Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nat Immunol ; 24(1): 42-54, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36050414

RESUMEN

Innate lymphoid cells (ILCs) are well-characterized immune cells that play key roles in host defense and tissue homeostasis. Yet, how the three-dimensional (3D) genome organization underlies the development and functions of ILCs is unknown. Herein, we carried out an integrative analysis of the 3D genome structure, chromatin accessibility and gene expression in mature ILCs. Our results revealed that the local 3D configuration of the genome is rewired specifically at loci associated with ILC biology to promote their development and functional differentiation. Importantly, we demonstrated that the ontogenesis of ILC2s and the progression of allergic airway inflammation are determined by a unique local 3D configuration of the region containing the ILC-lineage-defining factor Id2, which is characterized by multiple interactions between the Id2 promoter and distal regulatory elements bound by the transcription factors GATA-3 and RORα, unveiling the mechanism whereby the Id2 expression is specifically controlled in group 2 ILCs.


Asunto(s)
Inmunidad Innata , Linfocitos , Humanos , Inflamación/genética , Inflamación/metabolismo , Linaje de la Célula , Regiones Promotoras Genéticas
2.
Immunity ; 56(7): 1451-1467.e12, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37263273

RESUMEN

Multi-enhancer hubs are spatial clusters of enhancers present across numerous developmental programs. Here, we studied the functional relevance of these three-dimensional structures in T cell biology. Mathematical modeling identified a highly connected multi-enhancer hub at the Ets1 locus, comprising a noncoding regulatory element that was a hotspot for sequence variation associated with allergic disease in humans. Deletion of this regulatory element in mice revealed that the multi-enhancer connectivity was dispensable for T cell development but required for CD4+ T helper 1 (Th1) differentiation. These mice were protected from Th1-mediated colitis but exhibited overt allergic responses. Mechanistically, the multi-enhancer hub controlled the dosage of Ets1 that was required for CTCF recruitment and assembly of Th1-specific genome topology. Our findings establish a paradigm wherein multi-enhancer hubs control cellular competence to respond to an inductive cue through quantitative control of gene dosage and provide insight into how sequence variation within noncoding elements at the Ets1 locus predisposes individuals to allergic responses.


Asunto(s)
Hipersensibilidad , Linfocitos T , Humanos , Ratones , Animales , Diferenciación Celular/genética , Hematopoyesis , Inflamación/genética , Secuencias Reguladoras de Ácidos Nucleicos , Hipersensibilidad/genética , Elementos de Facilitación Genéticos/genética
3.
Immunity ; 43(6): 1087-100, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26682983

RESUMEN

The initiation of cytotoxic immune responses by dendritic cells (DCs) requires the presentation of antigenic peptides derived from phagocytosed microbes and infected or dead cells to CD8(+) T cells, a process called cross-presentation. Antigen cross-presentation by non-activated DCs, however, is not sufficient for the effective induction of immune responses. Additionally, DCs need to be activated through innate receptors, like Toll-like receptors (TLRs). During DC maturation, cross-presentation efficiency is first upregulated and then turned off. Here we show that during this transient phase of enhanced cross-presentation, phago-lysosome fusion was blocked by the topological re-organization of lysosomes into perinuclear clusters. LPS-induced lysosomal clustering, inhibition of phago-lysosome fusion and enhanced cross-presentation, all required expression of the GTPase Rab34. We conclude that TLR4 engagement induces a Rab34-dependent re-organization of lysosomal distribution that delays antigen degradation to transiently enhance cross-presentation, thereby optimizing the priming of CD8(+) T cell responses against pathogens.


Asunto(s)
Presentación de Antígeno/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Receptor Toll-Like 4/inmunología , Animales , Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica/inmunología , Femenino , Citometría de Flujo , Lisosomas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagosomas/inmunología , ARN Interferente Pequeño , Transfección , Proteínas de Unión al GTP rab/inmunología
4.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260403

RESUMEN

Injection of effector proteins to block host innate immune signaling is a common strategy used by many pathogenic organisms to establish an infection. For example, pathogenic Yersinia species inject the acetyltransferase YopJ into target cells to inhibit NF-κB and MAPK signaling. To counteract this, detection of YopJ activity in myeloid cells promotes the assembly of a RIPK1-caspase-8 death-inducing platform that confers antibacterial defense. While recent studies revealed that caspase-8 cleaves the pore-forming protein gasdermin D to trigger pyroptosis in macrophages, whether RIPK1 activates additional substrates downstream of caspase-8 to promote host defense is unclear. Here, we report that the related gasdermin family member gasdermin E (GSDME) is activated upon detection of YopJ activity in a RIPK1 kinase-dependent manner. Specifically, GSDME promotes neutrophil pyroptosis and IL-1ß release, which is critical for anti-Yersinia defense. During in vivo infection, IL-1ß neutralization increases bacterial burden in wild-type but not Gsdme-deficient mice. Thus, our study establishes GSDME as an important mediator that counteracts pathogen blockade of innate immune signaling.


Asunto(s)
Inmunidad Innata , Macrófagos/metabolismo , Proteínas de Neoplasias/metabolismo , Neutrófilos/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Yersinia pseudotuberculosis/fisiología , Células 3T3 , Animales , Citocinas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Piroptosis , Infecciones por Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/microbiología
5.
PLoS Pathog ; 17(10): e1009967, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34648590

RESUMEN

Cell death plays a critical role in inflammatory responses. During pyroptosis, inflammatory caspases cleave Gasdermin D (GSDMD) to release an N-terminal fragment that generates plasma membrane pores that mediate cell lysis and IL-1 cytokine release. Terminal cell lysis and IL-1ß release following caspase activation can be uncoupled in certain cell types or in response to particular stimuli, a state termed hyperactivation. However, the factors and mechanisms that regulate terminal cell lysis downstream of GSDMD cleavage remain poorly understood. In the course of studies to define regulation of pyroptosis during Yersinia infection, we identified a line of Card19-deficient mice (Card19lxcn) whose macrophages were protected from cell lysis and showed reduced apoptosis and pyroptosis, yet had wild-type levels of caspase activation, IL-1 secretion, and GSDMD cleavage. Unexpectedly, CARD19, a mitochondrial CARD-containing protein, was not directly responsible for this, as an independently-generated CRISPR/Cas9 Card19 knockout mouse line (Card19Null) showed no defect in macrophage cell lysis. Notably, Card19 is located on chromosome 13, immediately adjacent to Ninj1, which was recently found to regulate cell lysis downstream of GSDMD activation. RNA-seq and western blotting revealed that Card19lxcn BMDMs have significantly reduced NINJ1 expression, and reconstitution of Ninj1 in Card19lxcn immortalized BMDMs restored their ability to undergo cell lysis in response to caspase-dependent cell death stimuli. Card19lxcn mice exhibited increased susceptibility to Yersinia infection, whereas independently-generated Card19Null mice did not, demonstrating that cell lysis itself plays a key role in protection against bacterial infection, and that the increased infection susceptibility of Card19lxcn mice is attributable to loss of NINJ1. Our findings identify genetic targeting of Card19 being responsible for off-target effects on the adjacent gene Ninj1, disrupting the ability of macrophages to undergo plasma membrane rupture downstream of gasdermin cleavage and impacting host survival and bacterial control during Yersinia infection.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Macrófagos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Yersiniosis/patología , Animales , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Piroptosis/fisiología , Yersiniosis/metabolismo
6.
Immunity ; 39(1): 160-70, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23850380

RESUMEN

Dendritic cell (DC) activation is essential for the induction of immune defense against pathogens, yet needs to be tightly controlled to avoid chronic inflammation and exaggerated immune responses. Here, we identify a mechanism of immune homeostasis by which adaptive immunity, once triggered, tempers DC activation and prevents overreactive immune responses. T cells, once activated, produced Protein S (Pros1) that signaled through TAM receptor tyrosine kinases in DCs to limit the magnitude of DC activation. Genetic ablation of Pros1 in mouse T cells led to increased expression of costimulatory molecules and cytokines in DCs and enhanced immune responses to T cell-dependent antigens, as well as increased colitis. Additionally, PROS1 was expressed in activated human T cells, and its ability to regulate DC activation was conserved. Our results identify a heretofore unrecognized, homeostatic negative feedback mechanism at the interface of adaptive and innate immunity that maintains the physiological magnitude of the immune response.


Asunto(s)
Inmunidad Adaptativa/inmunología , Células Dendríticas/inmunología , Proteína S/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Animales , Células Cultivadas , Colitis/genética , Colitis/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/metabolismo , Citometría de Flujo , Expresión Génica/inmunología , Humanos , Immunoblotting , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína S/genética , Proteína S/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/metabolismo
7.
Proc Natl Acad Sci U S A ; 116(24): 11916-11925, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31138702

RESUMEN

The transcriptional programs that regulate CD8 T-cell differentiation and function in the context of viral infections or tumor immune surveillance have been extensively studied; yet how long noncoding RNAs (lncRNAs) and the loci that transcribe them contribute to the regulation of CD8 T cells during viral infections remains largely unexplored. Here, we report that transcription of the lncRNA Morrbid is specifically induced by T-cell receptor (TCR) and type I IFN stimulation during the early stages of acute and chronic lymphocytic choriomeningitis virus (LCMV) infection. In response to type I IFN, the Morrbid RNA and its locus control CD8 T cell expansion, survival, and effector function by regulating the expression of the proapoptotic factor, Bcl2l11, and by modulating the strength of the PI3K-AKT signaling pathway. Thus, our results demonstrate that inflammatory cue-responsive lncRNA loci represent fundamental mechanisms by which CD8 T cells are regulated in response to pathogens and potentially cancer.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Coriomeningitis Linfocítica/inmunología , ARN Largo no Codificante/inmunología , Animales , Linfocitos T CD8-positivos/virología , Diferenciación Celular/inmunología , Interferón Tipo I/inmunología , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología
8.
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
9.
Proc Natl Acad Sci U S A ; 110(32): 13091-6, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23878224

RESUMEN

The receptor tyrosine kinases Axl and Mer, belonging to the Tyro3, Axl and Mer (TAM) receptor family, are expressed in a number of tumor cells and have well-characterized oncogenic roles. The therapeutic targeting of these kinases is considered an anticancer strategy, and various inhibitors are currently under development. At the same time, Axl and Mer are expressed in dendritic cells and macrophages and have an essential function in limiting inflammation. Inflammation is an enabling characteristic of multiple cancer hallmarks. These contrasting oncogenic and anti-inflammatory functions of Axl and Mer posit a potential paradox in terms of anticancer therapy. Here we demonstrate that azoxymethane (AOM) and dextran sulfate sodium (DSS)-induced inflammation-associated cancer is exacerbated in mice lacking Axl and Mer. Ablation of Axl and Mer signaling is associated with increased production of proinflammatory cytokines and failure to clear apoptotic neutrophils in the intestinal lamina propria, thereby favoring a tumor-promoting environment. Interestingly, loss of these genes in the hematopoietic compartment is not associated with increased colitis. Axl and Mer are expressed in radioresistant intestinal macrophages, and the loss of these genes is associated with an increased inflammatory signature in this compartment. Our results raise the possibility of potential adverse effects of systemic anticancer therapies with Axl and Mer inhibitors, and underscore the importance of understanding their tissue and cell type-specific functions in cancer.


Asunto(s)
Colitis/inmunología , Neoplasias del Colon/inmunología , Proteínas Proto-Oncogénicas/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Animales , Apoptosis/genética , Apoptosis/inmunología , Azoximetano , Colitis/inducido químicamente , Colitis/genética , Colon/inmunología , Colon/metabolismo , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Citocinas/genética , Citocinas/inmunología , Sulfato de Dextran , Femenino , Citometría de Flujo , Expresión Génica/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Fagocitosis/genética , Fagocitosis/inmunología , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/inmunología , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
10.
J Hepatol ; 63(3): 670-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25908269

RESUMEN

BACKGROUND & AIMS: Liver fibrosis, an important health concern associated to chronic liver injury that provides a permissive environment for cancer development, is characterized by accumulation of extracellular matrix components mainly derived from activated hepatic stellate cells (HSCs). Axl, a receptor tyrosine kinase and its ligand Gas6, are involved in cell differentiation, immune response and carcinogenesis. METHODS: HSCs were obtained from WT and Axl(-/-) mice, treated with recombinant Gas6 protein (rGas6), Axl siRNAs or the Axl inhibitor BGB324, and analyzed by western blot and real-time PCR. Experimental fibrosis was studied in CCl4-treated WT and Axl(-/-) mice, and in combination with Axl inhibitor. Gas6 and Axl serum levels were measured in alcoholic liver disease (ALD) and hepatitis C virus (HCV) patients. RESULTS: In primary mouse HSCs, Gas6 and Axl levels paralleled HSC activation. rGas6 phosphorylated Axl and AKT prior to HSC phenotypic changes, while Axl siRNA silencing reduced HSC activation. Moreover, BGB324 blocked Axl/AKT phosphorylation and diminished HSC activation. In addition, Axl(-/-) mice displayed decreased HSC activation in vitro and liver fibrogenesis after chronic damage by CCl4 administration. Similarly, BGB324 reduced collagen deposition and CCl4-induced liver fibrosis in mice. Importantly, Gas6 and Axl serum levels increased in ALD and HCV patients, inversely correlating with liver functionality. CONCLUSIONS: The Gas6/Axl axis is required for full HSC activation. Gas6 and Axl serum levels increase in parallel to chronic liver disease progression. Axl targeting may be a therapeutic strategy for liver fibrosis management.


Asunto(s)
Células Estrelladas Hepáticas/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Cirrosis Hepática/etiología , Transducción de Señal/fisiología , Adulto , Anciano , Animales , Tetracloruro de Carbono , Proliferación Celular , Células Cultivadas , Enfermedad Crónica , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Cirrosis Hepática/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , FN-kappa B/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Tirosina Quinasa c-Mer
11.
Nat Commun ; 15(1): 5394, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918428

RESUMEN

Adipose tissue macrophages (ATMs) influence obesity-associated metabolic dysfunction, but the mechanisms by which they do so are not well understood. We show that miR-6236 is a bona fide miRNA that is secreted by ATMs during obesity. Global or myeloid cell-specific deletion of miR-6236 aggravates obesity-associated adipose tissue insulin resistance, hyperglycemia, hyperinsulinemia, and hyperlipidemia. miR-6236 augments adipocyte insulin sensitivity by inhibiting translation of negative regulators of insulin signaling, including PTEN. The human genome harbors a miR-6236 homolog that is highly expressed in the serum and adipose tissue of obese people. hsa-MIR-6236 expression negatively correlates with hyperglycemia and glucose intolerance, and positively correlates with insulin sensitivity. Together, our findings establish miR-6236 as an ATM-secreted miRNA that potentiates adipocyte insulin signaling and protects against metabolic dysfunction during obesity.


Asunto(s)
Adipocitos , Hiperglucemia , Resistencia a la Insulina , Insulina , MicroARNs , Obesidad , Fosfohidrolasa PTEN , Transducción de Señal , MicroARNs/metabolismo , MicroARNs/genética , Obesidad/metabolismo , Obesidad/genética , Animales , Adipocitos/metabolismo , Hiperglucemia/metabolismo , Hiperglucemia/genética , Humanos , Insulina/metabolismo , Resistencia a la Insulina/genética , Ratones , Masculino , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética , Ratones Endogámicos C57BL , Macrófagos/metabolismo , Tejido Adiposo/metabolismo , Células Mieloides/metabolismo , Ratones Noqueados , Hiperinsulinismo/metabolismo , Hiperinsulinismo/genética
12.
Cell Metab ; 35(8): 1441-1456.e9, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37494932

RESUMEN

This study reveals a previously uncharacterized mechanism to restrict intestinal inflammation via a regulatory RNA transcribed from a noncoding genomic locus. We identified a novel transcript of the lncRNA HOXA11os specifically expressed in the distal colon that is reduced to undetectable levels in colitis. HOXA11os is localized to mitochondria under basal conditions and interacts with a core subunit of complex 1 of the electron transport chain (ETC) to maintain its activity. Deficiency of HOXA11os in colonic myeloid cells results in complex I deficiency, dysfunctional oxidative phosphorylation (OXPHOS), and the production of mitochondrial reactive oxygen species (mtROS). As a result, HOXA11os-deficient mice develop spontaneous intestinal inflammation and are hypersusceptible to colitis. Collectively, these studies identify a new regulatory axis whereby a lncRNA maintains intestinal homeostasis and restricts inflammation in the colon through the regulation of complex I activity.


Asunto(s)
Colitis , ARN Largo no Codificante , Animales , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Colitis/genética , Colitis/metabolismo , Inflamación/metabolismo , Mitocondrias/genética , Homeostasis , Mucosa Intestinal/metabolismo
13.
J Exp Med ; 219(12)2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36074090

RESUMEN

The intestinal epithelium is a key physical interface that integrates dietary and microbial signals to regulate nutrient uptake and mucosal immune cell function. The transcriptional programs that regulate intestinal epithelial cell (IEC) quiescence, proliferation, and differentiation have been well characterized. However, how gene expression networks critical for IECs are posttranscriptionally regulated during homeostasis or inflammatory disease remains poorly understood. Herein, we show that a conserved family of microRNAs, miR-181, is significantly downregulated in IECs from patients with inflammatory bowel disease and mice with chemical-induced colitis. Strikingly, we showed that miR-181 expression within IECs, but not the hematopoietic system, is required for protection against severe colonic inflammation in response to epithelial injury in mice. Mechanistically, we showed that miR-181 expression increases the proliferative capacity of IECs, likely through the regulation of Wnt signaling, independently of the gut microbiota composition. As epithelial reconstitution is crucial to restore intestinal homeostasis after injury, the miR-181 family represents a potential therapeutic target against severe intestinal inflammation.


Asunto(s)
Colitis , MicroARNs , Animales , Colitis/inducido químicamente , Colitis/genética , Células Epiteliales/metabolismo , Inflamación/genética , Inflamación/metabolismo , Mucosa Intestinal , Ratones , MicroARNs/genética , MicroARNs/metabolismo
14.
Cell Rep ; 32(2): 107905, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32668257

RESUMEN

Cross-presentation of antigens by dendritic cells (DCs) is critical for initiation of anti-tumor immune responses. Yet, key steps involved in trafficking of antigens taken up by DCs remain incompletely understood. Here, we screen 700 US Food and Drug Administration (FDA)-approved drugs and identify 37 enhancers of antigen import from endolysosomes into the cytosol. To reveal their mechanism of action, we generate proteomic organellar maps of control and drug-treated DCs (focusing on two compounds, prazosin and tamoxifen). By combining organellar mapping, quantitative proteomics, and microscopy, we conclude that import enhancers undergo lysosomal trapping leading to membrane permeation and antigen release. Enhancing antigen import facilitates cross-presentation of soluble and cell-associated antigens. Systemic administration of prazosin leads to reduced growth of MC38 tumors and to a synergistic effect with checkpoint immunotherapy in a melanoma model. Thus, inefficient antigen import into the cytosol limits antigen cross-presentation, restraining the potency of anti-tumor immune responses and efficacy of checkpoint blockers.


Asunto(s)
Antineoplásicos/farmacología , Citosol/metabolismo , Endosomas/metabolismo , Inmunidad , Neoplasias/inmunología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antígenos/metabolismo , Transporte Biológico/efectos de los fármacos , Reactividad Cruzada/efectos de los fármacos , Citosol/efectos de los fármacos , Células Dendríticas/metabolismo , Degradación Asociada con el Retículo Endoplásmico/efectos de los fármacos , Endosomas/efectos de los fármacos , Inmunidad/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/tratamiento farmacológico , Permeabilidad , Prazosina/farmacología , Quinazolinas/farmacología , Tamoxifeno/farmacología , beta-Lactamasas/metabolismo
15.
Sci Transl Med ; 11(496)2019 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-31189717

RESUMEN

The gut microbiota is a key environmental determinant of mammalian metabolism. Regulation of white adipose tissue (WAT) by the gut microbiota is a process critical to maintaining metabolic fitness, and gut dysbiosis can contribute to the development of obesity and insulin resistance (IR). However, how the gut microbiota regulates WAT function remains largely unknown. Here, we show that tryptophan-derived metabolites produced by the gut microbiota controlled the expression of the miR-181 family in white adipocytes in mice to regulate energy expenditure and insulin sensitivity. Moreover, dysregulation of the gut microbiota-miR-181 axis was required for the development of obesity, IR, and WAT inflammation in mice. Our results indicate that regulation of miR-181 in WAT by gut microbiota-derived metabolites is a central mechanism by which host metabolism is tuned in response to dietary and environmental changes. As we also found that MIR-181 expression in WAT and the plasma abundance of tryptophan-derived metabolites were dysregulated in a cohort of obese human children, the MIR-181 family may represent a potential therapeutic target to modulate WAT function in the context of obesity.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Inflamación/metabolismo , Obesidad/metabolismo , Adipocitos/metabolismo , Animales , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Microbioma Gastrointestinal/genética , Inflamación/genética , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Obesidad/genética , Triptófano/metabolismo
16.
J Exp Med ; 215(4): 1245-1265, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29440364

RESUMEN

The adapter molecule linker for activation of T cells (LAT) orchestrates the formation of signalosomes upon T cell receptor (TCR) stimulation. LAT is present in different intracellular pools and is dynamically recruited to the immune synapse upon stimulation. However, the intracellular traffic of LAT and its function in T lymphocyte activation are ill defined. We show herein that LAT, once internalized, transits through the Golgi-trans-Golgi network (TGN), where it is repolarized to the immune synapse. This retrograde transport of LAT depends on the small GTPase Rab6 and the target soluble N-ethylmaleimide-sensitive factor attachment protein receptor (t-SNARE) Syntaxin-16, two regulators of the endosome-to-Golgi/TGN retrograde transport. We also show in vitro in Syntaxin-16- or Rab6-silenced human cells and in vivo in CD4+ T lymphocytes of the Rab6 knockout mouse that this retrograde traffic controls TCR stimulation. These results establish that the retrograde traffic of LAT from the plasma membrane to the Golgi-TGN controls the polarized delivery of LAT at the immune synapse and T lymphocyte activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Sinapsis Inmunológicas/metabolismo , Activación de Linfocitos/inmunología , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Linfocitos T/inmunología , Proteínas de Unión al GTP rab/metabolismo , Animales , Membrana Celular/metabolismo , Endosomas/metabolismo , Humanos , Interleucina-2/metabolismo , Células Jurkat , Ratones , Modelos Biológicos , Fosforilación , Transporte de Proteínas , Proteínas R-SNARE/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Sintaxina 16/metabolismo , Red trans-Golgi
17.
J Exp Med ; 214(8): 2231-2241, 2017 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-28663435

RESUMEN

CD8+ T cells mediate antigen-specific immune responses that can induce rejection of solid tumors. In this process, dendritic cells (DCs) are thought to take up tumor antigens, which are processed into peptides and loaded onto MHC-I molecules, a process called "cross-presentation." Neither the actual contribution of cross-presentation to antitumor immune responses nor the intracellular pathways involved in vivo are clearly established because of the lack of experimental tools to manipulate this process. To develop such tools, we generated mice bearing a conditional DC-specific mutation in the sec22b gene, a critical regulator of endoplasmic reticulum-phagosome traffic required for cross-presentation. DCs from these mice show impaired cross-presentation ex vivo and defective cross-priming of CD8+ T cell responses in vivo. These mice are also defective for antitumor immune responses and are resistant to treatment with anti-PD-1. We conclude that Sec22b-dependent cross-presentation in DCs is required to initiate CD8+ T cell responses to dead cells and to induce effective antitumor immune responses during anti-PD-1 treatment in mice.


Asunto(s)
Reactividad Cruzada/inmunología , Neoplasias/inmunología , Proteínas R-SNARE/fisiología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/fisiología , Muerte Celular/inmunología , Células Dendríticas/inmunología , Femenino , Inmunidad Celular/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas R-SNARE/genética , Células RAW 264.7
18.
Science ; 352(6281): 99-103, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27034374

RESUMEN

Host responses against metazoan parasites or an array of environmental substances elicit type 2 immunity. Despite its protective function, type 2 immunity also drives allergic diseases. The mechanisms that regulate the magnitude of the type 2 response remain largely unknown. Here, we show that genetic ablation of a receptor tyrosine kinase encoded byTyro3in mice or the functional neutralization of its ortholog in human dendritic cells resulted in enhanced type 2 immunity. Furthermore, the TYRO3 agonist PROS1 was induced in T cells by the quintessential type 2 cytokine, interleukin-4. T cell-specificPros1knockouts phenocopied the loss ofTyro3 Thus, a PROS1-mediated feedback from adaptive immunity engages a rheostat, TYRO3, on innate immune cells to limit the intensity of type 2 responses.


Asunto(s)
Inmunidad Adaptativa/genética , Asma/inmunología , Interacciones Huésped-Parásitos/inmunología , Inmunidad Innata/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Animales , Asma/genética , Proteínas Sanguíneas/antagonistas & inhibidores , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Interacciones Huésped-Parásitos/genética , Humanos , Interleucina-4/inmunología , Interleucina-4/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nippostrongylus/inmunología , Proteína S , Pyroglyphidae/inmunología , Proteínas Tirosina Quinasas Receptoras/genética , Infecciones por Strongylida/inmunología , Linfocitos T/inmunología
20.
Biopharm Drug Dispos ; 28(6): 283-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17570125

RESUMEN

It has been shown that the expression of the morphine (MOR) withdrawal syndrome precipitated by naloxone (NAL) is more intense in male mice than in females, but the reasons for this phenomenon remain uncertain. The purpose of the present study was to evaluate whether this sexual dimorphism might be due to differences in MOR and/or NAL plasma levels after a chronic treatment with MOR. Prepubertal Swiss male and female mice were rendered dependent by intraperitoneal (i.p.) injection of MOR (2 mg/kg), twice daily for 9 days. On day 10 dependent mice received NAL (6 mg/kg, i.p.) 60 min after MOR injection. Blood samples were taken at different times in order to determine MOR and NAL plasma levels by gas chromatography-mass spectrometry (GC-MS) and high-performance liquid chromatography (HPLC), respectively. Pharmacokinetic analysis showed no differences between male and female mice either for MOR or for NAL. In conclusion, although males and females respond differentially to NAL-precipitated withdrawal, this dimorphic behavior would not be influenced by a pharmacokinetic factor.


Asunto(s)
Morfina/farmacocinética , Naloxona/farmacocinética , Síndrome de Abstinencia a Sustancias/metabolismo , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacocinética , Animales , Área Bajo la Curva , Cromatografía Líquida de Alta Presión , Femenino , Cromatografía de Gases y Espectrometría de Masas , Semivida , Inyecciones Intraperitoneales , Masculino , Ratones , Morfina/administración & dosificación , Dependencia de Morfina/sangre , Dependencia de Morfina/complicaciones , Naloxona/administración & dosificación , Naloxona/sangre , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacocinética , Factores Sexuales , Maduración Sexual , Síndrome de Abstinencia a Sustancias/etiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA