Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29.698
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 184(10): 2665-2679.e19, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33882274

RESUMEN

The bacterial flagellar motor is a supramolecular protein machine that drives rotation of the flagellum for motility, which is essential for bacterial survival in different environments and a key determinant of pathogenicity. The detailed structure of the flagellar motor remains unknown. Here we present an atomic-resolution cryoelectron microscopy (cryo-EM) structure of the bacterial flagellar motor complexed with the hook, consisting of 175 subunits with a molecular mass of approximately 6.3 MDa. The structure reveals that 10 peptides protruding from the MS ring with the FlgB and FliE subunits mediate torque transmission from the MS ring to the rod and overcome the symmetry mismatch between the rotational and helical structures in the motor. The LP ring contacts the distal rod and applies electrostatic forces to support its rotation and torque transmission to the hook. This work provides detailed molecular insights into the structure, assembly, and torque transmission mechanisms of the flagellar motor.


Asunto(s)
Flagelos/fisiología , Flagelos/ultraestructura , Salmonella typhimurium/fisiología , Microscopía por Crioelectrón , Conformación Proteica , Torque
2.
Nat Immunol ; 24(9): 1473-1486, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37580603

RESUMEN

Omnivorous animals, including mice and humans, tend to prefer energy-dense nutrients rich in fat over plant-based diets, especially for short periods of time, but the health consequences of this short-term consumption of energy-dense nutrients are unclear. Here, we show that short-term reiterative switching to 'feast diets', mimicking our social eating behavior, breaches the potential buffering effect of the intestinal microbiota and reorganizes the immunological architecture of mucosa-associated lymphoid tissues. The first dietary switch was sufficient to induce transient mucosal immune depression and suppress systemic immunity, leading to higher susceptibility to Salmonella enterica serovar Typhimurium and Listeria monocytogenes infections. The ability to respond to antigenic challenges with a model antigen was also impaired. These observations could be explained by a reduction of CD4+ T cell metabolic fitness and cytokine production due to impaired mTOR activity in response to reduced microbial provision of fiber metabolites. Reintroducing dietary fiber rewired T cell metabolism and restored mucosal and systemic CD4+ T cell functions and immunity. Finally, dietary intervention with human volunteers confirmed the effect of short-term dietary switches on human CD4+ T cell functionality. Therefore, short-term nutritional changes cause a transient depression of mucosal and systemic immunity, creating a window of opportunity for pathogenic infection.


Asunto(s)
Membrana Mucosa , Salmonella typhimurium , Humanos , Ratones , Animales , Linfocitos T , Inmunidad Mucosa
3.
Annu Rev Immunol ; 31: 73-106, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23215645

RESUMEN

Inflammasomes are cytosolic multiprotein complexes that assemble in response to a variety of infectious and noxious insults. Inflammasomes play a critical role in the initiation of innate immune responses, primarily by serving as platforms for the activation of inflammatory caspase proteases. One such caspase, CASPASE-1 (CASP1), initiates innate immune responses by cleaving pro-IL-1ß and pro-IL-18, leading to their activation and release. CASP1 and another inflammatory caspase termed CASP11 can also initiate a rapid and inflammatory form of cell death termed pyroptosis. Several distinct inflammasomes have been described, each of which contains a unique sensor protein of the NLR (nucleotide-binding domain, leucine-rich repeat-containing) superfamily or the PYHIN (PYRIN and HIN-200 domain-containing) superfamily. Here we describe the surprisingly diverse mechanisms by which NLR/PYHIN proteins sense bacteria and initiate innate immune responses. We conclude that inflammasomes represent a highly adaptable scaffold ideally suited for detecting and initiating rapid innate responses to diverse and rapidly evolving bacteria.


Asunto(s)
Bacterias/patogenicidad , Inflamasomas/metabolismo , Animales , Bacillus anthracis/patogenicidad , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas Adaptadoras de Señalización CARD/fisiología , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/fisiología , Flagelos/metabolismo , Flagelos/fisiología , Humanos , Inflamasomas/genética , Inflamasomas/fisiología , Legionella pneumophila/patogenicidad , Listeria monocytogenes/patogenicidad , Salmonella typhimurium/patogenicidad
4.
Cell ; 180(1): 33-49.e22, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31813624

RESUMEN

Gut-innervating nociceptor sensory neurons respond to noxious stimuli by initiating protective responses including pain and inflammation; however, their role in enteric infections is unclear. Here, we find that nociceptor neurons critically mediate host defense against the bacterial pathogen Salmonella enterica serovar Typhimurium (STm). Dorsal root ganglia nociceptors protect against STm colonization, invasion, and dissemination from the gut. Nociceptors regulate the density of microfold (M) cells in ileum Peyer's patch (PP) follicle-associated epithelia (FAE) to limit entry points for STm invasion. Downstream of M cells, nociceptors maintain levels of segmentous filamentous bacteria (SFB), a gut microbe residing on ileum villi and PP FAE that mediates resistance to STm infection. TRPV1+ nociceptors directly respond to STm by releasing calcitonin gene-related peptide (CGRP), a neuropeptide that modulates M cells and SFB levels to protect against Salmonella infection. These findings reveal a major role for nociceptor neurons in sensing and defending against enteric pathogens.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Interacciones Microbiota-Huesped/fisiología , Nociceptores/fisiología , Animales , Epitelio/metabolismo , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/microbiología , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Nociceptores/metabolismo , Ganglios Linfáticos Agregados/inervación , Ganglios Linfáticos Agregados/metabolismo , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología
5.
Cell ; 180(1): 50-63.e12, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31923399

RESUMEN

Mucosal barrier immunity is essential for the maintenance of the commensal microflora and combating invasive bacterial infection. Although immune and epithelial cells are thought to be the canonical orchestrators of this complex equilibrium, here, we show that the enteric nervous system (ENS) plays an essential and non-redundant role in governing the antimicrobial protein (AMP) response. Using confocal microscopy and single-molecule fluorescence in situ mRNA hybridization (smFISH) studies, we observed that intestinal neurons produce the pleiotropic cytokine IL-18. Strikingly, deletion of IL-18 from the enteric neurons alone, but not immune or epithelial cells, rendered mice susceptible to invasive Salmonella typhimurium (S.t.) infection. Mechanistically, unbiased RNA sequencing and single-cell sequencing revealed that enteric neuronal IL-18 is specifically required for homeostatic goblet cell AMP production. Together, we show that neuron-derived IL-18 signaling controls tissue-wide intestinal immunity and has profound consequences on the mucosal barrier and invasive bacterial killing.


Asunto(s)
Inmunidad Mucosa/inmunología , Interleucina-18/inmunología , Mucosa Intestinal/inmunología , Animales , Citocinas/inmunología , Sistema Nervioso Entérico/inmunología , Sistema Nervioso Entérico/metabolismo , Células Epiteliales/inmunología , Femenino , Células Caliciformes/inmunología , Interleucina-18/biosíntesis , Mucosa Intestinal/metabolismo , Intestino Delgado/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/inmunología , Ratas , Ratas Sprague-Dawley , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Transducción de Señal/inmunología
6.
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
7.
Cell ; 168(3): 503-516.e12, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28129542

RESUMEN

Sickness-induced anorexia is a conserved behavior induced during infections. Here, we report that an intestinal pathogen, Salmonella Typhimurium, inhibits anorexia by manipulating the gut-brain axis. Inhibition of inflammasome activation by the S. Typhimurium effector, SlrP, prevented anorexia caused by IL-1ß-mediated signaling to the hypothalamus via the vagus nerve. Rather than compromising host defenses, pathogen-mediated inhibition of anorexia increased host survival. SlrP-mediated inhibition of anorexia prevented invasion and systemic infection by wild-type S. Typhimurium, reducing virulence while increasing transmission to new hosts, suggesting that there are trade-offs between transmission and virulence. These results clarify the complex and contextual role of anorexia in host-pathogen interactions and suggest that microbes have evolved mechanisms to modulate sickness-induced behaviors to promote health of their host and their transmission at the expense of virulence.


Asunto(s)
Anorexia/microbiología , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/transmisión , Salmonella typhimurium/patogenicidad , Animales , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inflamasomas/inmunología , Interleucina-1beta/inmunología , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas , Infecciones por Salmonella/inmunología , Salmonella typhimurium/fisiología , Organismos Libres de Patógenos Específicos , Virulencia
8.
Cell ; 168(6): 1065-1074.e10, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28283062

RESUMEN

Type III protein secretion systems have specifically evolved to deliver bacterially encoded proteins into target eukaryotic cells. The core elements of this multi-protein machine are the envelope-associated needle complex, the inner membrane export apparatus, and a large cytoplasmic sorting platform. Here, we report a high-resolution in situ structure of the Salmonella Typhimurium type III secretion machine obtained by high-throughput cryo-electron tomography and sub-tomogram averaging. Through molecular modeling and comparative analysis of machines assembled with protein-tagged components or from different deletion mutants, we determined the molecular architecture of the secretion machine in situ and localized its structural components. We also show that docking of the sorting platform results in significant conformational changes in the needle complex to provide the symmetry adaptation required for the assembly of the entire secretion machine. These studies provide major insight into the structure and assembly of a broadly distributed protein secretion machine.


Asunto(s)
Sistemas de Secreción Bacterianos/ultraestructura , Salmonella typhimurium/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/genética , Microscopía por Crioelectrón , Transporte de Proteínas , Virulencia
9.
Cell ; 171(4): 783-794.e13, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28942917

RESUMEN

Intestinal intraepithelial lymphocytes (IELs) are located at the critical interface between the intestinal lumen, which is chronically exposed to food and microbes, and the core of the body. Using high-resolution microscopy techniques and intersectional genetic tools, we investigated the nature of IEL responses to luminal microbes. We observed that TCRγδ IELs exhibit unique microbiota-dependent location and movement patterns in the epithelial compartment. This behavioral pattern quickly changes upon exposure to different enteric pathogens, resulting in increased interepithelial cell (EC) scanning, expression of antimicrobial genes, and glycolysis. Both dynamic and metabolic changes to γδ IEL depend on pathogen sensing by ECs. Direct modulation of glycolysis is sufficient to change γδ IEL behavior and susceptibility to early pathogen invasion. Our results uncover a coordinated EC-IEL response to enteric infections that modulates lymphocyte energy utilization and dynamics and supports maintenance of the intestinal epithelial barrier. VIDEO ABSTRACT.


Asunto(s)
Intestinos/citología , Intestinos/inmunología , Infecciones por Salmonella/inmunología , Linfocitos T/inmunología , Animales , Células Epiteliales/metabolismo , Vigilancia Inmunológica , Mucosa Intestinal/inmunología , Ratones , Infecciones por Salmonella/microbiología , Salmonella typhimurium/fisiología
10.
Cell ; 164(3): 378-91, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26777404

RESUMEN

Proper adaptation to environmental perturbations is essential for tissue homeostasis. In the intestine, diverse environmental cues can be sensed by immune cells, which must balance resistance to microorganisms with tolerance, avoiding excess tissue damage. By applying imaging and transcriptional profiling tools, we interrogated how distinct microenvironments in the gut regulate resident macrophages. We discovered that macrophages exhibit a high degree of gene-expression specialization dependent on their proximity to the gut lumen. Lamina propria macrophages (LpMs) preferentially expressed a pro-inflammatory phenotype when compared to muscularis macrophages (MMs), which displayed a tissue-protective phenotype. Upon luminal bacterial infection, MMs further enhanced tissue-protective programs, and this was attributed to swift activation of extrinsic sympathetic neurons innervating the gut muscularis and norepinephrine signaling to ß2 adrenergic receptors on MMs. Our results reveal unique intra-tissue macrophage specialization and identify neuro-immune communication between enteric neurons and macrophages that induces rapid tissue-protective responses to distal perturbations.


Asunto(s)
Intestino Delgado/fisiología , Macrófagos/inmunología , Neuronas/citología , Animales , Línea Celular , Mucosa Intestinal/citología , Mucosa Intestinal/fisiología , Intestino Delgado/citología , Intestino Delgado/inmunología , Macrófagos/citología , Ratones , Membrana Mucosa/citología , Membrana Mucosa/fisiología , Neuroinmunomodulación , Neuronas/fisiología , Receptores Adrenérgicos beta 2/metabolismo , Infecciones por Salmonella/inmunología , Salmonella typhimurium/fisiología , Organismos Libres de Patógenos Específicos
11.
Cell ; 167(2): 444-456.e14, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27716507

RESUMEN

While conventional pathogenic protists have been extensively studied, there is an underappreciated constitutive protist microbiota that is an integral part of the vertebrate microbiome. The impact of these species on the host and their potential contributions to mucosal immune homeostasis remain poorly studied. Here, we show that the protozoan Tritrichomonas musculis activates the host epithelial inflammasome to induce IL-18 release. Epithelial-derived IL-18 promotes dendritic cell-driven Th1 and Th17 immunity and confers dramatic protection from mucosal bacterial infections. Along with its role as a "protistic" antibiotic, colonization with T. musculis exacerbates the development of T-cell-driven colitis and sporadic colorectal tumors. Our findings demonstrate a novel mutualistic host-protozoan interaction that increases mucosal host defenses at the cost of an increased risk of inflammatory disease.


Asunto(s)
Colitis/inmunología , Colitis/parasitología , Interacciones Huésped-Parásitos , Inflamasomas/inmunología , Mucosa Intestinal/parasitología , Microbiota/inmunología , Tricomoniasis/inmunología , Trichomonas/inmunología , Animales , Colitis/microbiología , Dientamoeba/inmunología , Inmunidad Mucosa , Interleucina-18/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Simbiosis , Células TH1/inmunología , Células Th17/inmunología
12.
Immunity ; 54(12): 2712-2723.e6, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34788598

RESUMEN

Interactions between intracellular bacteria and mononuclear phagocytes give rise to diverse cellular phenotypes that may determine the outcome of infection. Recent advances in single-cell RNA sequencing (scRNA-seq) have identified multiple subsets within the mononuclear population, but implications to their function during infection are limited. Here, we surveyed the mononuclear niche of intracellular Salmonella Typhimurium (S.Tm) during early systemic infection in mice. We described eclipse-like growth kinetics in the spleen, with a first phase of bacterial control mediated by tissue-resident red-pulp macrophages. A second phase involved extensive bacterial replication within a macrophage population characterized by CD9 expression. We demonstrated that CD9+ macrophages induced pathways for detoxificating oxidized lipids, that may be utilized by intracellular S.Tm. We established that CD9+ macrophages originated from non-classical monocytes (NCM), and NCM-depleted mice were more resistant to S.Tm infection. Our study defines macrophage subset-specific host-pathogen interactions that determine early infection dynamics and infection outcome of the entire organism.


Asunto(s)
Macrófagos/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/fisiología , Bazo/inmunología , Animales , Interacciones Huésped-Patógeno , Humanos , Espacio Intracelular , Metabolismo de los Lípidos , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxidación-Reducción , Análisis de la Célula Individual , Bazo/microbiología , Tetraspanina 29/metabolismo
13.
Cell ; 162(6): 1208-10, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26359980

RESUMEN

Using single-cell RNA sequencing, Avraham et al. investigate how variability in macrophage response to infection is controlled by variability within the pathogen population. They find that heterogeneous expression of the Salmonella virulence factor PhoP and subsequent cell-wall modifications lead to the bimodal induction of the interferon-response in infected macrophages.


Asunto(s)
Interacciones Huésped-Patógeno , Macrófagos/inmunología , Salmonella typhimurium/fisiología , Animales
14.
Cell ; 162(6): 1309-21, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26343579

RESUMEN

Encounters between immune cells and invading bacteria ultimately determine the course of infection. These interactions are usually measured in populations of cells, masking cell-to-cell variation that may be important for infection outcome. To characterize the gene expression variation that underlies distinct infection outcomes and monitor infection phenotypes, we developed an experimental system that combines single-cell RNA-seq with fluorescent markers. Probing the responses of individual macrophages to invading Salmonella, we find that variation between individual infected host cells is determined by the heterogeneous activity of bacterial factors in individual infecting bacteria. We illustrate how variable PhoPQ activity in the population of invading bacteria drives variable host type I IFN responses by modifying LPS in a subset of bacteria. This work demonstrates a causative link between host and bacterial variability, with cell-to-cell variation between different bacteria being sufficient to drive radically different host immune responses. This co-variation has implications for host-pathogen dynamics in vivo.


Asunto(s)
Interacciones Huésped-Patógeno , Macrófagos/inmunología , Salmonella typhimurium/fisiología , Animales , Interferón Tipo I/inmunología , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/microbiología , Organismos Libres de Patógenos Específicos
15.
Cell ; 158(4): 722-733, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25126781

RESUMEN

Antibiotic therapy often fails to eliminate a fraction of transiently refractory bacteria, causing relapses and chronic infections. Multiple mechanisms can induce such persisters with high antimicrobial tolerance in vitro, but their in vivo relevance remains unclear. Using a fluorescent growth rate reporter, we detected extensive phenotypic variation of Salmonella in host tissues. This included slow-growing subsets as well as well-nourished fast-growing subsets driving disease progression. Monitoring of Salmonella growth and survival during chemotherapy revealed that antibiotic killing correlated with single-cell division rates. Nondividing Salmonella survived best but were rare, limiting their impact. Instead, most survivors originated from abundant moderately growing, partially tolerant Salmonella. These data demonstrate that host tissues diversify pathogen physiology, with major consequences for disease progression and control.


Asunto(s)
Antibacterianos/administración & dosificación , Fluoroquinolonas/administración & dosificación , Imagen Óptica/métodos , Salmonella typhimurium/efectos de los fármacos , Fiebre Tifoidea/tratamiento farmacológico , Fiebre Tifoidea/microbiología , Animales , Proteínas Bacterianas/análisis , Enrofloxacina , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Proteoma/análisis , Salmonella typhimurium/citología , Salmonella typhimurium/crecimiento & desarrollo , Bazo/microbiología , Bazo/patología
16.
Nat Immunol ; 17(4): 441-50, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26855029

RESUMEN

Epithelial tissues continually undergo apoptosis. Commensal organisms that inhabit the epithelium influence tissue homeostasis, in which regulatory T cells (Treg cells) have a central role. However, the physiological importance of epithelial cell apoptosis and how the number of Treg cells is regulated are both incompletely understood. Here we found that apoptotic epithelial cells negatively regulated the commensal-stimulated proliferation of Treg cells. Gut commensals stimulated CX3CR1(+)CD103(-)CD11b(+) dendritic cells (DCs) to produce interferon-ß (IFN-ß), which augmented the proliferation of Treg cells in the intestine. Conversely, phosphatidylserine exposed on apoptotic epithelial cells suppressed IFN-ß production by the DCs via inhibitory signaling mediated by the cell-surface glycoprotein CD300a and thus suppressed Treg cell proliferation. Our findings reveal a regulatory role for apoptotic epithelial cells in maintaining the number of Treg cell and tissue homeostasis.


Asunto(s)
Apoptosis/inmunología , Epidermis/inmunología , Células Epiteliales/inmunología , Microbioma Gastrointestinal/inmunología , Interferón beta/inmunología , Mucosa Intestinal/inmunología , Mucosa Respiratoria/inmunología , Linfocitos T Reguladores/inmunología , Alérgenos/toxicidad , Animales , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Colon/citología , Colon/inmunología , Células Dendríticas/inmunología , Dermatitis Alérgica por Contacto/etiología , Dermatitis Alérgica por Contacto/inmunología , Dermatitis Alérgica por Contacto/patología , Sulfato de Dextran/toxicidad , Células Epidérmicas , Citometría de Flujo , Inmunohistoquímica , Mucosa Intestinal/citología , Células de Langerhans/inmunología , Pulmón/citología , Pulmón/inmunología , Ratones , Ratones Noqueados , Ovalbúmina/toxicidad , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Inmunológicos/genética , Mucosa Respiratoria/citología , Infecciones por Salmonella/inmunología , Salmonella typhimurium
17.
Immunity ; 50(1): 137-151.e6, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30650373

RESUMEN

Fever is an evolutionarily conserved response that confers survival benefits during infection. However, the underlying mechanism remains obscure. Here, we report that fever promoted T lymphocyte trafficking through heat shock protein 90 (Hsp90)-induced α4 integrin activation and signaling in T cells. By inducing selective binding of Hsp90 to α4 integrins, but not ß2 integrins, fever increased α4-integrin-mediated T cell adhesion and transmigration. Mechanistically, Hsp90 bound to the α4 tail and activated α4 integrins via inside-out signaling. Moreover, the N and C termini of one Hsp90 molecule simultaneously bound to two α4 tails, leading to dimerization and clustering of α4 integrins on the cell membrane and subsequent activation of the FAK-RhoA pathway. Abolishment of Hsp90-α4 interaction inhibited fever-induced T cell trafficking to draining lymph nodes and impaired the clearance of bacterial infection. Our findings identify the Hsp90-α4-integrin axis as a thermal sensory pathway that promotes T lymphocyte trafficking and enhances immune surveillance during infection.


Asunto(s)
Fiebre/inmunología , Proteínas HSP90 de Choque Térmico/metabolismo , Integrina alfa4/metabolismo , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Linfocitos T/inmunología , Animales , Carga Bacteriana , Adhesión Celular , Movimiento Celular , Dimerización , Quinasa 1 de Adhesión Focal/metabolismo , Vigilancia Inmunológica , Integrina alfa4/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Transducción de Señal , Proteína de Unión al GTP rhoA/metabolismo
18.
Cell ; 154(1): 146-56, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23827679

RESUMEN

Several intracellular pathogens, including Salmonella enterica and Mycobacterium tuberculosis, require the virulence protein MgtC to survive within macrophages and to cause a lethal infection in mice. We now report that, unlike secreted virulence factors that target the host vacuolar ATPase to withstand phagosomal acidity, the MgtC protein acts on Salmonella's own F1Fo ATP synthase. This complex couples proton translocation to ATP synthesis/hydrolysis and is required for virulence. We establish that MgtC interacts with the a subunit of the F1Fo ATP synthase, hindering ATP-driven proton translocation and NADH-driven ATP synthesis in inverted vesicles. An mgtC null mutant displays heightened ATP levels and an acidic cytoplasm, whereas mgtC overexpression decreases ATP levels. A single amino acid substitution in MgtC that prevents binding to the F1Fo ATP synthase abolishes control of ATP levels and attenuates pathogenicity. MgtC provides a singular example of a virulence protein that promotes pathogenicity by interfering with another virulence protein.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Transporte de Catión/metabolismo , ATPasas de Translocación de Protón/antagonistas & inhibidores , Infecciones por Salmonella/microbiología , Salmonella typhimurium/citología , Salmonella typhimurium/patogenicidad , Factores de Virulencia/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Femenino , Concentración de Iones de Hidrógeno , Macrófagos/microbiología , Potenciales de la Membrana , Ratones , Ratones Endogámicos C3H , Subunidades de Proteína/antagonistas & inhibidores , Salmonella typhimurium/enzimología , Virulencia
19.
Cell ; 153(2): 426-37, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23582330

RESUMEN

Glucose homeostasis is strictly controlled in all domains of life. Bacteria that are unable to balance intracellular sugar levels and deal with potentially toxic phosphosugars cease growth and risk being outcompeted. Here, we identify the conserved haloacid dehalogenase (HAD)-like enzyme YigL as the previously hypothesized phosphatase for detoxification of phosphosugars and reveal that its synthesis is activated by an Hfq-dependent small RNA in Salmonella typhimurium. We show that the glucose-6-P-responsive small RNA SgrS activates YigL synthesis in a translation-independent fashion by the selective stabilization of a decay intermediate of the dicistronic pldB-yigL messenger RNA (mRNA). Intriguingly, the major endoribonuclease RNase E, previously known to function together with small RNAs to degrade mRNA targets, is also essential for this process of mRNA activation. The exploitation of and targeted interference with regular RNA turnover described here may constitute a general route for small RNAs to rapidly activate both coding and noncoding genes.


Asunto(s)
Glucosa/metabolismo , Hidrolasas/genética , ARN Bacteriano/metabolismo , ARN Pequeño no Traducido/metabolismo , Salmonella typhimurium/metabolismo , Secuencia de Bases , Escherichia coli/enzimología , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Hidrolasas/metabolismo , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/metabolismo , Operón , Monoéster Fosfórico Hidrolasas/genética , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Salmonella typhimurium/enzimología , Salmonella typhimurium/genética
20.
Cell ; 152(4): 743-54, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23415224

RESUMEN

Long noncoding RNAs (lncRNAs) are increasingly appreciated as regulators of cell-specific gene expression. Here, an enhancer-like lncRNA termed NeST (nettoie Salmonella pas Theiler's [cleanup Salmonella not Theiler's]) is shown to be causal for all phenotypes conferred by murine viral susceptibility locus Tmevp3. This locus was defined by crosses between SJL/J and B10.S mice and contains several candidate genes, including NeST. The SJL/J-derived locus confers higher lncRNA expression, increased interferon-γ (IFN-γ) abundance in activated CD8(+) T cells, increased Theiler's virus persistence, and decreased Salmonella enterica pathogenesis. Transgenic expression of NeST lncRNA alone was sufficient to confer all phenotypes of the SJL/J locus. NeST RNA was found to bind WDR5, a component of the histone H3 lysine 4 methyltransferase complex, and to alter histone 3 methylation at the IFN-γ locus. Thus, this lncRNA regulates epigenetic marking of IFN-γ-encoding chromatin, expression of IFN-γ, and susceptibility to a viral and a bacterial pathogen.


Asunto(s)
Susceptibilidad a Enfermedades , Epigénesis Genética , Interferón gamma/genética , ARN Largo no Codificante/genética , Animales , Linfocitos T CD8-positivos/inmunología , Infecciones por Cardiovirus/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Theilovirus/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA