Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 42(17): e113012, 2023 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-37409490

RESUMEN

Invasive bacteria enter the cytosol of host cells through initial uptake into bacteria-containing vacuoles (BCVs) and subsequent rupture of the BCV membrane, thereby exposing to the cytosol intraluminal, otherwise shielded danger signals such as glycans and sphingomyelin. The detection of glycans by galectin-8 triggers anti-bacterial autophagy, but how cells sense and respond to cytosolically exposed sphingomyelin remains unknown. Here, we identify TECPR1 (tectonin beta-propeller repeat containing 1) as a receptor for cytosolically exposed sphingomyelin, which recruits ATG5 into an E3 ligase complex that mediates lipid conjugation of LC3 independently of ATG16L1. TECPR1 binds sphingomyelin through its N-terminal DysF domain (N'DysF), a feature not shared by other mammalian DysF domains. Solving the crystal structure of N'DysF, we identified key residues required for the interaction, including a solvent-exposed tryptophan (W154) essential for binding to sphingomyelin-positive membranes and the conjugation of LC3 to lipids. Specificity of the ATG5/ATG12-E3 ligase responsible for the conjugation of LC3 is therefore conferred by interchangeable receptor subunits, that is, the canonical ATG16L1 and the sphingomyelin-specific TECPR1, in an arrangement reminiscent of certain multi-subunit ubiquitin E3 ligases.


Asunto(s)
Proteínas Asociadas a Microtúbulos , Esfingomielinas , Animales , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas Portadoras/metabolismo , Autofagia , Ubiquitina-Proteína Ligasas/metabolismo , Proteína 5 Relacionada con la Autofagia/metabolismo , Mamíferos
2.
EMBO J ; 39(17): e104469, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32657447

RESUMEN

Upon invasive bacterial infection of colonic epithelium, host cells induce several types of cell death to eliminate pathogens. For instance, necroptosis is a RIPK-dependent lytic cell death that serves as a backup system to fully eliminate intracellular pathogens when apoptosis is inhibited; this phenomenon has been termed "cell death crosstalk". To maintain their replicative niche and multiply within cells, some enteric pathogens prevent epithelial cell death by delivering effectors via the type III secretion system. In this study, we found that Shigella hijacks host cell death crosstalk via a dual mechanism: inhibition of apoptosis by the OspC1 effector and inhibition of necroptosis by the OspD3 effector. Upon infection by Shigella, host cells recognize blockade of caspase-8 apoptosis signaling by OspC1 effector as a key danger signal and trigger necroptosis as a backup form of host defense. To counteract this backup defense, Shigella delivers the OspD3 effector, a protease, to degrade RIPK1 and RIPK3, preventing necroptosis. We believe that blockade of host cell death crosstalk by Shigella is a unique intracellular survival tactic for prolonging the bacterium's replicative niche.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Caspasa 8/metabolismo , Necroptosis , Péptido Hidrolasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Shigella flexneri/metabolismo , Células HCT116 , Células HT29 , Células HeLa , Humanos , Shigella flexneri/patogenicidad
3.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33753513

RESUMEN

Helicobacter suis, a bacterial species naturally hosted by pigs, can colonize the human stomach in the context of gastric diseases such as gastric mucosa-associated lymphoid tissue (MALT) lymphoma. Because H. suis has been successfully isolated from pigs, but not from humans, evidence linking human H. suis infection to gastric diseases has remained incomplete. In this study, we successfully in vitro cultured H. suis directly from human stomachs. Unlike Helicobacter pylori, the viability of H. suis decreases significantly on neutral pH; therefore, we achieved this using a low-pH medium for transport of gastric biopsies. Ultimately, we isolated H. suis from three patients with gastric diseases, including gastric MALT lymphoma. Successful eradication of H. suis yielded significant improvements in endoscopic and histopathological findings. Oral infection of mice with H. suis clinical isolates elicited gastric and systemic inflammatory responses; in addition, progression of gastric mucosal metaplasia was observed 4 mo postinfection. Because H. suis could be isolated from the stomachs of infected mice, our findings satisfied Koch's postulates. Although further prospective clinical studies are needed, H. suis, like H. pylori, is likely a gastric pathogen in humans. Furthermore, comparative genomic analysis of H. suis using complete genomes of clinical isolates revealed that the genome of each H. suis isolate contained highly plastic genomic regions encoding putative strain-specific virulence factors, including type IV secretion system-associated genes, and that H. suis isolates from humans and pigs were genetically very similar, suggesting possible pig-to-human transmission.


Asunto(s)
Infecciones por Helicobacter/genética , Helicobacter heilmannii/genética , Helicobacter heilmannii/patogenicidad , Gastropatías/microbiología , Estómago/microbiología , Factores de Virulencia/genética , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Genoma Bacteriano , Helicobacter heilmannii/aislamiento & purificación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Porcinos , Sistemas de Secreción Tipo IV/genética , Virulencia/genética
4.
Curr Microbiol ; 78(1): 55-66, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33145611

RESUMEN

Erysipelothrix rhusiopathiae causes swine erysipelas (SE), which results in considerable economic loss on pig farms. During SE outbreaks that occurred sporadically from 2008 to 2011 in Japan, new E. rhusiopathiae strains were isolated with a specific surface protective antigen (Spa)A protein characterized by methionine at position 203 and isoleucine at position 257 (M203/I257 SpaA type). To determine whether strains with the M203/I257 SpaA type are still prevalent in Japan, we collected 79 strains of E. rhusiopathiae from pigs showing various SE symptoms from 2012 to 2019 and classified them based on serovar typing, spaA gene sequence analysis, and lineage typing. We found that the majority of recent E. rhusiopathiae strains (59/79) belonged to the serovar 1a strain, and that the M203/I257 SpaA type (56/59) was predominant continuing from 2008 to 2011. Furthermore, serovar 1a strains with IVb-1 and IVb-2 lineages that had been isolated in specific regions of Japan were no longer local but were found across Japan. The pathogenicity of recent isolates tested in mice was not significantly changed when compared to that of previously isolated strains. Our results suggest that recent SE outbreaks were not due to changes in the SpaA protein or to altered virulence of E. rhusiopathiae but were rather caused by the persistent presence of E. rhusiopathiae with the M203/I257 SpaA type.


Asunto(s)
Erysipelothrix , Erisipela Porcina , Animales , Erysipelothrix/genética , Japón , Ratones , Serogrupo , Porcinos , Virulencia
5.
Cell Microbiol ; 21(3): e12974, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30414351

RESUMEN

Subversion of antigen-specific immune responses by intracellular pathogens is pivotal for successful colonisation. Bacterial pathogens, including Shigella, deliver effectors into host cells via the type III secretion system (T3SS) in order to manipulate host innate and adaptive immune responses, thereby promoting infection. However, the strategy for subverting antigen-specific immunity is not well understood. Here, we show that Shigella flexneri invasion plasmid antigen H (IpaH) 4.5, a member of the E3 ubiquitin ligase effector family, targets the proteasome regulatory particle non-ATPase 13 (RPN13) and induces its degradation via the ubiquitin-proteasome system (UPS). IpaH4.5-mediated RPN13 degradation causes dysfunction of the 19S regulatory particle (RP) in the 26S proteasome, inhibiting guidance of ubiquitinated proteins to the proteolytically active 20S core particle (CP) of 26S proteasome and thereby suppressing proteasome-catalysed peptide splicing. This, in turn, reduces antigen cross-presentation to CD8+ T cells via major histocompatibility complex (MHC) class I in vitro. In RPN13 knockout mouse embryonic fibroblasts (MEFs), loss of RPN13 suppressed CD8+ T cell priming during Shigella infection. Our results uncover the unique tactics employed by Shigella to dampen the antigen-specific cytotoxic T lymphocyte (CTL) response.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Evasión Inmune , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Complejo de la Endopetidasa Proteasomal/metabolismo , Shigella flexneri/crecimiento & desarrollo , Linfocitos T Citotóxicos/inmunología , Animales , Células Cultivadas , Análisis por Conglomerados , ADN Ribosómico/química , ADN Ribosómico/genética , Modelos Animales de Enfermedad , Disentería Bacilar/microbiología , Disentería Bacilar/patología , Humanos , Activación de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Teóricos , Filogenia , ARN Ribosómico/genética , Análisis de Secuencia de ADN , Shigella flexneri/inmunología , Shigella flexneri/patogenicidad , Linfocitos T Citotóxicos/microbiología , Factores de Virulencia/metabolismo
6.
EMBO Rep ; 19(1): 89-101, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29191979

RESUMEN

Shigella deploys a unique mechanism to manipulate macrophage pyroptosis by delivering the IpaH7.8 E3 ubiquitin ligase via its type III secretion system. IpaH7.8 ubiquitinates glomulin (GLMN) and elicits its degradation, thereby inducing inflammasome activation and pyroptotic cell death of macrophages. Here, we show that GLMN specifically binds cellular inhibitor of apoptosis proteins 1 and 2 (cIAP1 and cIAP2), members of the inhibitor of apoptosis (IAP) family of RING-E3 ligases, which results in reduced E3 ligase activity, and consequently inflammasome-mediated death of macrophages. Importantly, reducing the levels of GLMN in macrophages via IpaH7.8, or siRNA-mediated knockdown, enhances inflammasome activation in response to infection by Shigella, Salmonella, or Pseudomonas, stimulation with NLRP3 inflammasome activators (including SiO2, alum, or MSU), or stimulation of the AIM2 inflammasome by poly dA:dT GLMN binds specifically to the RING domain of both cIAPs, which inhibits their self-ubiquitination activity. These findings suggest that GLMN is a negative regulator of cIAP-mediated inflammasome activation, and highlight a unique Shigella stratagem to kill macrophages, promoting severe inflammation.


Asunto(s)
Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Interacciones Huésped-Patógeno , Inflamasomas/genética , Proteínas Inhibidoras de la Apoptosis/genética , Macrófagos/microbiología , Proteínas Musculares/genética , Shigella flexneri/inmunología , Secuencia de Aminoácidos , Animales , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Regulación de la Expresión Génica , Inflamasomas/inmunología , Proteínas Inhibidoras de la Apoptosis/inmunología , Isoenzimas/genética , Isoenzimas/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/inmunología , Cultivo Primario de Células , Unión Proteica , Piroptosis/genética , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/inmunología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Shigella flexneri/crecimiento & desarrollo , Transducción de Señal , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/inmunología
7.
EMBO J ; 33(22): 2598-600, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25246515

RESUMEN

Bacterial pathogens alter host transcriptional programs to promote infection. Shigella OspF is an essential virulence protein with a unique phosphothreonine lyase activity. A new study in The EMBO Journal (Harouz et al, 2014) reveals a novel function of OspF: targeting of heterochromatin protein 1γ (HP1γ) and downregulation of a subset of immune genes. These results illustrate how bacterial pathogens exploit epigenetic modifications to counteract host immune responses.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Liasas de Carbono-Oxígeno/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Disentería Bacilar/metabolismo , Enterocolitis/metabolismo , Shigella flexneri/metabolismo , Transcriptoma , Animales , Homólogo de la Proteína Chromobox 5
8.
Virol J ; 15(1): 28, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29394943

RESUMEN

BACKGROUND: Porcine epidemic diarrhea (PED) is a lethal infectious disease in suckling piglets with symptoms including watery diarrhea caused by PED virus (PEDV). Since the late 1990's, live vaccines based on genogroup 1 virus have been used in Japan, and a significant amount of the vaccine has been used even after new genogroups invaded in 2013. In this study, we evaluated the effect of a conventional PED live vaccine on a newly prevalent genogroup 2 field strain in experimental and field situations. METHODS: Two pregnant sows were administered twice the live vaccine before farrowing. A pregnant sow was served as a negative control. All newborn piglets were challenged with the genogroup 2 virus, and clinical signs were monitored for 7 days post challenge. PEDV-specific immune responses in serum and milk of the sows were assayed by virus neutralization assay. The efficacy of PED live vaccine in vaccinated or non-vaccinated farms was evaluated by comparing the mortality rate of suckling piglets after the onset of PED. RESULTS: The challenged piglets exhibited watery diarrhea with or without vaccination. However, the clinical score of piglets born from vaccinated sows significantly improved after the 4th day of the challenge. The survival rate of piglets in the vaccinated group at the end of the experimental period was 80%, whereas in the control group was 0%. Neutralizing antibody titers in serum and milk of control sow was negative throughout the experimental period, whereas high titers were observed in the vaccinated sows. The vaccinated farms significantly reduced the mortality rate of suckling piglets after the onset of PED, compared to farms not vaccinated. CONCLUSIONS: The conventional PED live vaccine induced the lactogenic immunity to vaccinated sows and showed partial protection against the genogroup 2 virus both under the experimental and field conditions.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Virus de la Diarrea Epidémica Porcina/inmunología , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/virología , Vacunas de Productos Inactivados/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Chlorocebus aethiops , Femenino , Genotipo , Inmunización , Japón , Pruebas de Neutralización , Evaluación de Resultado en la Atención de Salud , Filogenia , Virus de la Diarrea Epidémica Porcina/genética , Embarazo , Porcinos , Enfermedades de los Porcinos/diagnóstico , Enfermedades de los Porcinos/inmunología , Células Vero
9.
Immunity ; 30(1): 108-19, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19144317

RESUMEN

Interleukin-17A (IL-17A) is a cytokine produced by T helper 17 (Th17) cells and plays important roles in the development of inflammatory diseases. Although IL-17F is highly homologous to IL-17A and binds the same receptor, the functional roles of this molecule remain largely unknown. Here, we demonstrated with Il17a(-/-), Il17f(-/-), and Il17a(-/-)Il17f(-/-) mice that IL-17F played only marginal roles, if at all, in the development of delayed-type and contact hypersensitivities, autoimmune encephalomyelitis, collagen-induced arthritis, and arthritis in Il1rn(-/-) mice. In contrast, both IL-17F and IL-17A were involved in host defense against mucoepithelial infection by Staphylococcus aureus and Citrobacter rodentium. IL-17A was produced mainly in T cells, whereas IL-17F was produced in T cells, innate immune cells, and epithelial cells. Although only IL-17A efficiently induced cytokines in macrophages, both cytokines activated epithelial innate immune responses. These observations indicate that IL-17A and IL-17F have overlapping yet distinct roles in host immune and defense mechanisms.


Asunto(s)
Artritis/inmunología , Infecciones Bacterianas/inmunología , Citocinas/metabolismo , Hipersensibilidad/inmunología , Interleucina-17/clasificación , Interleucina-17/fisiología , Animales , Artritis/genética , Infecciones Bacterianas/prevención & control , Células Cultivadas , Citometría de Flujo , Interleucina-17/genética , Ratones , Ratones Noqueados
10.
Nature ; 483(7391): 623-6, 2012 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-22407319

RESUMEN

Many bacterial pathogens can enter various host cells and then survive intracellularly, transiently evade humoral immunity, and further disseminate to other cells and tissues. When bacteria enter host cells and replicate intracellularly, the host cells sense the invading bacteria as damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) by way of various pattern recognition receptors. As a result, the host cells induce alarm signals that activate the innate immune system. Therefore, bacteria must modulate host inflammatory signalling and dampen these alarm signals. How pathogens do this after invading epithelial cells remains unclear, however. Here we show that OspI, a Shigella flexneri effector encoded by ORF169b on the large plasmid and delivered by the type ΙΙΙ secretion system, dampens acute inflammatory responses during bacterial invasion by suppressing the tumour-necrosis factor (TNF)-receptor-associated factor 6 (TRAF6)-mediated signalling pathway. OspI is a glutamine deamidase that selectively deamidates the glutamine residue at position 100 in UBC13 to a glutamic acid residue. Consequently, the E2 ubiquitin-conjugating activity required for TRAF6 activation is inhibited, allowing S. flexneri OspI to modulate the diacylglycerol-CBM (CARD-BCL10-MALT1) complex-TRAF6-nuclear-factor-κB signalling pathway. We determined the 2.0 Å crystal structure of OspI, which contains a putative cysteine-histidine-aspartic acid catalytic triad. A mutational analysis showed this catalytic triad to be essential for the deamidation of UBC13. Our results suggest that S. flexneri inhibits acute inflammatory responses in the initial stage of infection by targeting the UBC13-TRAF6 complex.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Amidohidrolasas/química , Amidohidrolasas/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Shigella flexneri/enzimología , Shigella flexneri/inmunología , Enzimas Ubiquitina-Conjugadoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Amidohidrolasas/genética , Secuencia de Aminoácidos , Animales , Ácido Aspártico/metabolismo , Proteína 10 de la LLC-Linfoma de Células B , Biocatálisis , Caspasas/metabolismo , Dominio Catalítico/genética , Cristalografía por Rayos X , Cisteína/metabolismo , Análisis Mutacional de ADN , Diglicéridos/antagonistas & inhibidores , Diglicéridos/metabolismo , Disentería Bacilar/microbiología , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Células HEK293 , Células HeLa , Histidina/metabolismo , Humanos , Inmunidad Innata , Inflamación/enzimología , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Shigella flexneri/genética , Shigella flexneri/patogenicidad , Factor 6 Asociado a Receptor de TNF/deficiencia , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Enzimas Ubiquitina-Conjugadoras/química , Enzimas Ubiquitina-Conjugadoras/genética , Factores de Virulencia/metabolismo
11.
Genes Cells ; 21(6): 608-23, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27091465

RESUMEN

Many bacterial pathogens hijack the host ubiquitin system for their own benefit by delivering effectors with ubiquitin ligase (E3) into host cells via the type III secretion system. Therefore, screening for small compounds that selectively inhibit bacterial but not mammalian E3 ligases is a promising strategy for identifying molecules that could substitute for antibiotics. To facilitate high-throughput screening for bacterial E3 ligase inhibitors, we developed a MiCy/mKO (Midori-ishi Cyan/monomeric Kusabira-Orange)-based FRET (fluorescence resonance energy transfer) assay and validated it on Shigella IpaH E3 ligase effectors. We showed the feasibility of using the MiCy/mKO-based FRET assay to identify the most appropriate ubiquitin-conjugating enzymes (E2s) and determine the lysine specificity of a given E3, both hallmarks of E3 activity. Furthermore, we showed the usefulness of the FRET assay in characterizing mammalian E3 ligases, such as TNF receptor-associated factor 6 (TRAF6) and mouse double minute 2 homologue (MDM2). In addition, we confirmed the feasibility of determining the efficiency of inhibition of E3 ligase activity using inhibitors of E1 ubiquitin-activating enzymes, such as UBE1-41, by measuring the IC50 . Based on these results, we concluded that the MiCy/mKO-based FRET assay is useful for characterizing E3 enzyme activity, as well as for high-throughput E3 inhibitor screening.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Fluorescentes Verdes/análisis , Shigella flexneri/enzimología , Ubiquitina-Proteína Ligasas/química , Animales , Disentería Bacilar/enzimología , Disentería Bacilar/microbiología , Ensayos Analíticos de Alto Rendimiento , Humanos , Concentración 50 Inhibidora , Proteínas Luminiscentes/análisis , Lisina/metabolismo , Ratones , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Proteína Fluorescente Roja
12.
Proc Natl Acad Sci U S A ; 111(40): E4254-63, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25246571

RESUMEN

When nucleotide-binding oligomerization domain-like receptors (NLRs) sense cytosolic-invading bacteria, they induce the formation of inflammasomes and initiate an innate immune response. In quiescent cells, inflammasome activity is tightly regulated to prevent excess inflammation and cell death. Many bacterial pathogens provoke inflammasome activity and induce inflammatory responses, including cell death, by delivering type III secreted effectors, the rod component flagellin, and toxins. Recent studies indicated that Shigella deploy multiple mechanisms to stimulate NLR inflammasomes through type III secretion during infection. Here, we show that Shigella induces rapid macrophage cell death by delivering the invasion plasmid antigen H7.8 (IpaH7.8) enzyme 3 (E3) ubiquitin ligase effector via the type III secretion system, thereby activating the NLR family pyrin domain-containing 3 (NLRP3) and NLR family CARD domain-containing 4 (NLRC4) inflammasomes and caspase-1 and leading to macrophage cell death in an IpaH7.8 E3 ligase-dependent manner. Mice infected with Shigella possessing IpaH7.8, but not with Shigella possessing an IpaH7.8 E3 ligase-null mutant, exhibited enhanced bacterial multiplication. We defined glomulin/flagellar-associated protein 68 (GLMN) as an IpaH7.8 target involved in IpaH7.8 E3 ligase-dependent inflammasome activation. This protein originally was identified through its association with glomuvenous malformations and more recently was described as a member of a Cullin ring ligase inhibitor. Modifying GLMN levels through overexpression or knockdown led to reduced or augmented inflammasome activation, respectively. Macrophages stimulated with lipopolysaccharide/ATP induced GLMN puncta that localized with the active form of caspase-1. Macrophages from GLMN(+/-) mice were more responsive to inflammasome activation than those from GLMN(+/+) mice. Together, these results highlight a unique bacterial adaptation that hijacks inflammasome activation via interactions between IpaH7.8 and GLMN.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Proteínas Musculares/metabolismo , Shigella flexneri/metabolismo , Animales , Antígenos Bacterianos/genética , Apoptosis , Proteínas Bacterianas/genética , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Femenino , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Immunoblotting , Células Jurkat , Macrófagos/microbiología , Ratones Endogámicos BALB C , Ratones Noqueados , Microscopía Fluorescente , Proteínas Musculares/genética , Unión Proteica , Shigella flexneri/genética , Shigella flexneri/fisiología , Técnicas del Sistema de Dos Híbridos
13.
PLoS Pathog ; 10(2): e1003926, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24516390

RESUMEN

Recognition of intracellular pathogenic bacteria by members of the nucleotide-binding domain and leucine-rich repeat containing (NLR) family triggers immune responses against bacterial infection. A major response induced by several Gram-negative bacteria is the activation of caspase-1 via the Nlrc4 inflammasome. Upon activation, caspase-1 regulates the processing of proIL-1ß and proIL-18 leading to the release of mature IL-1ß and IL-18, and induction of pyroptosis. The activation of the Nlrc4 inflammasome requires the presence of an intact type III or IV secretion system that mediates the translocation of small amounts of flagellin or PrgJ-like rod proteins into the host cytosol to induce Nlrc4 activation. Using the Salmonella system, it was shown that Naip2 and Naip5 link flagellin and the rod protein PrgJ, respectively, to Nlrc4. Furthermore, phosphorylation of Nlrc4 at Ser533 by Pkcδ was found to be critical for the activation of the Nlrc4 inflammasome. Here, we show that Naip2 recognizes the Shigella T3SS inner rod protein MxiI and induces Nlrc4 inflammasome activation. The expression of MxiI in primary macrophages was sufficient to induce pyroptosis and IL-1ß release, which were prevented in macrophages deficient in Nlrc4. In the presence of MxiI or Shigella infection, MxiI associated with Naip2, and Naip2 interacted with Nlrc4. siRNA-mediated knockdown of Naip2, but not Naip5, inhibited Shigella-induced caspase-1 activation, IL-1ß maturation and Asc pyroptosome formation. Notably, the Pkcδ kinase was dispensable for caspase-1 activation and secretion of IL-1ß induced by Shigella or Salmonella infection. These results indicate that activation of caspase-1 by Shigella is triggered by the rod protein MxiI that interacts with Naip2 to induce activation of the Nlrc4 inflammasome independently of the Pkcδ kinase.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Interacciones Huésped-Parásitos/inmunología , Inflamasomas/metabolismo , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Proteína Quinasa C-delta/metabolismo , Animales , Caspasa 1/metabolismo , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Transfección
14.
J Virol ; 88(16): 8981-97, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24899188

RESUMEN

UNLABELLED: Occasional transmission of highly pathogenic avian H5N1 influenza viruses to humans causes severe pneumonia with high mortality. To better understand the mechanisms via which H5N1 viruses induce severe disease in humans, we infected cynomolgus macaques with six different H5N1 strains isolated from human patients and compared their pathogenicity and the global host responses to the virus infection. Although all H5N1 viruses replicated in the respiratory tract, there was substantial heterogeneity in their replicative ability and in the disease severity induced, which ranged from asymptomatic to fatal. A comparison of global gene expression between severe and mild disease cases indicated that interferon-induced upregulation of genes related to innate immunity, apoptosis, and antigen processing/presentation in the early phase of infection was limited in severe disease cases, although interferon expression was upregulated in both severe and mild cases. Furthermore, coexpression analysis of microarray data, which reveals the dynamics of host responses during the infection, demonstrated that the limited expression of these genes early in infection led to a failure to suppress virus replication and to the hyperinduction of genes related to immunity, inflammation, coagulation, and homeostasis in the late phase of infection, resulting in a more severe disease. Our data suggest that the attenuated interferon-induced activation of innate immunity, apoptosis, and antigen presentation in the early phase of H5N1 virus infection leads to subsequent severe disease outcome. IMPORTANCE: Highly pathogenic avian H5N1 influenza viruses sometimes transmit to humans and cause severe pneumonia with ca. 60% lethality. The continued circulation of these viruses poses a pandemic threat; however, their pathogenesis in mammals is not fully understood. We, therefore, investigated the pathogenicity of six H5N1 viruses and compared the host responses of cynomolgus macaques to the virus infection. We identified differences in the viral replicative ability of and in disease severity caused by these H5N1 viruses. A comparison of global host responses between severe and mild disease cases identified the limited upregulation of interferon-stimulated genes early in infection in severe cases. The dynamics of the host responses indicated that the limited response early in infection failed to suppress virus replication and led to hyperinduction of pathological condition-related genes late in infection. These findings provide insight into the pathogenesis of H5N1 viruses in mammals.


Asunto(s)
Regulación Viral de la Expresión Génica/genética , Expresión Génica/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/virología , Primates/virología , Animales , Presentación de Antígeno/inmunología , Apoptosis/inmunología , Células Cultivadas , Perros , Expresión Génica/inmunología , Regulación Viral de la Expresión Génica/inmunología , Humanos , Inmunidad Innata/inmunología , Inflamación/inmunología , Inflamación/virología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Macaca/inmunología , Macaca/virología , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Células de Riñón Canino Madin Darby , Infecciones por Orthomyxoviridae/inmunología , Primates/inmunología , Sistema Respiratorio/inmunología , Sistema Respiratorio/virología , Índice de Severidad de la Enfermedad , Replicación Viral/genética , Replicación Viral/inmunología
15.
PLoS Pathog ; 9(6): e1003409, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23754945

RESUMEN

NF-κB plays a central role in modulating innate immune responses to bacterial infections. Therefore, many bacterial pathogens deploy multiple mechanisms to counteract NF-κB activation. The invasion of and subsequent replication of Shigella within epithelial cells is recognized by various pathogen recognition receptors as pathogen-associated molecular patterns. These receptors trigger innate defense mechanisms via the activation of the NF-κB signaling pathway. Here, we show the inhibition of the NF-κB activation by the delivery of the IpaH E3 ubiquitin ligase family member IpaH0722 using Shigella's type III secretion system. IpaH0722 dampens the acute inflammatory response by preferentially inhibiting the PKC-mediated activation of NF-κB by ubiquitinating TRAF2, a molecule downstream of PKC, and by promoting its proteasome-dependent degradation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Disentería Bacilar/enzimología , Células Epiteliales/metabolismo , FN-kappa B/metabolismo , Proteína Quinasa C/metabolismo , Proteolisis , Shigella/enzimología , Factor 2 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas Bacterianas/genética , Sistemas de Secreción Bacterianos/genética , Células COS , Chlorocebus aethiops , Disentería Bacilar/genética , Disentería Bacilar/patología , Células Epiteliales/microbiología , Células Epiteliales/patología , Células HeLa , Humanos , Ratones , Ratones Noqueados , FN-kappa B/genética , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Quinasa C/genética , Shigella/genética , Transducción de Señal/genética , Factor 2 Asociado a Receptor de TNF/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/genética
16.
Cell Microbiol ; 16(12): 1757-66, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25264025

RESUMEN

Host cells deploy multiple defences against microbial infection. One prominent host defence mechanism, the death of infected cells, plays a pivotal role in clearing damaged cells, eliminating pathogens, removing replicative niches, exposing intracellular bacterial pathogens to extracellular immune surveillance and presenting bacteria-derived antigens to the adaptive immune system. Although cell death can occur under either physiological or pathophysiological conditions, it acts as an innate defence mechanism against bacterial pathogens by limiting their persistent colonization. However, many bacterial pathogens, including Shigella, have evolved mechanisms that manipulate host cell death for their own benefit.


Asunto(s)
Apoptosis , Interacciones Huésped-Patógeno , Evasión Inmune , Shigella/crecimiento & desarrollo , Shigella/inmunología , Shigella/metabolismo
17.
Nature ; 459(7246): 578-82, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19489119

RESUMEN

The rapid turnover and exfoliation of mucosal epithelial cells provides an innate defence system against bacterial infection. Nevertheless, many pathogenic bacteria, including Shigella, are able to surmount exfoliation and colonize the epithelium efficiently. Here we show that the Shigella flexneri effector OspE (consisting of OspE1 and OspE2 proteins), which is highly conserved among enteropathogenic Escherichia coli, enterohaemorrhagic E. coli, Citrobacter rodentium and Salmonella strains, reinforces host cell adherence to the basement membrane by interacting with integrin-linked kinase (ILK). The number of focal adhesions was augmented along with membrane fraction ILK by ILK-OspE binding. The interaction between ILK and OspE increased cell surface levels of 1 integrin and suppressed phosphorylation of focal adhesion kinase and paxillin, which are required for rapid turnover of focal adhesion in cell motility. Nocodazole-washout-induced focal adhesion disassembly was blocked by expression of OspE. Polarized epithelial cells infected with a Shigella mutant lacking the ospE gene underwent more rapid cell detachment than cells infected with wild-type Shigella. Infection of guinea pig colons with Shigella corroborated the pivotal role of the OspE-ILK interaction in suppressing epithelial detachment, increasing bacterial cell-to-cell spreading, and promoting bacterial colonization. These results indicate that Shigella sustain their infectious foothold by using special tactics to prevent detachment of infected cells.


Asunto(s)
Adhesión Celular/fisiología , Adhesiones Focales/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Shigella flexneri/fisiología , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Adhesión Celular/efectos de los fármacos , Polaridad Celular , Colon/microbiología , Células Epiteliales/citología , Células Epiteliales/microbiología , Adhesiones Focales/efectos de los fármacos , Cobayas , Células HeLa , Humanos , Integrina beta1/metabolismo , Ratones , Nocodazol/farmacología , Fosforilación , Unión Proteica , Shigella flexneri/patogenicidad , Factores de Virulencia/deficiencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
18.
Nat Cell Biol ; 9(1): 121-8, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17173036

RESUMEN

Shigella use a special mechanism to invade epithelial cells called 'the trigger mechanism of entry', which allows epithelial cells to trap several bacteria simultaneously. On contact, Shigella deliver effectors into epithelial cells through the type III secretion system. Here, we show that one of the effectors, IpgB1, has a pivotal role in producing membrane ruffles by exploiting the RhoG-ELMO-Dock180 pathway to stimulate Rac1 activity. Using pulldown assays, we identified engulfment and cell motility (ELMO) protein as the IpgB1 binding partner. IpgB1 colocalized with ELMO and Dock180 in membrane ruffles induced by Shigella. Shigella invasiveness and IpgB1-induced ruffles were less in ELMO- and Dock180-knockdown cells compared with wild-type cells. Membrane association of ELMO-Dock180 with ruffles were promoted when cells expressed an IpgB1-ELMO chimera, establishing that IpgB1 mimics the role of RhoG in producing membrane ruffles. Taken together, our findings show that IpgB1 mimicry is the key to invasion by Shigella.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adhesión Bacteriana , Shigella/patogenicidad , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Membrana Celular/metabolismo , Perros , Células HeLa , Humanos , Inmunoprecipitación , Ratones , Modelos Biológicos , Células 3T3 NIH , Transporte de Proteínas , Interferencia de ARN , Shigella/fisiología , Transducción de Señal , Transducción Genética , Transfección , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/fisiología
19.
RNA Biol ; 11(11): 1347-54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25483038

RESUMEN

Overexpression of SIRT1 is frequently observed in various types of cancers, suggesting its potential role in malignancies. However, the molecular basis of how SIRT1 is elevated in cancer is less understood. Here we show that cancer-related SIRT1 overexpression is due to evasion of Sirt1 mRNA from repression by a group of Sirt1-targeting microRNAs (miRNAs) that might be robustly silenced in cancer. Our comprehensive library-based screening and subsequent miRNA gene profiling revealed a housekeeping gene-like broad expression pattern and strong CpG island-association of the Sirt1-targeting miRNA genes. This suggests aberrant CpG DNA methylation as the mechanistic background for malignant SIRT1 elevation. Our work also provides an example where epigenetic mechanisms cause the group-wide regulation of miRNAs sharing a common key target.


Asunto(s)
Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Sirtuina 1/genética , Regiones no Traducidas 3'/genética , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Islas de CpG/genética , Metilación de ADN , Etopósido/farmacología , Células HeLa , Humanos , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Nat Chem Biol ; 8(1): 36-45, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22173358

RESUMEN

The gut mucosa acts as a barrier against microbial invaders, whereas resident commensal and foreign invading bacteria interact intimately with the gut epithelium and influence the host cellular and immune systems. The epithelial barrier serves as an infectious foothold for many bacterial pathogens and as an entry port for pathogens to disseminate into deeper tissues. Enteric bacterial pathogens can efficiently infect the gut mucosa using highly sophisticated virulence mechanisms that allow bacteria to circumvent the defense barriers in the gut. We provide an overview of the components of the mucosal barrier and discuss the bacterial stratagems that circumvent these barriers with particular emphasis on the roles of bacterial effector proteins.


Asunto(s)
Células Epiteliales/microbiología , Interacciones Huésped-Patógeno , Mucosa Intestinal/microbiología , Animales , Humanos , Metagenoma , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA