17(3): 331-43, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26779602

RESUMEN

The transcription factor Blimp-1 is necessary for the generation of plasma cells. Here we studied its functions in plasmablast differentiation by identifying regulated Blimp-1 target genes. Blimp-1 promoted the migration and adhesion of plasmablasts. It directly repressed genes encoding several transcription factors and Aicda (which encodes the cytidine deaminase AID) and thus silenced B cell-specific gene expression, antigen presentation and class-switch recombination in plasmablasts. It directly activated genes, which led to increased expression of the plasma cell regulator IRF4 and proteins involved in immunoglobulin secretion. Blimp-1 induced the transcription of immunoglobulin genes by controlling the 3' enhancers of the loci encoding the immunoglobulin heavy chain (Igh) and κ-light chain (Igk) and, furthermore, regulated the post-transcriptional expression switch from the membrane-bound form of the immunoglobulin heavy chain to its secreted form by activating Ell2 (which encodes the transcription-elongation factor ELL2). Notably, Blimp-1 recruited chromatin-remodeling and histone-modifying complexes to regulate its target genes. Hence, many essential functions of plasma cells are under the control of Blimp-1.


Asunto(s)
Diferenciación Celular/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Cadenas kappa de Inmunoglobulina/inmunología , Factores Reguladores del Interferón/inmunología , Células Plasmáticas/inmunología , Factores de Transcripción/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Adhesión Celular/genética , Adhesión Celular/inmunología , Diferenciación Celular/genética , Ensayos de Migración de Leucocitos , Movimiento Celular/genética , Movimiento Celular/inmunología , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Regulación de la Expresión Génica , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/genética , Factores Reguladores del Interferón/genética , Espectrometría de Masas , Ratones , Células Plasmáticas/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN , Factores de Transcripción/genética
12.
J Exp Med ; 212(11): 1771-81, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26392221

RESUMEN

Studies investigating the causes of autism spectrum disorder (ASD) point to genetic, as well as epigenetic, mechanisms of the disease. Identification of epigenetic processes that contribute to ASD development and progression is of major importance and may lead to the development of novel therapeutic strategies. Here, we identify the bromodomain and extraterminal domain-containing proteins (BETs) as epigenetic regulators of genes involved in ASD-like behaviors in mice. We found that the pharmacological suppression of BET proteins in the brain of young mice, by the novel, highly specific, brain-permeable inhibitor I-BET858 leads to selective suppression of neuronal gene expression followed by the development of an autism-like syndrome. Many of the I-BET858-affected genes have been linked to ASD in humans, thus suggesting the key role of the BET-controlled gene network in the disorder. Our studies suggest that environmental factors controlling BET proteins or their target genes may contribute to the epigenetic mechanism of ASD.


Asunto(s)
Trastorno del Espectro Autista/etiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Trastorno del Espectro Autista/genética , Factor Neurotrófico Derivado del Encéfalo/farmacología , Epigénesis Genética , Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Proc Natl Acad Sci U S A ; 112(32): E4448-57, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26216992

RESUMEN

Aire controls immunologic tolerance by inducing a battery of thymic transcripts encoding proteins characteristic of peripheral tissues. Its unusually broad effect is achieved by releasing RNA polymerase II paused just downstream of transcriptional start sites. We explored Aire's collaboration with the bromodomain-containing protein, Brd4, uncovering an astonishing correspondence between those genes induced by Aire and those inhibited by a small-molecule bromodomain blocker. Aire:Brd4 binding depended on an orchestrated series of posttranslational modifications within Aire's caspase activation and recruitment domain. This interaction attracted P-TEFb, thereby mobilizing downstream transcriptional elongation and splicing machineries. Aire:Brd4 association was critical for tolerance induction, and its disruption could account for certain point mutations that provoke human autoimmune disease. Our findings evoke the possibility of unanticipated immunologic mechanisms subtending the potent antitumor effects of bromodomain blockers.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Timo/citología , Elongación de la Transcripción Genética , Factores de Transcripción/metabolismo , Acetilación/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/genética , Lisina/metabolismo , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/química , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Elongación de la Transcripción Genética/efectos de los fármacos , Factores de Transcripción/química , Factores de Transcripción/genética , Transcriptoma/genética , Proteína AIRE
14.
J Exp Med ; 212(3): 297-306, 2015 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-25687282

RESUMEN

The fidelity of T cell immunity depends greatly on coupling T cell receptor signaling with specific T cell effector functions. Here, we describe a chromatin-based mechanism that enables integration of TCR specificity into definite T cell lineage commitment. Using natural killer T cells (iNKT cell) as a model of a T cell subset that differentiates in response to specific TCR signaling, we identified a key role of histone H3 lysine 27 trimethylation (H3K27me3) in coupling iNKT cell TCR specificity with the generation of iNKT cells. We found that the Zbtb16/PLZF gene promoter that drives iNKT cell differentiation possesses a bivalent chromatin state characterized by the simultaneous presence of negative and positive H3K27me3 and H3K4me3 modifications. Depletion of H3K27me3 at the Zbtb16/PLZF promoter leads to uncoupling of iNKT cell development from TCR specificity and is associated with accumulation of iNKT-like CD4(+) cells that express a non-iNKT cell specific T cell repertoire. In turn, stabilization of H3K27me3 leads to a drastic reduction of the iNKT cell population. Our data suggest that H3K27me3 levels at the bivalent Zbtb16/PLZF gene define a threshold enabling precise coupling of TCR specificity to lineage commitment.


Asunto(s)
Histonas/metabolismo , Células T Asesinas Naturales/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T , Animales , Antígenos CD4/metabolismo , Linfocitos T CD8-positivos/fisiología , Cromatina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Lisina/metabolismo , Metilación , Ratones Endogámicos C57BL , Ratones Transgénicos , Células T Asesinas Naturales/metabolismo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Receptores de Antígenos de Linfocitos T/metabolismo
15.
Cell ; 158(5): 989-999, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25131989

RESUMEN

Latent reservoirs of HIV-1-infected cells are refractory to antiretroviral therapies (ART) and remain the major barrier to curing HIV-1. Because latently infected cells are long-lived, immunologically invisible, and may undergo homeostatic proliferation, a "shock and kill" approach has been proposed to eradicate this reservoir by combining ART with inducers of viral transcription. However, all attempts to alter the HIV-1 reservoir in vivo have failed to date. Using humanized mice, we show that broadly neutralizing antibodies (bNAbs) can interfere with establishment of a silent reservoir by Fc-FcR-mediated mechanisms. In established infection, bNAbs or bNAbs plus single inducers are ineffective in preventing viral rebound. However, bNAbs plus a combination of inducers that act by independent mechanisms synergize to decrease the reservoir as measured by viral rebound. Thus, combinations of inducers and bNAbs constitute a therapeutic strategy that impacts the establishment and maintenance of the HIV-1 reservoir in humanized mice.


Asunto(s)
Anticuerpos Neutralizantes/administración & dosificación , Infecciones por VIH/inmunología , VIH-1/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Latencia del Virus/efectos de los fármacos , Animales , Fármacos Anti-VIH/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígeno CTLA-4/administración & dosificación , Infecciones por VIH/virología , VIH-1/genética , VIH-1/fisiología , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Humanos , Ácidos Hidroxámicos/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/inmunología , Ratones , Receptores Fc/inmunología , Vorinostat
16.
PLoS One ; 9(7): e101749, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24992693

RESUMEN

Argonaute 2 (Ago2) is the only mammalian Ago protein capable of mRNA cleavage. It has been reported that the activity of the short interfering RNA targeting coding sequence (CDS), but not 3' untranslated region (3'UTR) of an mRNA, is solely dependent on Ago2 in vitro. These studies utilized extremely high doses of siRNAs and overexpressed Ago proteins, as well as were directed at various highly expressed reporter transgenes. Here we report the effect of Ago2 in vivo on targeted knockdown of several endogenous genes by siRNAs, targeting both CDS and 3'UTR. We show that siRNAs targeting CDS lose their activity in the absence of Ago2, whereas both Ago1 and Ago3 proteins contribute to residual 3'UTR-targeted siRNA-mediated knockdown observed in the absence of Ago2 in mouse liver. Our results provide mechanistic insight into two components mediating RNAi under physiological conditions: mRNA cleavage dependent and independent. In addition our results contribute a novel consideration for designing most efficacious siRNA molecules with the preference given to 3'UTR targeting as to harness the activity of several Ago proteins.


Asunto(s)
Proteínas Argonautas/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Fibroblastos/citología , Hígado/metabolismo , ARN Mensajero/genética , Animales , Proteínas Argonautas/genética , Células Cultivadas , Embrión de Mamíferos/citología , Femenino , Fibroblastos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células 3T3 NIH , Interferencia de ARN
17.
Artículo en Inglés | MEDLINE | ID: mdl-24890513

RESUMEN

Immunity relies on the heterogeneity of immune cells and their ability to respond to pathogen challenges. In the adaptive immune system, lymphocytes display a highly diverse antigen receptor repertoire that matches the vast diversity of pathogens. In the innate immune system, the cell's heterogeneity and phenotypic plasticity enable flexible responses to changes in tissue homeostasis caused by infection or damage. The immune responses are calibrated by the graded activity of immune cells that can vary from yeast-like proliferation to lifetime dormancy. This article describes key epigenetic processes that contribute to the function of immune cells during health and disease.


Asunto(s)
Epigénesis Genética , Inmunidad/genética , Inmunidad Adaptativa , Linaje de la Célula , Humanos , Linfocitos/citología , Linfocitos/inmunología
18.
Annu Rev Immunol ; 32: 489-511, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24555473

RESUMEN

A fundamental property of cells of the innate immune system is their ability to elicit a transcriptional response to a microbial stimulus or danger signal with a high degree of cell type and stimulus specificity. The selective response activates effector pathways to control the insult and plays a central role in regulating adaptive immunity through the differential regulation of cytokine genes. Selectivity is dictated by signaling pathways and their transcription factor targets. However, a growing body of evidence supports models in which different subsets of genes exhibit distinct chromatin features that play active roles in shaping the response. Chromatin also participates in innate memory mechanisms that can promote tolerance to a stimulus or prime cells for a more robust response. These findings have generated interest in the capacity to modulate chromatin regulators with small-molecule compounds for the treatment of diseases associated with innate or adaptive immunity.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Inmunidad Innata/fisiología , Animales , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Memoria Inmunológica/genética , Memoria Inmunológica/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/terapia , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Transcripción Genética
19.
Genes Dev ; 27(16): 1731-8, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23964091

RESUMEN

Recent advances in the enzymology of transcription and chromatin regulation have led to the discovery of proteins that play a prominent role in cell differentiation and the maintenance of specialized cell functions. Knowledge about post-synthetic DNA and histone modifications as well as information about the rules that guide the formation of multimolecular chromatin-bound complexes have helped to delineate gene-regulating pathways and describe how these pathways are altered in various pathological conditions. The present review focuses on the emerging area of therapeutic interference with chromatin function for the purpose of cancer treatment and immunomodulation.


Asunto(s)
Antineoplásicos/uso terapéutico , Cromatina/efectos de los fármacos , Inmunidad , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Humanos , Inmunidad/efectos de los fármacos , Inmunidad/genética , Inmunomodulación/efectos de los fármacos , Complejo Represivo Polycomb 2/metabolismo , Factores de Transcripción/metabolismo
20.
Adv Immunol ; 119: 107-33, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23886066

RESUMEN

Immune response to pathogens depends on coordinated regulation of numerous genes that contribute collectively to pathogen elimination and restoration of the integrity of the affected tissue. The pathogen-induced gene expression is governed largely by the signal-induced posttranslational histone modifications that facilitate assembly of the functionally distinct chromatin complexes. In this review, we describe the principles of chromatin-based gene regulation during innate immune responses. We discuss the ability of pathogens to hijack the host response by interfering with various arms of transcriptional machinery involved in the responses. In particular, we discuss the phenomenon of the histone mimicry where interaction between histones and transcriptional regulators is targeted by pathogens that carry the histone-like sequences (histone mimics). We show how the principle of isotone mimicry as an efficient way to control host gene expression has been sued for the development of novel anti-inflammatory pharmacological approaches.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Evasión Inmune/inmunología , Inflamación/genética , Inflamación/inmunología , Empalme del ARN , Cromatina/metabolismo , Regulación de la Expresión Génica , Histonas/genética , Histonas/inmunología , Humanos , Inmunidad Innata , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...