Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.572
Filtrar
Más filtros

Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
PLoS Biol ; 22(6): e3002616, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38865418

RESUMEN

The gastrointestinal tract is densely colonized by a polymicrobial community known as the microbiota which serves as primary line of defence against pathogen invasion. The microbiota can limit gut-luminal pathogen growth at different stages of infection. This can be traced to specific commensal strains exhibiting direct or indirect protective functions. Although these mechanisms hold the potential to develop new approaches to combat enteric pathogens, they remain far from being completely described. In this study, we investigated how a mouse commensal Escherichia coli can outcompete Salmonella enterica serovar Typhimurium (S. Tm). Using a salmonellosis mouse model, we found that the commensal E. coli 8178 strain relies on a trojan horse trap strategy to limit S. Tm expansion in the inflamed gut. Combining mutants and reporter tools, we demonstrated that inflammation triggers the expression of the E. coli 8178 antimicrobial microcin H47 toxin which, when fused to salmochelin siderophores, can specifically alter S. Tm growth. This protective function was compromised upon disruption of the E. coli 8178 tonB-dependent catecholate siderophore uptake system, highlighting a previously unappreciated crosstalk between iron intake and microcin H47 activity. By identifying the genetic determinants mediating S. Tm competition, our work not only provides a better mechanistic understanding of the protective function displayed by members of the gut microbiota but also further expands the general contribution of microcins in bacterial antagonistic relationships. Ultimately, such insights can open new avenues for developing microbiota-based approaches to better control intestinal infections.


Asunto(s)
Escherichia coli , Inflamación , Salmonella typhimurium , Sideróforos , Animales , Escherichia coli/metabolismo , Escherichia coli/genética , Sideróforos/metabolismo , Ratones , Salmonella typhimurium/patogenicidad , Salmonella typhimurium/metabolismo , Inflamación/metabolismo , Inflamación/microbiología , Ratones Endogámicos C57BL , Bacteriocinas/metabolismo , Bacteriocinas/farmacología , Microbioma Gastrointestinal , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/metabolismo , Femenino , Hierro/metabolismo , Simbiosis , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/metabolismo
7.
PLoS Biol ; 22(4): e3002597, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38684033

RESUMEN

Intestinal epithelial cells (IECs) play pivotal roles in nutrient uptake and in the protection against gut microorganisms. However, certain enteric pathogens, such as Salmonella enterica serovar Typhimurium (S. Tm), can invade IECs by employing flagella and type III secretion systems (T3SSs) with cognate effector proteins and exploit IECs as a replicative niche. Detection of flagella or T3SS proteins by IECs results in rapid host cell responses, i.e., the activation of inflammasomes. Here, we introduce a single-cell manipulation technology based on fluidic force microscopy (FluidFM) that enables direct bacteria delivery into the cytosol of single IECs within a murine enteroid monolayer. This approach allows to specifically study pathogen-host cell interactions in the cytosol uncoupled from preceding events such as docking, initiation of uptake, or vacuole escape. Consistent with current understanding, we show using a live-cell inflammasome reporter that exposure of the IEC cytosol to S. Tm induces NAIP/NLRC4 inflammasomes via its known ligands flagellin and T3SS rod and needle. Injected S. Tm mutants devoid of these invasion-relevant ligands were able to grow in the cytosol of IECs despite the absence of T3SS functions, suggesting that, in the absence of NAIP/NLRC4 inflammasome activation and the ensuing cell death, no effector-mediated host cell manipulation is required to render the epithelial cytosol growth-permissive for S. Tm. Overall, the experimental system to introduce S. Tm into single enteroid cells enables investigations into the molecular basis governing host-pathogen interactions in the cytosol with high spatiotemporal resolution.


Asunto(s)
Proteínas de Unión al Calcio , Citosol , Flagelina , Interacciones Huésped-Patógeno , Inflamasomas , Salmonella typhimurium , Sistemas de Secreción Tipo III , Citosol/metabolismo , Citosol/microbiología , Animales , Salmonella typhimurium/patogenicidad , Salmonella typhimurium/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Inflamasomas/metabolismo , Ratones , Flagelina/metabolismo , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Proteína Inhibidora de la Apoptosis Neuronal/genética , Células Epiteliales/microbiología , Células Epiteliales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Ratones Endogámicos C57BL , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas Adaptadoras de Señalización CARD/genética , Análisis de la Célula Individual/métodos , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo
8.
Mol Cell ; 74(2): 320-329.e6, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30853402

RESUMEN

Xenophagy, a selective autophagy pathway that protects the cytosol against bacterial invasion, relies on cargo receptors that juxtapose bacteria and phagophore membranes. Whether phagophores are recruited from a constitutive pool or are generated de novo at prospective cargo remains unknown. Phagophore formation in situ would require recruitment of the upstream autophagy machinery to prospective cargo. Here, we show that, essential for anti-bacterial autophagy, the cargo receptor NDP52 forms a trimeric complex with FIP200 and SINTBAD/NAP1, which are subunits of the autophagy-initiating ULK and the TBK1 kinase complex, respectively. FIP200 and SINTBAD/NAP1 are each recruited independently to bacteria via NDP52, as revealed by selective point mutations in their respective binding sites, but only in their combined presence does xenophagy proceed. Such recruitment of the upstream autophagy machinery by NDP52 reveals how detection of cargo-associated "eat me" signals, induction of autophagy, and juxtaposition of cargo and phagophores are integrated in higher eukaryotes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Autofagia/genética , Proteínas Nucleares/genética , Proteínas Tirosina Quinasas/genética , Proteínas Adaptadoras Transductoras de Señales/química , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Proteínas Relacionadas con la Autofagia , Sitios de Unión/genética , Citoplasma/microbiología , Citosol/microbiología , Humanos , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Proteínas Nucleares/química , Mutación Puntual/genética , Unión Proteica/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/química , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidad
9.
Cell ; 144(5): 675-88, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21376231

RESUMEN

Toll-like receptors (TLRs) contribute to host resistance to microbial pathogens and can drive the evolution of virulence mechanisms. We have examined the relationship between host resistance and pathogen virulence using mice with a functional allele of the nramp-1 gene and lacking combinations of TLRs. Mice deficient in both TLR2 and TLR4 were highly susceptible to the intracellular bacterial pathogen Salmonella typhimurium, consistent with reduced innate immune function. However, mice lacking additional TLRs involved in S. typhimurium recognition were less susceptible to infection. In these TLR-deficient cells, bacteria failed to upregulate Salmonella pathogenicity island 2 (SPI-2) genes and did not form a replicative compartment. We demonstrate that TLR signaling enhances the rate of acidification of the Salmonella-containing phagosome, and inhibition of this acidification prevents SPI-2 induction. Our results indicate that S. typhimurium requires cues from the innate immune system to regulate virulence genes necessary for intracellular survival, growth, and systemic infection.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Innata , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Transducción de Señal , Receptores Toll-Like/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Humanos , Macrófagos/inmunología , Macrófagos/microbiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Receptores Toll-Like/inmunología
10.
Annu Rev Microbiol ; 74: 787-813, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32692613

RESUMEN

Food has a major impact on all aspects of health. Recent data suggest that food composition can also affect susceptibility to infections by enteropathogenic bacteria. Here, we discuss how food may alter the microbiota as well as mucosal defenses and how this can affect infection. Salmonella Typhimurium diarrhea serves as a paradigm, and complementary evidence comes from other pathogens. We discuss the effects of food composition on colonization resistance, host defenses, and the infection process as well as the merits and limitations of mouse models and experimental foods, which are available to decipher the underlying mechanisms.


Asunto(s)
Dieta , Enterobacteriaceae/patogenicidad , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Infecciones por Salmonella/prevención & control , Animales , Diarrea/microbiología , Diarrea/prevención & control , Modelos Animales de Enfermedad , Análisis de los Alimentos , Humanos , Ratones , Infecciones por Salmonella/microbiología , Salmonella typhimurium/patogenicidad
11.
J Biol Chem ; 299(12): 105387, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37890783

RESUMEN

The expression of virulence factors essential for the invasion of host cells by Salmonella enterica is tightly controlled by a network of transcription regulators. The AraC/XylS transcription factor HilD is the main integration point of environmental signals into this regulatory network, with many factors affecting HilD activity. Long-chain fatty acids, which are highly abundant throughout the host intestine, directly bind to and repress HilD, acting as environmental cues to coordinate virulence gene expression. The regulatory protein HilE also negatively regulates HilD activity, through a protein-protein interaction. Both of these regulators inhibit HilD dimerization, preventing HilD from binding to target DNA. We investigated the structural basis of these mechanisms of HilD repression. Long-chain fatty acids bind to a conserved pocket in HilD, in a comparable manner to that reported for other AraC/XylS regulators, whereas HilE forms a stable heterodimer with HilD by binding to the HilD dimerization interface. Our results highlight two distinct, mutually exclusive mechanisms by which HilD activity is repressed, which could be exploited for the development of new antivirulence leads.


Asunto(s)
Proteínas Bacterianas , Intestinos , Salmonella typhimurium , Proteínas Bacterianas/metabolismo , Ácidos Grasos/metabolismo , Regulación Bacteriana de la Expresión Génica , Intestinos/metabolismo , Intestinos/microbiología , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Virulencia , Animales , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/microbiología
12.
J Cell Physiol ; 239(1): 36-50, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37877586

RESUMEN

Human enterocytes are primary targets of infection by invasive bacterium Salmonella Typhimurium, and studies using nonintestinal epithelial cells established that S. Typhimurium activates Rho family GTPases, primarily CDC42, to modulate the actin cytoskeletal network for invasion. The host intracellular protein network that engages CDC42 and influences the pathogen's invasive capacity are relatively unclear. Here, proteomic analyses of canonical and variant CDC42 interactomes identified a poorly characterized CDC42 interacting protein, CDC42EP1, whose intracellular localization is rapidly redistributed and aggregated around the invading bacteria. CDC42EP1 associates with SEPTIN-7 and Villin, and its relocalization and bacterial engagement depend on host CDC42 and S. Typhimurium's capability of activating CDC42. Unlike CDC42, CDC42EP1 is not required for S. Typhimurium's initial cellular entry but is found to associate with Salmonella-containing vacuoles after long-term infections, indicating a contribution to the pathogen's intracellular growth and replication. These results uncover a new host regulator of enteric Salmonella infections, which may be targeted to restrict bacterial load at the primary site of infection to prevent systemic spread.


Asunto(s)
Proteínas del Citoesqueleto , Salmonella typhimurium , Proteínas de Unión al GTP rho , Humanos , Actinas/metabolismo , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Citoesqueleto/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Salmonella typhimurium/patogenicidad , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo
13.
PLoS Pathog ; 18(1): e1009718, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35073381

RESUMEN

Salmonella enterica serovar Typhimurium is a Gram-negative pathogen that uses two distinct type III secretion systems (T3SSs), termed Salmonella pathogenicity island (SPI)-1 and SPI-2, to deliver virulence factors into the host cell. The SPI-1 T3SS enables Salmonella to invade host cells, while the SPI-2 T3SS facilitates Salmonella's intracellular survival. In mice, a family of cytosolic immune sensors, including NAIP1, NAIP2, and NAIP5/6, recognizes the SPI-1 T3SS needle, inner rod, and flagellin proteins, respectively. Ligand recognition triggers assembly of the NAIP/NLRC4 inflammasome, which mediates caspase-1 activation, IL-1 family cytokine secretion, and pyroptosis of infected cells. In contrast to mice, humans encode a single NAIP that broadly recognizes all three ligands. The role of NAIP/NLRC4 or other inflammasomes during Salmonella infection of human macrophages is unclear. We find that although the NAIP/NLRC4 inflammasome is essential for detecting T3SS ligands in human macrophages, it is partially required for responses to infection, as Salmonella also activated the NLRP3 and CASP4/5 inflammasomes. Importantly, we demonstrate that combinatorial NAIP/NLRC4 and NLRP3 inflammasome activation restricts Salmonella replication in human macrophages. In contrast to SPI-1, the SPI-2 T3SS inner rod is not sensed by human or murine NAIPs, which is thought to allow Salmonella to evade host recognition and replicate intracellularly. Intriguingly, we find that human NAIP detects the SPI-2 T3SS needle protein. Critically, in the absence of both flagellin and the SPI-1 T3SS, the NAIP/NLRC4 inflammasome still controlled intracellular Salmonella burden. These findings reveal that recognition of Salmonella SPI-1 and SPI-2 T3SSs and engagement of both the NAIP/NLRC4 and NLRP3 inflammasomes control Salmonella infection in human macrophages.


Asunto(s)
Inflamasomas/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Infecciones por Salmonella/inmunología , Sistemas de Secreción Tipo III/inmunología , Proteínas Adaptadoras de Señalización CARD/inmunología , Proteínas de Unión al Calcio/inmunología , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteína Inhibidora de la Apoptosis Neuronal/inmunología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Virulencia
14.
Nat Immunol ; 13(10): 954-62, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22922364

RESUMEN

Salmonella enterica serovar Typhimurium (S. Typhimurium) is a virulent pathogen that induces rapid host death. Here we observed that host survival after infection with S. Typhimurium was enhanced in the absence of type I interferon signaling, with improved survival of mice deficient in the receptor for type I interferons (Ifnar1(-/-) mice) that was attributed to macrophages. Although there was no impairment in cytokine expression or inflammasome activation in Ifnar1(-/-) macrophages, they were highly resistant to S. Typhimurium-induced cell death. Specific inhibition of the kinase RIP1 or knockdown of the gene encoding the kinase RIP3 prevented the death of wild-type macrophages, which indicated that necroptosis was a mechanism of cell death. Finally, RIP3-deficient macrophages, which cannot undergo necroptosis, had similarly less death and enhanced control of S. Typhimurium in vivo. Thus, we propose that S. Typhimurium induces the production of type I interferon, which drives necroptosis of macrophages and allows them to evade the immune response.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Interferón Tipo I/metabolismo , Macrófagos/fisiología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Salmonelosis Animal/inmunología , Salmonella typhimurium/patogenicidad , Animales , Apoptosis , Proteínas Activadoras de GTPasa/antagonistas & inhibidores , Evasión Inmune , Inflamasomas , Interferón Tipo I/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Salmonelosis Animal/metabolismo , Salmonelosis Animal/microbiología , Salmonella typhimurium/inmunología , Transducción de Señal
15.
Arch Microbiol ; 206(7): 302, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38874634

RESUMEN

Host-generated antimicrobial peptides (AMPs) play a pivotal role in defense against bacterial pathogens. AMPs kill invading bacteria majorly by disrupting the bacterial cell walls. AMPs are actively synthesized and released into the lumen of the gastrointestinal tract to limit colonization of enteric pathogens like Salmonella typhimurium (S. typhimurium). However, S. typhimurium has evolved several resistance mechanisms to defend AMPs. The multicomponent SapABCDF uptake transporter is one such system that helps in resisting AMPs. In the current study, we analyzed the role of S. typhimurium SapA against stress survival and virulence of this bacterium. ∆sapA mutant strain showed hypersensitivity to AMPs, like melittin and mastoparan. Further, ∆sapA mutant showed more than 22 folds (p = 0.019) hypersensitivity to neutrophils as compared to the WT strain of S. typhimurium. In addition, ∆sapA strain showed defective survival in mice. In conclusion, the results of the current study suggest that the SapA is essential for survival against AMPs and virulence of S. typhimurium.


Asunto(s)
Neutrófilos , Salmonella typhimurium , Animales , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/patogenicidad , Salmonella typhimurium/genética , Ratones , Neutrófilos/inmunología , Neutrófilos/microbiología , Virulencia , Péptidos Antimicrobianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Femenino , Ratones Endogámicos BALB C , Infecciones por Salmonella/microbiología , Péptidos Catiónicos Antimicrobianos/farmacología
16.
Cell ; 137(2): 283-94, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19379694

RESUMEN

Many bacterial pathogens and symbionts utilize type III secretion systems to deliver bacterial effector proteins into host cells. These effector proteins have the capacity to modulate a large variety of cellular functions in a highly regulated manner. Here, we report that the phosphoinositide phosphatase SopB, a Salmonella Typhimurium type III secreted effector protein, diversifies its function by localizing to different cellular compartments in a ubiquitin-dependent manner. We show that SopB utilizes the same enzymatic activity to modulate actin-mediated bacterial internalization and Akt activation at the plasma membrane and vesicular trafficking and intracellular bacterial replication at the phagosome. Thus, by exploiting the host cellular machinery, Salmonella Typhimurium has evolved the capacity to broaden the functional repertoire of a virulence factor to maximize its ability to modulate cellular functions.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ubiquitina/metabolismo , Actinas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Pinocitosis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad
17.
Mol Cell ; 63(1): 86-96, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27264868

RESUMEN

The recalcitrance of many bacterial infections to antibiotic treatment is thought to be due to the presence of persisters that are non-growing, antibiotic-insensitive cells. Eventually, persisters resume growth, accounting for relapses of infection. Salmonella is an important pathogen that causes disease through its ability to survive inside macrophages. After macrophage phagocytosis, a significant proportion of the Salmonella population forms non-growing persisters through the action of toxin-antitoxin modules. Here we reveal that one such toxin, TacT, is an acetyltransferase that blocks the primary amine group of amino acids on charged tRNA molecules, thereby inhibiting translation and promoting persister formation. Furthermore, we report the crystal structure of TacT and note unique structural features, including two positively charged surface patches that are essential for toxicity. Finally, we identify a detoxifying mechanism in Salmonella wherein peptidyl-tRNA hydrolase counteracts TacT-dependent growth arrest, explaining how bacterial persisters can resume growth.


Asunto(s)
Acetiltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , ARN Bacteriano/metabolismo , ARN de Transferencia/metabolismo , Salmonella typhimurium/enzimología , Aminoacilación de ARN de Transferencia , Acetiltransferasas/química , Proteínas Bacterianas/química , Toxinas Bacterianas/química , Hidrolasas de Éster Carboxílico/metabolismo , Farmacorresistencia Bacteriana/genética , Modelos Moleculares , Conformación Proteica , ARN Bacteriano/genética , ARN de Transferencia/genética , Salmonella typhimurium/genética , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/patogenicidad , Relación Estructura-Actividad , Propiedades de Superficie , Factores de Tiempo , Virulencia
18.
Mol Cell ; 62(6): 967-981, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27211868

RESUMEN

Ubiquitination serves as a critical signal in the host immune response to infection. Many pathogens have evolved strategies to exploit the ubiquitin (Ub) system to promote their own survival through a complex interplay between host defense machinery and bacterial virulence factors. Here we report dynamic changes in the global ubiquitinome of host epithelial cells and invading pathogen in response to Salmonella Typhimurium infection. The most significant alterations in the host ubiquitinome concern components of the actin cytoskeleton, NF-κB and autophagy pathways, and the Ub and RHO GTPase systems. Specifically, infection-induced ubiquitination promotes CDC42 activity and linear ubiquitin chain formation, both being required for NF-κB activation. Conversely, the bacterial ubiquitinome exhibited extensive ubiquitination of various effectors and several outer membrane proteins. Moreover, we reveal that bacterial Ub-modifying enzymes modulate a unique subset of host targets, affecting different stages of Salmonella infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Proteómica/métodos , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Ubiquitinación , Células Epiteliales/microbiología , Células HCT116 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Infecciones por Salmonella/genética , Salmonella typhimurium/patogenicidad , Factores de Tiempo , Transfección , Proteína de Unión al GTP cdc42/metabolismo
19.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34706941

RESUMEN

Type III secretion systems are multiprotein molecular machines required for the virulence of several important bacterial pathogens. The central element of these machines is the injectisome, a ∼5-Md multiprotein structure that mediates the delivery of bacterially encoded proteins into eukaryotic target cells. The injectisome is composed of a cytoplasmic sorting platform, and a membrane-embedded needle complex, which is made up of a multiring base and a needle-like filament that extends several nanometers from the bacterial surface. The needle filament is capped at its distal end by another substructure known as the tip complex, which is crucial for the translocation of effector proteins through the eukaryotic cell plasma membrane. Here we report the cryo-EM structure of the Salmonella Typhimurium needle tip complex docked onto the needle filament tip. Combined with a detailed analysis of structurally guided mutants, this study provides major insight into the assembly and function of this essential component of the type III secretion protein injection machine.


Asunto(s)
Salmonella typhimurium/ultraestructura , Sistemas de Secreción Tipo III/ultraestructura , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Sistemas de Secreción Bacterianos/metabolismo , Sistemas de Secreción Bacterianos/ultraestructura , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Microscopía por Crioelectrón/métodos , Citoesqueleto/metabolismo , Citosol/metabolismo , Transporte de Proteínas/fisiología , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Sistemas de Secreción Tipo III/metabolismo , Sistemas de Secreción Tipo III/fisiología
20.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33846244

RESUMEN

The gut epithelium serves to maximize the surface for nutrient and fluid uptake, but at the same time must provide a tight barrier to pathogens and remove damaged intestinal epithelial cells (IECs) without jeopardizing barrier integrity. How the epithelium coordinates these tasks remains a question of significant interest. We used imaging and an optical flow analysis pipeline to study the dynamicity of untransformed murine and human intestinal epithelia, cultured atop flexible hydrogel supports. Infection with the pathogen Salmonella Typhimurium (STm) within minutes elicited focal contractions with inward movements of up to ∼1,000 IECs. Genetics approaches and chimeric epithelial monolayers revealed contractions to be triggered by the NAIP/NLRC4 inflammasome, which sensed type-III secretion system and flagellar ligands upon bacterial invasion, converting the local tissue into a contraction epicenter. Execution of the response required swift sublytic Gasdermin D pore formation, ion fluxes, and the propagation of a myosin contraction pulse across the tissue. Importantly, focal contractions preceded, and could be uncoupled from, the death and expulsion of infected IECs. In both two-dimensional monolayers and three-dimensional enteroids, multiple infection-elicited contractions coalesced to produce shrinkage of the epithelium as a whole. Monolayers deficient for Caspase-1(-11) or Gasdermin D failed to elicit focal contractions but were still capable of infected IEC death and expulsion. Strikingly, these monolayers lost their integrity to a markedly higher extent than wild-type counterparts. We propose that prompt NAIP/NLRC4/Caspase-1/Gasdermin D/myosin-dependent contractions allow the epithelium to densify its cell packing in infected regions, thereby preventing tissue disintegration due to the subsequent IEC death and expulsion process.


Asunto(s)
Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiología , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Animales , Infecciones Bacterianas/fisiopatología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Caspasa 1/metabolismo , Caspasas/metabolismo , Células Epiteliales/metabolismo , Epitelio/metabolismo , Humanos , Inflamasomas , Mucosa Intestinal/microbiología , Intestinos , Ratones , Contracción Muscular/fisiología , Cultivo Primario de Células , Receptores de Reconocimiento de Patrones/metabolismo , Salmonella typhimurium/patogenicidad , Sistemas de Secreción Tipo III/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA