Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(1): 149-168.e17, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33278357

RESUMEN

COVID-19 is characterized by excessive production of pro-inflammatory cytokines and acute lung damage associated with patient mortality. While multiple inflammatory cytokines are produced by innate immune cells during SARS-CoV-2 infection, we found that only the combination of TNF-α and IFN-γ induced inflammatory cell death characterized by inflammatory cell death, PANoptosis. Mechanistically, TNF-α and IFN-γ co-treatment activated the JAK/STAT1/IRF1 axis, inducing nitric oxide production and driving caspase-8/FADD-mediated PANoptosis. TNF-α and IFN-γ caused a lethal cytokine shock in mice that mirrors the tissue damage and inflammation of COVID-19, and inhibiting PANoptosis protected mice from this pathology and death. Furthermore, treating with neutralizing antibodies against TNF-α and IFN-γ protected mice from mortality during SARS-CoV-2 infection, sepsis, hemophagocytic lymphohistiocytosis, and cytokine shock. Collectively, our findings suggest that blocking the cytokine-mediated inflammatory cell death signaling pathway identified here may benefit patients with COVID-19 or other infectious and autoinflammatory diseases by limiting tissue damage/inflammation.


Asunto(s)
COVID-19/inmunología , COVID-19/patología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/patología , Interferón gamma/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Anticuerpos Neutralizantes/administración & dosificación , Muerte Celular , Modelos Animales de Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/inmunología , Inflamación/patología , Linfohistiocitosis Hemofagocítica/inducido químicamente , Masculino , Ratones , Ratones Transgénicos , Células THP-1
2.
Cell ; 173(4): 920-933.e13, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29576451

RESUMEN

Inflammasome activation is critical for host defenses against various microbial infections. Activation of the NLRC4 inflammasome requires detection of flagellin or type III secretion system (T3SS) components by NLR family apoptosis inhibitory proteins (NAIPs); yet how this pathway is regulated is unknown. Here, we found that interferon regulatory factor 8 (IRF8) is required for optimal activation of the NLRC4 inflammasome in bone-marrow-derived macrophages infected with Salmonella Typhimurium, Burkholderia thailandensis, or Pseudomonas aeruginosa but is dispensable for activation of the canonical and non-canonical NLRP3, AIM2, and Pyrin inflammasomes. IRF8 governs the transcription of Naips to allow detection of flagellin or T3SS proteins to mediate NLRC4 inflammasome activation. Furthermore, we found that IRF8 confers protection against bacterial infection in vivo, owing to its role in inflammasome-dependent cytokine production and pyroptosis. Altogether, our findings suggest that IRF8 is a critical regulator of NAIPs and NLRC4 inflammasome activation for defense against bacterial infection.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Inflamasomas/metabolismo , Factores Reguladores del Interferón/metabolismo , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al Calcio/genética , Células Cultivadas , Citocinas/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Flagelina/metabolismo , Factores Reguladores del Interferón/antagonistas & inhibidores , Factores Reguladores del Interferón/genética , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína Inhibidora de la Apoptosis Neuronal/genética , Regiones Promotoras Genéticas , Unión Proteica , Pseudomonas aeruginosa/patogenicidad , Piroptosis , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Salmonella typhimurium/patogenicidad , Transcripción Genética
3.
Cell ; 166(5): 1132-1146.e7, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27565343

RESUMEN

Cancers are distributed unevenly across the body, but the importance of cell intrinsic factors such as stem cell function in determining organ cancer risk is unknown. Therefore, we used Cre-recombination of conditional lineage tracing, oncogene, and tumor suppressor alleles to define populations of stem and non-stem cells in mouse organs and test their life-long susceptibility to tumorigenesis. We show that tumor incidence is determined by the life-long generative capacity of mutated cells. This relationship held true in the presence of multiple genotypes and regardless of developmental stage, strongly supporting the notion that stem cells dictate organ cancer risk. Using the liver as a model system, we further show that damage-induced activation of stem cell function markedly increases cancer risk. Therefore, we propose that a combination of stem cell mutagenesis and extrinsic factors that enhance the proliferation of these cell populations, creates a "perfect storm" that ultimately determines organ cancer risk. VIDEO ABSTRACT.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Oncogenes , Células Madre , Alelos , Animales , Genes Supresores de Tumor , Humanos , Integrasas , Ratones , Modelos Biológicos , Mutagénesis , Recombinación Genética , Riesgo , Células Madre/metabolismo , Células Madre/patología
4.
Cell ; 167(2): 382-396.e17, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27693356

RESUMEN

The inflammasome is an intracellular signaling complex, which on recognition of pathogens and physiological aberration, drives activation of caspase-1, pyroptosis, and the release of the pro-inflammatory cytokines IL-1ß and IL-18. Bacterial ligands must secure entry into the cytoplasm to activate inflammasomes; however, the mechanisms by which concealed ligands are liberated in the cytoplasm have remained unclear. Here, we showed that the interferon-inducible protein IRGB10 is essential for activation of the DNA-sensing AIM2 inflammasome by Francisella novicida and contributed to the activation of the LPS-sensing caspase-11 and NLRP3 inflammasome by Gram-negative bacteria. IRGB10 directly targeted cytoplasmic bacteria through a mechanism requiring guanylate-binding proteins. Localization of IRGB10 to the bacterial cell membrane compromised bacterial structural integrity and mediated cytosolic release of ligands for recognition by inflammasome sensors. Overall, our results reveal IRGB10 as part of a conserved signaling hub at the interface between cell-autonomous immunity and innate immune sensing pathways.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Francisella/inmunología , GTP Fosfohidrolasas/metabolismo , Infecciones por Bacterias Gramnegativas/inmunología , Interacciones Huésped-Patógeno/inmunología , Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Linfocitos B/inmunología , Caspasas/metabolismo , Caspasas Iniciadoras , Citosol/inmunología , Citosol/microbiología , GTP Fosfohidrolasas/genética , Infecciones por Bacterias Gramnegativas/microbiología , Inmunidad Celular , Inmunidad Innata , Inflamasomas/metabolismo , Ligandos , Ratones , Ratones Mutantes , Células Mieloides/inmunología , Linfocitos T/inmunología
5.
Cell ; 162(1): 45-58, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26095253

RESUMEN

Colorectal cancer is a leading cause of cancer-related deaths. Mutations in the innate immune sensor AIM2 are frequently identified in patients with colorectal cancer, but how AIM2 modulates colonic tumorigenesis is unknown. Here, we found that Aim2-deficient mice were hypersusceptible to colonic tumor development. Production of inflammasome-associated cytokines and other inflammatory mediators was largely intact in Aim2-deficient mice; however, intestinal stem cells were prone to uncontrolled proliferation. Aberrant Wnt signaling expanded a population of tumor-initiating stem cells in the absence of AIM2. Susceptibility of Aim2-deficient mice to colorectal tumorigenesis was enhanced by a dysbiotic gut microbiota, which was reduced by reciprocal exchange of gut microbiota with healthy wild-type mice. These findings uncover a synergy between a specific host genetic factor and gut microbiota in determining the susceptibility to colorectal cancer. Therapeutic modulation of AIM2 expression and microbiota has the potential to prevent colorectal cancer.


Asunto(s)
Proliferación Celular , Neoplasias Colorrectales/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre/patología , Animales , Azoximetano , Colitis/inducido químicamente , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Sulfato de Dextran , Enterocitos/patología , Tracto Gastrointestinal/microbiología , Inflamasomas/metabolismo , Ratones , Mutación , Células Madre/metabolismo
6.
Nat Immunol ; 17(3): 277-85, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26808230

RESUMEN

Regulatory T (Treg) cells respond to immune and inflammatory signals to mediate immunosuppression, but how the functional integrity of Treg cells is maintained under activating environments is unclear. Here we show that autophagy is active in Treg cells and supports their lineage stability and survival fitness. Treg cell-specific deletion of Atg7 or Atg5, two essential genes in autophagy, leads to loss of Treg cells, greater tumor resistance and development of inflammatory disorders. Atg7-deficient Treg cells show increased apoptosis and readily lose expression of the transcription factor Foxp3, especially after activation. Mechanistically, autophagy deficiency upregulates metabolic regulators mTORC1 and c-Myc and glycolysis, which contribute to defective Treg function. Therefore, autophagy couples environmental signals and metabolic homeostasis to protect lineage and survival integrity of Treg cells in activating contexts.


Asunto(s)
Apoptosis/genética , Autofagia/genética , Factores de Transcripción Forkhead/genética , Proteínas Asociadas a Microtúbulos/genética , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Adenocarcinoma/inmunología , Traslado Adoptivo , Animales , Apoptosis/inmunología , Autofagia/inmunología , Proteína 5 Relacionada con la Autofagia , Proteína 7 Relacionada con la Autofagia , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Metilación de ADN , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucólisis , Homeostasis , Immunoblotting , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Trasplante de Neoplasias , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba
7.
Nat Immunol ; 16(2): 178-87, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25559258

RESUMEN

The interplay between effector T cells and regulatory T cells (Treg cells) is crucial for adaptive immunity, but how Treg cells control diverse effector responses is elusive. We found that the phosphatase PTEN links Treg cell stability to repression of type 1 helper T cell (TH1 cell) and follicular helper T cell (TFH cell) responses. Depletion of PTEN in Treg cells resulted in excessive TFH cell and germinal center responses and spontaneous inflammatory disease. These defects were considerably blocked by deletion of interferon-γ, indicating coordinated control of TH1 and TFH responses. Mechanistically, PTEN maintained Treg cell stability and metabolic balance between glycolysis and mitochondrial fitness. Moreover, PTEN deficiency upregulates activity of the metabolic checkpoint kinase complex mTORC2 and the serine-threonine kinase Akt, and loss of this activity restores functioning of PTEN-deficient Treg cells. Our studies establish a PTEN-mTORC2 axis that maintains Treg cell stability and coordinates Treg cell-mediated control of effector responses.


Asunto(s)
Fosfohidrolasa PTEN/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Animales , Linfocitos B/inmunología , Factores de Transcripción Forkhead/metabolismo , Humanos , Activación de Linfocitos , Ratones , Proteínas Represoras/metabolismo , Transducción de Señal , Células TH1/enzimología
8.
Nat Immunol ; 16(5): 467-75, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25774715

RESUMEN

Inflammasomes are critical for mounting host defense against pathogens. The molecular mechanisms that control activation of the AIM2 inflammasome in response to different cytosolic pathogens remain unclear. Here we found that the transcription factor IRF1 was required for activation of the AIM2 inflammasome during infection with the Francisella tularensis subspecies novicida (F. novicida), whereas engagement of the AIM2 inflammasome by mouse cytomegalovirus (MCMV) or transfected double-stranded DNA did not require IRF1. Infection of F. novicida detected by the DNA sensor cGAS and its adaptor STING induced type I interferon-dependent expression of IRF1, which drove the expression of guanylate-binding proteins (GBPs); this led to intracellular killing of bacteria and DNA release. Our results reveal a specific requirement for IRF1 and GBPs in the liberation of DNA for sensing by AIM2 depending on the pathogen encountered by the cell.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Francisella tularensis/fisiología , Proteínas de Unión al GTP/metabolismo , Inflamasomas/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Tularemia/inmunología , Animales , Bacteriólisis/genética , Células Cultivadas , ADN/inmunología , ADN Bacteriano/genética , Regulación de la Expresión Génica/genética , Factor 1 Regulador del Interferón/genética , Interferón Tipo I/metabolismo , Ratones , Ratones Noqueados , Nucleotidiltransferasas/metabolismo
9.
Immunity ; 49(3): 515-530.e5, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30231985

RESUMEN

Fungi represent a significant proportion of the gut microbiota. Aberrant immune responses to fungi are frequently observed in inflammatory bowel diseases (IBD) and colorectal cancer (CRC), and mutations in the fungal-sensing pathways are associated with the pathogenesis of IBD. Fungal recognition receptors trigger downstream signaling via the common adaptor protein CARD9 and the kinase SYK. Here we found that commensal gut fungi promoted inflammasome activation during AOM-DSS-induced colitis. Myeloid cell-specific deletion of Card9 or Syk reduced inflammasome activation and interleukin (IL)-18 maturation and increased susceptibility to colitis and CRC. IL-18 promoted epithelial barrier restitution and interferon-γ production by intestinal CD8+ T cells. Supplementation of IL-18 or transfer of wild-type myeloid cells reduced tumor burden in AOM-DSS-treated Card9-/- and Sykfl/flLysMCre/+ mice, whereas treatment with anti-fungal agents exacerbated colitis and CRC. CARD9 deletion changes the gut microbial landscape, suggesting that SYK-CARD9 signaling maintains a microbial ecology that promotes inflammasome activation and thereby restrains colitis and colon tumorigenesis.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Colitis/inmunología , Neoplasias del Colon/inmunología , Hongos/inmunología , Microbioma Gastrointestinal/inmunología , Inflamasomas/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/fisiología , Células Mieloides/fisiología , Quinasa Syk/metabolismo , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Células Cultivadas , Colitis/inducido químicamente , Modelos Animales de Enfermedad , Humanos , Interleucina-18/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Dodecil Sulfato de Sodio , Quinasa Syk/genética
10.
Immunity ; 49(5): 899-914.e6, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30413360

RESUMEN

Interleukin-2 (IL-2) and downstream transcription factor STAT5 are important for maintaining regulatory T (Treg) cell homeostasis and function. Treg cells can respond to low IL-2 levels, but the mechanisms of STAT5 activation during partial IL-2 deficiency remain uncertain. We identified the serine-threonine kinase Mst1 as a signal-dependent amplifier of IL-2-STAT5 activity in Treg cells. High Mst1 and Mst2 (Mst1-Mst2) activity in Treg cells was crucial to prevent tumor resistance and autoimmunity. Mechanistically, Mst1-Mst2 sensed IL-2 signals to promote the STAT5 activation necessary for Treg cell homeostasis and lineage stability and to maintain the highly suppressive phosphorylated-STAT5+ Treg cell subpopulation. Unbiased quantitative proteomics revealed association of Mst1 with the cytoskeletal DOCK8-LRCHs module. Mst1 deficiency limited Treg cell migration and access to IL-2 and activity of the small GTPase Rac, which mediated downstream STAT5 activation. Collectively, IL-2-STAT5 signaling depends upon Mst1-Mst2 functions to maintain a stable Treg cell pool and immune tolerance.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Interleucina-2/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Linaje de la Célula/genética , Factor de Crecimiento de Hepatocito/genética , Vía de Señalización Hippo , Interleucina-2/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Serina-Treonina Quinasa 3 , Linfocitos T Reguladores/inmunología , Proteínas de Unión al GTP rac/metabolismo
11.
Nature ; 580(7803): 396-401, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32296180

RESUMEN

Cancer genomics has revealed many genes and core molecular processes that contribute to human malignancies, but the genetic and molecular bases of many rare cancers remains unclear. Genetic predisposition accounts for 5 to 10% of cancer diagnoses in children1,2, and genetic events that cooperate with known somatic driver events are poorly understood. Pathogenic germline variants in established cancer predisposition genes have been recently identified in 5% of patients with the malignant brain tumour medulloblastoma3. Here, by analysing all protein-coding genes, we identify and replicate rare germline loss-of-function variants across ELP1 in 14% of paediatric patients with the medulloblastoma subgroup Sonic Hedgehog (MBSHH). ELP1 was the most common medulloblastoma predisposition gene and increased the prevalence of genetic predisposition to 40% among paediatric patients with MBSHH. Parent-offspring and pedigree analyses identified two families with a history of paediatric medulloblastoma. ELP1-associated medulloblastomas were restricted to the molecular SHHα subtype4 and characterized by universal biallelic inactivation of ELP1 owing to somatic loss of chromosome arm 9q. Most ELP1-associated medulloblastomas also exhibited somatic alterations in PTCH1, which suggests that germline ELP1 loss-of-function variants predispose individuals to tumour development in combination with constitutive activation of SHH signalling. ELP1 is the largest subunit of the evolutionarily conserved Elongator complex, which catalyses translational elongation through tRNA modifications at the wobble (U34) position5,6. Tumours from patients with ELP1-associated MBSHH were characterized by a destabilized Elongator complex, loss of Elongator-dependent tRNA modifications, codon-dependent translational reprogramming, and induction of the unfolded protein response, consistent with loss of protein homeostasis due to Elongator deficiency in model systems7-9. Thus, genetic predisposition to proteome instability may be a determinant in the pathogenesis of paediatric brain cancers. These results support investigation of the role of protein homeostasis in other cancer types and potential for therapeutic interference.


Asunto(s)
Neoplasias Cerebelosas/metabolismo , Mutación de Línea Germinal , Meduloblastoma/metabolismo , Factores de Elongación Transcripcional/metabolismo , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Niño , Femenino , Humanos , Masculino , Meduloblastoma/genética , Linaje , ARN de Transferencia/metabolismo , Factores de Elongación Transcripcional/genética
12.
Immunity ; 45(3): 540-554, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27637146

RESUMEN

Follicular helper T (Tfh) cells are crucial for germinal center (GC) formation and humoral adaptive immunity. Mechanisms underlying Tfh cell differentiation in peripheral and mucosal lymphoid organs are incompletely understood. We report here that mTOR kinase complexes 1 and 2 (mTORC1 and mTORC2) are essential for Tfh cell differentiation and GC reaction under steady state and after antigen immunization and viral infection. Loss of mTORC1 and mTORC2 in T cells exerted distinct effects on Tfh cell signature gene expression, whereas increased mTOR activity promoted Tfh responses. Deficiency of mTORC2 impaired CD4(+) T cell accumulation and immunoglobulin A production and aberrantly induced the transcription factor Foxo1. Mechanistically, the costimulatory molecule ICOS activated mTORC1 and mTORC2 to drive glycolysis and lipogenesis, and glucose transporter 1-mediated glucose metabolism promoted Tfh cell responses. Altogether, mTOR acts as a central node in Tfh cells by linking immune signals to anabolic metabolism and transcriptional activity.


Asunto(s)
Diferenciación Celular/inmunología , Glucosa/metabolismo , Complejos Multiproteicos/metabolismo , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Centro Germinal/inmunología , Centro Germinal/metabolismo , Inmunidad Humoral/inmunología , Activación de Linfocitos/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos/inmunología , Serina-Treonina Quinasas TOR/inmunología
13.
Nature ; 565(7737): 101-105, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30568299

RESUMEN

A defining feature of adaptive immunity is the development of long-lived memory T cells to curtail infection. Recent studies have identified a unique stem-like T-cell subset amongst exhausted CD8-positive T cells in chronic infection1-3, but it remains unclear whether CD4-positive T-cell subsets with similar features exist in chronic inflammatory conditions. Amongst helper T cells, TH17 cells have prominent roles in autoimmunity and tissue inflammation and are characterized by inherent plasticity4-7, although how such plasticity is regulated is poorly understood. Here we demonstrate that TH17 cells in a mouse model of autoimmune disease are functionally and metabolically heterogeneous; they contain a subset with stemness-associated features but lower anabolic metabolism, and a reciprocal subset with higher metabolic activity that supports transdifferentiation into TH1-like cells. These two TH17-cell subsets are defined by selective expression of the transcription factors TCF-1 and T-bet, and by discrete levels of CD27 expression. We also identify signalling via the kinase complex mTORC1 as a central regulator of TH17-cell fate decisions by coordinating metabolic and transcriptional programmes. TH17 cells with disrupted mTORC1 signalling or anabolic metabolism fail to induce autoimmune neuroinflammation or to develop into TH1-like cells, but instead upregulate TCF-1 expression and acquire stemness-associated features. Single-cell RNA sequencing and experimental validation reveal heterogeneity in fate-mapped TH17 cells, and a developmental arrest in the TH1 transdifferentiation trajectory upon loss of mTORC1 activity or metabolic perturbation. Our results establish that the dichotomy of stemness and effector function underlies the heterogeneous TH17 responses and autoimmune pathogenesis, and point to previously unappreciated metabolic control of plasticity in helper T cells.


Asunto(s)
Transdiferenciación Celular , Células Madre/citología , Células Madre/metabolismo , Células Th17/citología , Células Th17/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Modelos Animales de Enfermedad , Femenino , Memoria Inmunológica/inmunología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Proteína Reguladora Asociada a mTOR/deficiencia , Proteína Reguladora Asociada a mTOR/genética , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual , Células Madre/inmunología , Factor 1 de Transcripción de Linfocitos T/biosíntesis , Factor 1 de Transcripción de Linfocitos T/metabolismo , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/metabolismo , Células Th17/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
14.
PLoS Pathog ; 18(4): e1009716, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35452499

RESUMEN

Human astroviruses (HAstV), positive sense single-stranded RNA viruses, are one of the leading causes of diarrhea worldwide. Despite their high prevalence, the cellular mechanisms of astrovirus pathogenesis remain ill-defined. Previous studies showed HAstV increased epithelial barrier permeability by causing a re-localization of the tight junction protein, occludin. In these studies, we demonstrate that HAstV replication induces epithelial-mesenchymal transition (EMT), by upregulating the transcription of EMT-related genes within 8 hours post-infection (hpi), followed by the loss of cell-cell contacts and disruption of polarity by 24 hpi. While multiple classical HAstV serotypes, including clinical isolates, induce EMT, the non-classical genotype HAstV-VA1 and two strains of reovirus are incapable of inducing EMT. Unlike the re-localization of tight junction proteins, HAstV-induced EMT requires productive replication and is dependent transforming growth factor-ß (TGF-ß) activity. Finally, inhibiting TGF-ß signaling and EMT reduces viral replication, highlighting its importance in the viral life cycle. This finding puts classical strains of HAstV-1 in an exclusive group of non-oncogenic viruses triggering EMT.


Asunto(s)
Infecciones por Astroviridae , Mamastrovirus , Transición Epitelial-Mesenquimal , Humanos , Mamastrovirus/genética , Factor de Crecimiento Transformador beta , Replicación Viral
15.
Nature ; 558(7708): 141-145, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29849151

RESUMEN

Dendritic cells orchestrate the crosstalk between innate and adaptive immunity. CD8α+ dendritic cells present antigens to CD8+ T cells and elicit cytotoxic T cell responses to viruses, bacteria and tumours 1 . Although lineage-specific transcriptional regulators of CD8α+ dendritic cell development have been identified 2 , the molecular pathways that selectively orchestrate CD8α+ dendritic cell function remain elusive. Moreover, metabolic reprogramming is important for dendritic cell development and activation3,4, but metabolic dependence and regulation of dendritic cell subsets are largely uncharacterized. Here we use a data-driven systems biology algorithm (NetBID) to identify a role of the Hippo pathway kinases Mst1 and Mst2 (Mst1/2) in selectively programming CD8α+ dendritic cell function and metabolism. Our NetBID analysis reveals a marked enrichment of the activities of Hippo pathway kinases in CD8α+ dendritic cells relative to CD8α- dendritic cells. Dendritic cell-specific deletion of Mst1/2-but not Lats1 and Lats2 (Lats1/2) or Yap and Taz (Yap/Taz), which mediate canonical Hippo signalling-disrupts homeostasis and function of CD8+ T cells and anti-tumour immunity. Mst1/2-deficient CD8α+ dendritic cells are impaired in presentation of extracellular proteins and cognate peptides to prime CD8+ T cells, while CD8α- dendritic cells that lack Mst1/2 have largely normal function. Mechanistically, compared to CD8α- dendritic cells, CD8α+ dendritic cells exhibit much stronger oxidative metabolism and critically depend on Mst1/2 signalling to maintain bioenergetic activities and mitochondrial dynamics for their functional capacities. Further, selective expression of IL-12 by CD8α+ dendritic cells depends on Mst1/2 and the crosstalk with non-canonical NF-κB signalling. Our findings identify Mst1/2 as selective drivers of CD8α+ dendritic cell function by integrating metabolic activity and cytokine signalling, and highlight that the interplay between immune signalling and metabolic reprogramming underlies the unique functions of dendritic cell subsets.


Asunto(s)
Antígenos CD8/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Algoritmos , Animales , Antígenos CD8/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/citología , Vía de Señalización Hippo , Homeostasis , Interleucina-12/inmunología , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Serina-Treonina Quinasa 3 , Proteínas Supresoras de Tumor
16.
Nat Immunol ; 12(9): 888-97, 2011 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-21765414

RESUMEN

The mechanisms that regulate T cell quiescence are poorly understood. We report that the tumor suppressor Tsc1 established a quiescence program in naive T cells by controlling cell size, cell cycle entry and responses to stimulation of the T cell antigen receptor. Abrogation of quiescence predisposed Tsc1-deficient T cells to apoptosis that resulted in loss of conventional T cells and invariant natural killer T cells. Loss of Tsc1 function dampened in vivo immune responses to bacterial infection. Tsc1-deficient T cells had more activity of the serine-threonine kinase complex mTORC1 but less mTORC2 activity, and activation of mTORC1 was essential for the disruption of immune homeostasis. Therefore, Tsc1-dependent control of mTOR is crucial in actively maintaining the quiescence of naive T cells to facilitate adaptive immune function.


Asunto(s)
Inmunidad Adaptativa , Linfocitos T CD4-Positivos/inmunología , Proteínas/inmunología , Transducción de Señal/inmunología , Transactivadores/inmunología , Proteínas Supresoras de Tumor , Animales , Apoptosis , Linfocitos T CD4-Positivos/metabolismo , Ciclo Celular/inmunología , Supervivencia Celular/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Homeostasis , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Complejos Multiproteicos , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/inmunología
17.
Nat Immunol ; 12(10): 1010-6, 2011 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-21892172

RESUMEN

The adaptor ASC contributes to innate immunity through the assembly of inflammasome complexes that activate the cysteine protease caspase-1. Here we demonstrate that ASC has an inflammasome-independent, cell-intrinsic role in cells of the adaptive immune response. ASC-deficient mice showed defective antigen presentation by dendritic cells (DCs) and lymphocyte migration due to impaired actin polymerization mediated by the small GTPase Rac. Genome-wide analysis showed that ASC, but not the cytoplasmic receptor NLRP3 or caspase-1, controlled the mRNA stability and expression of Dock2, a guanine nucleotide-exchange factor that mediates Rac-dependent signaling in cells of the immune response. Dock2-deficient DCs showed defective antigen uptake similar to that of ASC-deficient cells. Ectopic expression of Dock2 in ASC-deficient cells restored Rac-mediated actin polymerization, antigen uptake and chemotaxis. Thus, ASC shapes adaptive immunity independently of inflammasomes by modulating Dock2-dependent Rac activation and actin polymerization in DCs and lymphocytes.


Asunto(s)
Actinas/química , Proteínas del Citoesqueleto/fisiología , Proteínas Activadoras de GTPasa/fisiología , Inflamasomas/fisiología , Proteínas de Unión al GTP rac/metabolismo , Actinas/metabolismo , Inmunidad Adaptativa , Animales , Presentación de Antígeno , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Movimiento Celular , Quimiotaxis de Leucocito , Células Dendríticas/inmunología , Proteínas Activadoras de GTPasa/genética , Factores de Intercambio de Guanina Nucleótido , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Polimerizacion , Estabilidad del ARN
18.
Immunity ; 41(6): 947-59, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25500368

RESUMEN

Nonresolving inflammation expands a heterogeneous population of myeloid suppressor cells capable of inhibiting T cell function. This heterogeneity has confounded the functional dissection of individual myeloid subpopulations and presents an obstacle for antitumor immunity and immunotherapy. Using genetic manipulation of cell death pathways, we found the monocytic suppressor-cell subset, but not the granulocytic subset, requires continuous c-FLIP expression to prevent caspase-8-dependent, RIPK3-independent cell death. Development of the granulocyte subset requires MCL-1-mediated control of the intrinsic mitochondrial death pathway. Monocytic suppressors tolerate the absence of MCL-1 provided cytokines increase expression of the MCL-1-related protein A1. Monocytic suppressors mediate T cell suppression, whereas their granulocytic counterparts lack suppressive function. The loss of the granulocytic subset via conditional MCL-1 deletion did not alter tumor incidence implicating the monocytic compartment as the functionally immunosuppressive subset in vivo. Thus, death pathway modulation defines the development, survival, and function of myeloid suppressor cells.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Granulocitos/fisiología , Monocitos/fisiología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Células Mieloides/fisiología , Neoplasias Experimentales/inmunología , Animales , Apoptosis/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Linfocitos T CD8-positivos/inmunología , Carcinogénesis/genética , Caspasa 8/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Técnicas de Cocultivo , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Tolerancia Inmunológica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antígenos de Histocompatibilidad Menor , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/genética
19.
Nature ; 548(7669): 602-606, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28847007

RESUMEN

Regulatory T cells (Treg cells) have a pivotal role in the establishment and maintenance of immunological self-tolerance and homeostasis. Transcriptional programming of regulatory mechanisms facilitates the functional activation of Treg cells in the prevention of diverse types of inflammatory responses. It remains unclear how Treg cells orchestrate their homeostasis and interplay with environmental signals. Here we show that liver kinase B1 (LKB1) programs the metabolic and functional fitness of Treg cells in the control of immune tolerance and homeostasis. Mice with a Treg-specific deletion of LKB1 developed a fatal inflammatory disease characterized by excessive TH2-type-dominant responses. LKB1 deficiency disrupted Treg cell survival and mitochondrial fitness and metabolism, but also induced aberrant expression of immune regulatory molecules including the negative co-receptor PD-1 and the TNF receptor superfamily proteins GITR and OX40. Unexpectedly, LKB1 function in Treg cells was independent of conventional AMPK signalling or the mTORC1-HIF-1α axis, but contributed to the activation of ß-catenin signalling for the control of PD-1 and TNF receptor proteins. Blockade of PD-1 activity reinvigorated the ability of LKB1-deficient Treg cells to suppress TH2 responses and the interplay with dendritic cells primed by thymic stromal lymphopoietin. Thus, Treg cells use LKB1 signalling to coordinate their metabolic and immunological homeostasis and to prevent apoptotic and functional exhaustion, thereby orchestrating the balance between immunity and tolerance.


Asunto(s)
Homeostasis , Tolerancia Inmunológica , Proteínas Serina-Treonina Quinasas/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Proteínas Quinasas Activadas por AMP , Animales , Apoptosis , Supervivencia Celular/genética , Citocinas/metabolismo , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/biosíntesis , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Receptores OX40/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Linfocitos T Reguladores/citología , Células Th2/inmunología , beta Catenina/metabolismo , Linfopoyetina del Estroma Tímico
20.
PLoS Pathog ; 16(3): e1008364, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32150572

RESUMEN

Innate immunity responds to pathogens by producing alarm signals and activating pathways that make host cells inhospitable for pathogen replication. The intracellular bacterium Burkholderia thailandensis invades the cytosol, hijacks host actin, and induces cell fusion to spread to adjacent cells, forming multinucleated giant cells (MNGCs) which promote bacterial replication. We show that type I interferon (IFN) restricts macrophage MNGC formation during B. thailandensis infection. Guanylate-binding proteins (GBPs) expressed downstream of type I IFN were required to restrict MNGC formation through inhibition of bacterial Arp2/3-dependent actin motility during infection. GTPase activity and the CAAX prenylation domain were required for GBP2 recruitment to B. thailandensis, which restricted bacterial actin polymerization required for MNGC formation. Consistent with the effects in in vitro macrophages, Gbp2-/-, Gbp5-/-, GbpChr3-KO mice were more susceptible to intranasal infection with B. thailandensis than wildtype mice. Our findings reveal that IFN and GBPs play a critical role in restricting cell-cell fusion and bacteria-induced pathology during infection.


Asunto(s)
Infecciones por Burkholderia/inmunología , Burkholderia/inmunología , Proteínas de Unión al GTP/inmunología , Células Gigantes/inmunología , Macrófagos/inmunología , Enfermedades Nasales/inmunología , Prenilación de Proteína/inmunología , Animales , Infecciones por Burkholderia/genética , Infecciones por Burkholderia/patología , Fusión Celular , Proteínas de Unión al GTP/genética , Células Gigantes/microbiología , Células Gigantes/patología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Enfermedades Nasales/genética , Enfermedades Nasales/microbiología , Enfermedades Nasales/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA