Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Nat Immunol ; 21(7): 777-789, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32572238

RESUMEN

T follicular helper (TFH) cells are a distinct type of CD4+ T cells that are essential for most antibody and B lymphocyte responses. TFH cell regulation and dysregulation is involved in a range of diseases. Bcl-6 is the lineage-defining transcription factor of TFH cells and its activity is essential for TFH cell differentiation and function. However, how Bcl-6 controls TFH biology has largely remained unclear, at least in part due to the intrinsic challenges of connecting repressors to gene upregulation in complex cell types with multiple possible differentiation fates. Multiple competing models were tested here by a series of experimental approaches to determine that Bcl-6 exhibits negative autoregulation and controls pleiotropic attributes of TFH differentiation and function, including migration, costimulation, inhibitory receptors and cytokines, via multiple repressor-of-repressor gene circuits.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Centro Germinal/inmunología , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Proteínas Represoras/genética , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Sistemas CRISPR-Cas/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Movimiento Celular/genética , Movimiento Celular/inmunología , Secuenciación de Inmunoprecipitación de Cromatina , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Redes Reguladoras de Genes , Centro Germinal/citología , Humanos , Masculino , Ratones , Mutación , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-bcl-6/genética , RNA-Seq , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
2.
Immunity ; 56(5): 959-978.e10, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37040762

RESUMEN

Although the importance of genome organization for transcriptional regulation of cell-fate decisions and function is clear, the changes in chromatin architecture and how these impact effector and memory CD8+ T cell differentiation remain unknown. Using Hi-C, we studied how genome configuration is integrated with CD8+ T cell differentiation during infection and investigated the role of CTCF, a key chromatin remodeler, in modulating CD8+ T cell fates through CTCF knockdown approaches and perturbation of specific CTCF-binding sites. We observed subset-specific changes in chromatin organization and CTCF binding and revealed that weak-affinity CTCF binding promotes terminal differentiation of CD8+ T cells through the regulation of transcriptional programs. Further, patients with de novo CTCF mutations had reduced expression of the terminal-effector genes in peripheral blood lymphocytes. Therefore, in addition to establishing genome architecture, CTCF regulates effector CD8+ T cell heterogeneity through altering interactions that regulate the transcription factor landscape and transcriptome.


Asunto(s)
Cromatina , Proteínas Represoras , Humanos , Sitios de Unión , Factor de Unión a CCCTC/metabolismo , Linfocitos T CD8-positivos/metabolismo , ADN/metabolismo , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
3.
Nat Immunol ; 18(5): 573-582, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28288100

RESUMEN

Dynamic changes in the expression of transcription factors (TFs) can influence the specification of distinct CD8+ T cell fates, but the observation of equivalent expression of TFs among differentially fated precursor cells suggests additional underlying mechanisms. Here we profiled the genome-wide histone modifications, open chromatin and gene expression of naive, terminal-effector, memory-precursor and memory CD8+ T cell populations induced during the in vivo response to bacterial infection. Integration of these data suggested that the expression and binding of TFs contributed to the establishment of subset-specific enhancers during differentiation. We developed a new bioinformatics method using the PageRank algorithm to reveal key TFs that influence the generation of effector and memory populations. The TFs YY1 and Nr3c1, both constitutively expressed during CD8+ T cell differentiation, regulated the formation of terminal-effector cell fates and memory-precursor cell fates, respectively. Our data define the epigenetic landscape of differentiation intermediates and facilitate the identification of TFs with previously unappreciated roles in CD8+ T cell differentiation.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Epigénesis Genética , Listeriosis/inmunología , Receptores de Glucocorticoides/metabolismo , Subgrupos de Linfocitos T/fisiología , Factor de Transcripción YY1/metabolismo , Animales , Linfocitos T CD8-positivos/microbiología , Diferenciación Celular/genética , Biología Computacional , Elementos de Facilitación Genéticos/genética , Perfilación de la Expresión Génica , Histonas/metabolismo , Memoria Inmunológica/genética , Ratones , Ratones Endogámicos C57BL , Receptores de Glucocorticoides/genética , Subgrupos de Linfocitos T/microbiología , Factor de Transcripción YY1/genética
4.
Immunity ; 48(4): 659-674.e6, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29669249

RESUMEN

T cell receptor (TCR) stimulation of naive CD8+ T cells initiates reprogramming of cis-regulatory landscapes that specify effector and memory cytotoxic T lymphocyte (CTL) differentiation. We mapped regions of hyper-accessible chromatin in naive cells during TCR stimulation and discovered that the transcription factor (TF) Runx3 promoted accessibility to memory CTL-specific cis-regulatory regions before the first cell division and was essential for memory CTL differentiation. Runx3 was specifically required for accessibility to regions highly enriched with IRF, bZIP and Prdm1-like TF motifs, upregulation of TFs Irf4 and Blimp1, and activation of fundamental CTL attributes in early effector and memory precursor cells. Runx3 ensured that nascent CTLs differentiated into memory CTLs by preventing high expression of the TF T-bet, slowing effector cell proliferation, and repressing terminal CTL differentiation. Runx3 overexpression enhanced memory CTL differentiation during iterative infections. Thus, Runx3 governs chromatin accessibility during TCR stimulation and enforces the memory CTL developmental program.


Asunto(s)
Cromatina/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Memoria Inmunológica/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Animales , Sitios de Unión/inmunología , Diferenciación Celular/inmunología , Línea Celular , Proliferación Celular , Chlorocebus aethiops , Cricetinae , Activación Enzimática/inmunología , Femenino , Humanos , Factores Reguladores del Interferón/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva/biosíntesis , Células Vero
5.
Nature ; 593(7857): 147-151, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33828301

RESUMEN

Bile acids are lipid-emulsifying metabolites synthesized in hepatocytes and maintained in vivo through enterohepatic circulation between the liver and small intestine1. As detergents, bile acids can cause toxicity and inflammation in enterohepatic tissues2. Nuclear receptors maintain bile acid homeostasis in hepatocytes and enterocytes3, but it is unclear how mucosal immune cells tolerate high concentrations of bile acids in the small intestine lamina propria (siLP). CD4+ T effector (Teff) cells upregulate expression of the xenobiotic transporter MDR1 (encoded by Abcb1a) in the siLP to prevent bile acid toxicity and suppress Crohn's disease-like small bowel inflammation4. Here we identify the nuclear xenobiotic receptor CAR (encoded by Nr1i3) as a regulator of MDR1 expression in T cells that can safeguard against bile acid toxicity and inflammation in the mouse small intestine. Activation of CAR induced large-scale transcriptional reprogramming in Teff cells that infiltrated the siLP, but not the colon. CAR induced the expression of not only detoxifying enzymes and transporters in siLP Teff cells, as in hepatocytes, but also the key anti-inflammatory cytokine IL-10. Accordingly, CAR deficiency in T cells exacerbated bile acid-driven ileitis in T cell-reconstituted Rag1-/- or Rag2-/- mice, whereas pharmacological activation of CAR suppressed it. These data suggest that CAR acts locally in T cells that infiltrate the small intestine to detoxify bile acids and resolve inflammation. Activation of this program offers an unexpected strategy to treat small bowel Crohn's disease and defines lymphocyte sub-specialization in the small intestine.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Regulación de la Expresión Génica , Intestino Delgado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Linfocitos T/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Receptor de Androstano Constitutivo , Enfermedad de Crohn/metabolismo , Femenino , Ileítis/metabolismo , Inflamación/metabolismo , Interleucina-10/biosíntesis , Interleucina-10/genética , Intestino Delgado/citología , Ratones
6.
Proc Natl Acad Sci U S A ; 120(1): e2217476120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36584296

RESUMEN

HIV gene expression is modulated by the combinatorial activity of the HIV transcriptional activator, Tat, host transcription factors, and chromatin remodeling complexes. To identify host factors regulating HIV transcription, we used specific single-guide RNAs and endonuclease-deficient Cas9 to perform chromatin affinity purification of the integrated HIV promoter followed by mass spectrometry. The scaffold protein, p32, also called ASF/SF2 splicing factor-associated protein, was identified among the top enriched factors present in actively transcribing HIV promoters but absent in silenced ones. Chromatin immunoprecipitation analysis confirmed the presence of p32 on active HIV promoters and its enhanced recruitment by Tat. HIV uses Tat to efficiently recruit positive transcription elongation factor b (p-TEFb) (CDK9/CCNT1) to TAR, an RNA secondary structure that forms from the first 59 bp of HIV transcripts, to enhance RNAPII transcriptional elongation. The RNA interference of p32 significantly reduced HIV transcription in primary CD4+T cells and in HIV chronically infected cells, independently of either HIV splicing or p32 anti-splicing activity. Conversely, overexpression of p32 specifically increased Tat-dependent HIV transcription. p32 was found to directly interact with Tat's basic domain enhancing Tat stability and half-life. Conversely, p32 associates with Tat via N- and C-terminal domains. Likely due its scaffold properties, p32 also promoted Tat association with TAR, p-TEFb, and RNAPII enhancing Tat-dependent HIV transcription. In sum, we identified p32 as a host factor that interacts with and stabilizes Tat protein, promotes Tat-dependent transcriptional regulation, and may be explored for HIV-targeted transcriptional inhibition.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , VIH-1/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Chaperonas Moleculares/metabolismo , Infecciones por VIH/genética , Transcripción Genética , Duplicado del Terminal Largo de VIH/genética
7.
J Immunol ; 210(11): 1752-1760, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37074193

RESUMEN

T follicular helper (TFH) cells are essential for developing protective Ab responses following vaccination. Greater understanding of the genetic program leading to TFH differentiation is needed. Chromatin modifications are central in the control of gene expression. However, detailed knowledge of how chromatin regulators (CRs) regulate differentiation of TFH cells is limited. We screened a large short hairpin RNA library targeting all known CRs in mice and identified the histone methyltransferase mixed lineage leukemia 1 (Mll1) as a positive regulator of TFH differentiation. Loss of Mll1 expression reduced formation of TFH cells following acute viral infection or protein immunization. In addition, expression of the TFH lineage-defining transcription factor Bcl6 was reduced in the absence of Mll1. Transcriptomics analysis identified Lef1 and Tcf7 as genes dependent on Mll1 for their expression, which provides one mechanism for the regulation of TFH differentiation by Mll1. Taken together, CRs such as Mll1 substantially influence TFH differentiation.


Asunto(s)
Cromatina , Células T Auxiliares Foliculares , Animales , Ratones , Diferenciación Celular , Cromatina/metabolismo , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Células T Auxiliares Foliculares/metabolismo , Linfocitos T Colaboradores-Inductores
8.
J Immunol ; 211(5): 721-726, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37486206

RESUMEN

CTL differentiation is controlled by the crosstalk of various transcription factors and epigenetic modulators. Uncovering this process is fundamental to improving immunotherapy and designing novel therapeutic approaches. In this study, we show that polycomb repressive complex 1 subunit chromobox (Cbx)4 favors effector CTL differentiation in a murine model. Cbx4 deficiency in CTLs induced a transcriptional signature of memory cells and increased the memory CTL population during acute viral infection. It has previously been shown that besides binding to H3K27me3 through its chromodomain, Cbx4 functions as a small ubiquitin-like modifier (SUMO) E3 ligase in a SUMO-interacting motifs (SIM)-dependent way. Overexpression of Cbx4 mutants in distinct domains showed that this protein regulates CTL differentiation primarily in an SIM-dependent way and partially through its chromodomain. Our data suggest a novel role of a polycomb group protein Cbx4 controlling CTL differentiation and indicated SUMOylation as a key molecular mechanism connected to chromatin modification in this process.


Asunto(s)
Complejo Represivo Polycomb 1 , Ubiquitina-Proteína Ligasas , Animales , Ratones , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
9.
Immunol Rev ; 300(1): 100-124, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33682165

RESUMEN

Adaptive immunity to intracellular pathogens and tumors is mediated by antigen-experienced CD8 T cells. Individual naive CD8 T cells have the potential to differentiate into a diverse array of antigen-experienced subsets that exhibit distinct effector functions, life spans, anatomic positioning, and potential for regenerating an entirely new immune response during iterative pathogenic exposures. The developmental process by which activated naive cells undergo diversification involves regulation of chromatin structure and transcription but is not entirely understood. This review examines how alterations in chromatin structure, transcription factor binding, extracellular signals, and single-cell gene expression explain the differential development of distinct effector (TEFF ) and memory (TMEM ) CD8 T cell subsets. Special emphasis is placed on how Runx proteins function with additional transcription factors to pioneer changes in chromatin accessibility and drive transcriptional programs that establish the core attributes of cytotoxic T lymphocytes, subdivide circulating and non-circulating TMEM cell subsets, and govern terminal differentiation. The discussion integrates the roles of specific cytokine signals, transcriptional circuits and how regulation of individual nucleosomes and RNA polymerase II activity can contribute to the process of differentiation. A model that integrates many of these features is discussed to conceptualize how activated CD8 T cells arrive at their fates.


Asunto(s)
Linfocitos T CD8-positivos , Subunidades alfa del Factor de Unión al Sitio Principal , Diferenciación Celular , Cromatina , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Memoria Inmunológica , Subgrupos de Linfocitos T
11.
Immunity ; 42(2): 265-278, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25680272

RESUMEN

During persistent antigen stimulation, CD8(+) T cells show a gradual decrease in effector function, referred to as exhaustion, which impairs responses in the setting of tumors and infections. Here we demonstrate that the transcription factor NFAT controls the program of T cell exhaustion. When expressed in cells, an engineered form of NFAT1 unable to interact with AP-1 transcription factors diminished T cell receptor (TCR) signaling, increased the expression of inhibitory cell surface receptors, and interfered with the ability of CD8(+) T cells to protect against Listeria infection and attenuate tumor growth in vivo. We defined the genomic regions occupied by endogenous and engineered NFAT1 in primary CD8(+) T cells and showed that genes directly induced by the engineered NFAT1 overlapped with genes expressed in exhausted CD8(+) T cells in vivo. Our data show that NFAT promotes T cell anergy and exhaustion by binding at sites that do not require cooperation with AP-1.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Anergia Clonal/genética , Factores de Transcripción NFATC/fisiología , Proteínas Recombinantes/farmacología , Factor de Transcripción AP-1/metabolismo , Animales , Células Cultivadas , Anergia Clonal/efectos de los fármacos , Regulación de la Expresión Génica/genética , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/microbiología , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Factores de Transcripción NFATC/genética , Neoplasias/inmunología , Regiones Promotoras Genéticas/genética , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes/genética
12.
J Immunol ; 209(8): 1566-1573, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36096645

RESUMEN

T follicular helper (TFH) cells are a specialized subset of CD4 T cells that deliver critical help signals to B cells for the production of high-affinity Abs. Understanding the genetic program regulating TFH differentiation is critical if one wants to manipulate TFH cells during vaccination. A large number of transcription factor (TFs) involved in the regulation of TFH differentiation have been characterized. However, there are likely additional unknown TFs required for this process. To identify new TFs, we screened a large short hairpin RNA library targeting 353 TFs in mice using an in vivo RNA interference screen. Yin Yang 1 (YY-1) was identified as a novel positive regulator of TFH differentiation. Ablation of YY-1 severely impaired TFH differentiation following acute viral infection and protein immunization. We found that the zinc fingers of YY-1 are critical to support TFH differentiation. Thus, we discovered a novel TF involved in the regulation of TFH cells.


Asunto(s)
Centro Germinal , Linfocitos T Colaboradores-Inductores , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Activación de Linfocitos , Ratones , ARN Interferente Pequeño/metabolismo , Células T Auxiliares Foliculares
14.
Immunity ; 41(2): 325-38, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25148027

RESUMEN

Classical genetic approaches to examine the requirements of genes for T cell differentiation during infection are time consuming. Here we developed a pooled approach to screen 30-100+ genes individually in separate antigen-specific T cells during infection using short hairpin RNAs in a microRNA context (shRNAmir). Independent screens using T cell receptor (TCR)-transgenic CD4(+) and CD8(+) T cells responding to lymphocytic choriomeningitis virus (LCMV) identified multiple genes that regulated development of follicular helper (Tfh) and T helper 1 (Th1) cells, and short-lived effector and memory precursor cytotoxic T lymphocytes (CTLs). Both screens revealed roles for the positive transcription elongation factor (P-TEFb) component Cyclin T1 (Ccnt1). Inhibiting expression of Cyclin T1, or its catalytic partner Cdk9, impaired development of Th1 cells and protective short-lived effector CTL and enhanced Tfh cell and memory precursor CTL formation in vivo. This pooled shRNA screening approach should have utility in numerous immunological studies.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Interferencia de ARN/inmunología , Animales , Diferenciación Celular/genética , Ciclina T/biosíntesis , Ciclina T/genética , Quinasa 9 Dependiente de la Ciclina/biosíntesis , Quinasa 9 Dependiente de la Ciclina/genética , Memoria Inmunológica/inmunología , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva , ARN Interferente Pequeño , Receptores de Antígenos de Linfocitos T/genética , Proteínas de Dominio T Box/genética , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Factores de Transcripción/genética , Transducción Genética/métodos
15.
J Immunol ; 206(9): 2170-2183, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33863789

RESUMEN

The transcriptional and epigenetic regulation of CD8+ T cell differentiation is critical for balancing pathogen eradication and long-term immunity by effector and memory CTLs, respectively. In this study, we demonstrate that the lysine demethylase 6b (Kdm6b) is essential for the proper generation and function of effector CD8+ T cells during acute infection and tumor eradication. We found that cells lacking Kdm6b (by either T cell-specific knockout mice or knockdown using short hairpin RNA strategies) show an enhanced generation of memory precursor and early effector cells upon acute viral infection in a cell-intrinsic manner. We also demonstrate that Kdm6b is indispensable for proper effector functions and tumor protection, and that memory CD8+ T cells lacking Kdm6b displayed a defective recall response. Mechanistically, we identified that Kdm6b, through induction of chromatin accessibility in key effector-associated gene loci, allows for the proper generation of effector CTLs. Our results pinpoint the essential function of Kdm6b in allowing chromatin accessibility in effector-associated genes, and identify Kdm6b as a potential target for therapeutics in diseases with dysregulated effector responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Cromatina/inmunología , Histona Demetilasas con Dominio de Jumonji/inmunología , Animales , Células Cultivadas , Cromatina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
16.
Nature ; 552(7684): 253-257, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29211713

RESUMEN

Tissue-resident memory CD8+ T (TRM) cells are found at common sites of pathogen exposure, where they elicit rapid and robust protective immune responses. However, the molecular signals that control TRM cell differentiation and homeostasis are not fully understood. Here we show that mouse TRM precursor cells represent a unique CD8+ T cell subset that is distinct from the precursors of circulating memory cell populations at the levels of gene expression and chromatin accessibility. Using computational and pooled in vivo RNA interference screens, we identify the transcription factor Runx3 as a key regulator of TRM cell differentiation and homeostasis. Runx3 was required to establish TRM cell populations in diverse tissue environments, and supported the expression of crucial tissue-residency genes while suppressing genes associated with tissue egress and recirculation. Furthermore, we show that human and mouse tumour-infiltrating lymphocytes share a core tissue-residency gene-expression signature with TRM cells that is associated with Runx3 activity. In a mouse model of adoptive T cell therapy for melanoma, Runx3-deficient CD8+ tumour-infiltrating lymphocytes failed to accumulate in tumours, resulting in greater rates of tumour growth and mortality. Conversely, overexpression of Runx3 enhanced tumour-specific CD8+ T cell abundance, delayed tumour growth, and prolonged survival. In addition to establishing Runx3 as a central regulator of TRM cell differentiation, these results provide insight into the signals that promote T cell residency in non-lymphoid sites, which could be used to enhance vaccine efficacy or adoptive cell therapy treatments that target cancer.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Memoria Inmunológica , Melanoma/inmunología , Especificidad de Órganos/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/citología , Diferenciación Celular , Proliferación Celular , Cromatina/genética , Cromatina/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Homeostasis , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Masculino , Melanoma/genética , Melanoma/patología , Melanoma/terapia , Ratones , Especificidad de Órganos/genética , Análisis de Supervivencia , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
17.
J Virol ; 95(4)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33239456

RESUMEN

HIV transcription requires assembly of cellular transcription factors at the HIV-1promoter. The TFIIH general transcription factor facilitates transcription initiation by opening the DNA strands around the transcription start site and phosphorylating the C-terminal domain for RNA polymerase II (RNAPII) for activation. Spironolactone (SP), an FDA approved aldosterone antagonist, triggers the proteasomal degradation of the XPB subunit of TFIIH, and concurrently suppresses acute HIV infection in vitro Here we investigated SP as a possible block-and-lock agent for a functional cure aimed at the transcriptional silencing of the viral reservoir. The long-term activity of SP was investigated in primary and cell line models of HIV-1 latency and reactivation. We show that SP rapidly inhibits HIV-1 transcription by reducing RNAPII recruitment to the HIV-1 genome. shRNA knockdown of XPB confirmed XPB degradation as the mechanism of action. Unfortunately, long-term pre-treatment with SP does not result in epigenetic suppression of HIV upon SP treatment interruption, since virus rapidly rebounds when XPB reemerges; however, SP alone without ART maintains the transcriptional suppression. Importantly, SP inhibits HIV reactivation from latency in both cell line models and resting CD4+T cells isolated from aviremic infected individuals upon cell stimulation with latency reversing agents. Furthermore, long-term treatment with concentrations of SP that potently degrade XPB does not lead to global dysregulation of cellular mRNA expression. Overall, these results suggest that XPB plays a key role in HIV transcriptional regulation and XPB degradation by SP strengthens the potential of HIV transcriptional inhibitors in block-and-lock HIV cure approaches.IMPORTANCE Antiretroviral therapy (ART) effectively reduces an individual's HIV loads to below the detection limit, nevertheless rapid viral rebound immediately ensues upon treatment interruption. Furthermore, virally suppressed individuals experience chronic immune activation from ongoing low-level virus expression. Thus, the importance of identifying novel therapeutics to explore in block-and-lock HIV functional cure approaches, aimed at the transcriptional and epigenetic silencing of the viral reservoir to block reactivation from latency. We investigated the potential of repurposing the FDA-approved spironolactone (SP), as one such drug. SP treatment rapidly degrades a host transcription factor subunit, XPB, inhibiting HIV transcription and blocking reactivation from latency. Long-term SP treatment does not affect cellular viability, cell cycle progression or global cellular transcription. SP alone blocks HIV transcription in the absence of ART but does not delay rebound upon drug removal as XPB rapidly reemerges. This study highlights XPB as a novel drug target in block-and-lock therapeutic approaches.

18.
Immunity ; 32(1): 79-90, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-20096607

RESUMEN

Interleukin(IL)-2 and inflammation regulate effector and memory cytolytic T-lymphocyte (CTL) generation during infection. We demonstrate a complex interplay between IL-2 and inflammatory signals during CTL differentiation. IL-2 stimulation induced the transcription factor eomesodermin (Eomes), upregulated perforin (Prf1) transcription, and repressed re-expression of memory CTL markers Bcl6 and IL-7Ralpha. Binding of Eomes and STAT5 to Prf1 cis-regulatory regions correlated with transcriptional initiation (increased recruitment of RNA polymerase II to the Prf1 promoter). Inflammation (CpG, IL-12) enhanced expression of IL-2Ralpha and the transcription factor T-bet, but countered late Eomes and perforin induction while preventing IL-7Ralpha repression by IL-2. After infection of mice with lymphocytic choriomeningitis virus, IL-2Ralpha-deficient effector CD8(+) T cells expressed more Bcl6 but less perforin and granzyme B, formed fewer KLRG-1(+) and T-bet-expressing CTL, and killed poorly. Thus, inflammation influences both effector and memory CTL differentiation, whereas persistent IL-2 stimulation promotes effector at the expense of memory CTL development.


Asunto(s)
Diferenciación Celular/inmunología , Regulación de la Expresión Génica/inmunología , Inflamación/inmunología , Interleucina-2/inmunología , Subgrupos de Linfocitos T/citología , Linfocitos T Citotóxicos/citología , Animales , Diferenciación Celular/genética , Separación Celular , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/inmunología , Citometría de Flujo , Expresión Génica , Memoria Inmunológica/genética , Memoria Inmunológica/inmunología , Inmunoprecipitación , Inflamación/metabolismo , Interleucina-2/genética , Interleucina-2/metabolismo , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Transcripción Genética
20.
Trends Immunol ; 36(5): 315-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25937561

RESUMEN

Functional genomics approaches that leverage the RNAi pathway have been applied in vivo to examine the roles of hundreds or thousands of genes; mainly in the context of cancer. Here, we discuss principles guiding the design of RNAi screens, parameters that determine success and recent developments that have improved accuracy and expanded the applicability of these approaches to other in vivo settings, including the immune system. We review recent studies that have applied in vivo RNAi screens in T cells to examine genes that regulate T cell differentiation during viral infection, and that control their accumulation in tumors in a model of adoptive T cell therapy. In this context, we put forward an argument as to why RNAi approaches in vivo are likely to provide particularly salient insight into immunology.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Estudios de Asociación Genética , Interferencia de ARN , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Virosis/genética , Virosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA