Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 12(1): 11662, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35804072

RESUMEN

Neuronal ceroid lipofuscinosis is one of many neurodegenerative storage diseases characterized by excessive accumulation of lipofuscins. CLN10 disease, an early infantile neuronal ceroid lipofuscinosis, is associated with a gene that encodes cathepsin D (CtsD), one of the major lysosomal proteases. Whole body CtsD-knockout mice show neurodegenerative phenotypes with the accumulation of lipofuscins in the brain and also show defects in other tissues including intestinal necrosis. To clarify the precise role of CtsD in the central nervous system (CNS), we generated a CNS-specific CtsD-knockout mouse (CtsD-CKO). CtsD-CKO mice were born normally but developed seizures and their growth stunted at around postnatal day 23 ± 1. CtsD-CKO did not exhibit apparent intestinal symptoms as those observed in whole body knockout. Histologically, autofluorescent materials were detected in several areas of the CtsD-CKO mouse's brain, including: thalamus, cerebral cortex, hippocampus, and cerebellum. Expression of ubiquitin and autophagy-associated proteins was also increased, suggesting that the autophagy-lysosome system was impaired. Microglia and astrocytes were activated in the CtsD-CKO thalamus, and inducible nitric oxide synthase (iNOS), an inflammation marker, was increased in the microglia. Interestingly, deposits of proteinopathy-related proteins, phosphorylated α-synuclein, and Tau protein were also increased in the thalamus of CtsD-CKO infant mice. Considering these results, we propose thatt the CtsD-CKO mouse is a useful mouse model to investigate the contribution of cathepsin D to the early phases of neurodegenerative diseases in relation to lipofuscins, proteinopathy-related proteins and activation of microglia and astrocytes.


Asunto(s)
Catepsina D/metabolismo , Lipofuscinosis Ceroideas Neuronales , Animales , Astrocitos/metabolismo , Catepsina D/genética , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Microglía/metabolismo , Lipofuscinosis Ceroideas Neuronales/patología
2.
Sci Rep ; 12(1): 11130, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35778550

RESUMEN

Biotin ligases have been developed as proximity biotinylation enzymes for analyses of the interactome. However, there has been no report on the application of proximity labeling for in-resin correlative light-electron microscopy of Epon-embedded cells. In this study, we established a proximity-labeled in-resin CLEM of Epon-embedded cells using miniTurbo, a biotin ligase. Biotinylation by miniTurbo was observed in cells within 10 min following the addition of biotin to the medium. Using fluorophore-conjugated streptavidin, intracellular biotinylated proteins were labeled after fixation of cells with a mixture of paraformaldehyde and glutaraldehyde. Fluorescence of these proteins was resistant to osmium tetroxide staining and was detected in 100-nm ultrathin sections of Epon-embedded cells. Ultrastructures of organelles were preserved well in the same sections. Fluorescence in sections was about 14-fold brighter than that in the sections of Epon-embedded cells expressing mCherry2 and was detectable for 14 days. When mitochondria-localized miniTurbo was expressed in the cells, mitochondria-like fluorescent signals were detected in the sections, and ultrastructures of mitochondria were observed as fluorescence-positive structures in the same sections by scanning electron microscopy. Proximity labeling using miniTurbo led to more stable and brighter fluorescent signals in the ultrathin sections of Epon-embedded cells, resulting in better performance of in-resin CLEM.


Asunto(s)
Biotina , Tetróxido de Osmio , Microscopía Electrónica de Rastreo , Orgánulos/ultraestructura , Resinas de Plantas , Coloración y Etiquetado
3.
Nat Commun ; 12(1): 2085, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33837194

RESUMEN

Long-term infection of the stomach with Helicobacter pylori can cause gastric cancer. However, the mechanisms by which the bacteria adapt to the stomach environment are poorly understood. Here, we show that a small non-coding RNA of H. pylori (HPnc4160, also known as IsoB or NikS) regulates the pathogen's adaptation to the host environment as well as bacterial oncoprotein production. In a rodent model of H. pylori infection, the genomes of bacteria isolated from the stomach possess an increased number of T-repeats upstream of the HPnc4160-coding region, and this leads to reduced HPnc4160 expression. We use RNA-seq and iTRAQ analyses to identify eight targets of HPnc4160, including genes encoding outer membrane proteins and oncoprotein CagA. Mutant strains with HPnc4160 deficiency display increased colonization ability of the mouse stomach, in comparison with the wild-type strain. Furthermore, HPnc4160 expression is lower in clinical isolates from gastric cancer patients than in isolates derived from non-cancer patients, while the expression of HPnc4160's targets is higher in the isolates from gastric cancer patients. Therefore, the small RNA HPnc4160 regulates H. pylori adaptation to the host environment and, potentially, gastric carcinogenesis.


Asunto(s)
Adaptación Fisiológica/genética , Infecciones por Helicobacter/patología , Helicobacter pylori/fisiología , ARN Bacteriano/metabolismo , ARN Pequeño no Traducido/metabolismo , Neoplasias Gástricas/microbiología , Animales , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Carcinogénesis , Modelos Animales de Enfermedad , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Regulación Bacteriana de la Expresión Génica/fisiología , Genoma Bacteriano/genética , Gerbillinae , Infecciones por Helicobacter/microbiología , Helicobacter pylori/aislamiento & purificación , Helicobacter pylori/patogenicidad , Interacciones Microbiota-Huesped , Humanos , Masculino , Mutación , ARN Bacteriano/genética , ARN Pequeño no Traducido/genética , RNA-Seq , Neoplasias Gástricas/patología
4.
Biochem Biophys Res Commun ; 525(3): 806-811, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32164943

RESUMEN

Helicobacter pylori, a pathogenic bacterium that colonizes in the human stomach, harbors DNA repair genes to counter the gastric environment during chronic infection. In addition, H. pylori adapts to the host environment by undergoing antigenic phase variation caused by genomic mutations. The emergence of mutations in nucleotide sequences is one of the major factors underlying drug resistance and genetic diversity in bacteria. However, it is not clear how DNA repair genes contribute to driving the genetic change of H. pylori during chronic infection. To elucidate the physiological roles of DNA repair genes, we generated DNA repair-deficient strains of H. pylori (ΔuvrA, ΔuvrB, ΔruvA, Δnth, ΔmutY, ΔmutS, and Δung). We performed susceptibility testing to rifampicin in vitro and found that ΔmutY exhibited the highest mutation frequency among the mutants. The number of bacteria colonizing the stomach was significantly lower with ΔmutY strain compared with wild-type strains in a Mongolian gerbil model of H. pylori infection. Furthermore, we performed a genomic sequence analysis of the strains isolated from the Mongolian gerbil stomachs eight weeks after infection. We found that the isolated ΔmutY strains exhibited a high frequency of spontaneous G:C to T:A mutations. However, the frequency of phase variations in the ΔmutY strain was almost similar to the wild-type strain. These results suggest that MutY may play a role in modes of gastric environmental adaptation distinct from phase variation.


Asunto(s)
Adaptación Fisiológica , ADN Glicosilasas/genética , Helicobacter pylori/genética , Mutación/genética , Estómago/microbiología , Animales , Proteínas Bacterianas/genética , Reparación del ADN/genética , Modelos Animales de Enfermedad , Gerbillinae , Infecciones por Helicobacter/microbiología , Helicobacter pylori/crecimiento & desarrollo , Tasa de Mutación , FN-kappa B/metabolismo
5.
Sci Rep ; 10(1): 3251, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32094510

RESUMEN

Group A Streptococcus (GAS) secretes deoxyribonucleases and evades neutrophil extracellular killing by degrading neutrophil extracellular traps (NETs). However, limited information is currently available on the interaction between GAS and NETs in the pathogenicity of GAS pharyngitis. In this study, we modified a mouse model of GAS pharyngitis and revealed an essential role for DNase in this model. After intranasal infection, the nasal mucosa was markedly damaged near the nasal cavity, at which GAS was surrounded by neutrophils. When neutrophils were depleted from mice, GAS colonization and damage to the nasal mucosa were significantly decreased. Furthermore, mice infected with deoxyribonuclease knockout GAS mutants (∆spd, ∆endA, and ∆sdaD2) survived significantly better than those infected with wild-type GAS. In addition, the supernatants of digested NETs enhanced GAS-induced cell death in vitro. Collectively, these results indicate that NET degradation products may contribute to the establishment of pharyngeal infection caused by GAS.


Asunto(s)
ADN/química , Trampas Extracelulares , Faringitis/microbiología , Faringe/microbiología , Infecciones Estreptocócicas/patología , Animales , Apoptosis , Desoxirribonucleasas/metabolismo , Modelos Animales de Enfermedad , Humanos , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Neutrófilos/microbiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Streptococcus pyogenes
6.
Microbiol Immunol ; 62(4): 221-228, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29446491

RESUMEN

Helicobacter pylori (H. pylori), a gram-negative microaerophilic bacterial pathogen that colonizes the stomachs of more than half of all humans, is linked to chronic gastritis, peptic ulcers and gastric cancer. Spiral-shaped H. pylori undergo morphologic conversion to a viable but not culturable coccoid form when they transit from the microaerobic stomach into the anaerobic intestinal tract. However, little is known about the morphological and pathogenic characteristics of H. pylori under prolonged anaerobic conditions. In this study, scanning electron microscopy was used to document anaerobiosis-induced morphological changes of H. pylori, from helical to coccoid to a newly defined fragmented form. Western blot analysis indicated that all three forms express certain pathogenic proteins, including the bacterial cytotoxin-associated gene A (CagA), components of the cag-Type IV secretion system (TFSS), the blood group antigen-binding adhesin BabA, and UreA (an apoenzyme of urease), almost equally. Similar urease activities were also detected in all three forms of H. pylori. However, in contrast to the helical form, bacterial motility and TFSS activity were found to have been abrogated in the anaerobiosis-induced coccoid and fragmented forms of H. pylori. Notably, it was demonstrated that some of the anaerobiosis-induced fragmented state cells could be converted to proliferation-competent helical bacteria in vitro. These results indicate that prolonged exposure to the anaerobic intestine may not eliminate the potential for H. pylori to revert to the helical pathogenic state.


Asunto(s)
Proteínas Bacterianas/genética , Helicobacter pylori/citología , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Anaerobiosis , Antibacterianos , Antígenos Bacterianos/genética , Línea Celular , Proliferación Celular , Regulación Bacteriana de la Expresión Génica , Infecciones por Helicobacter/microbiología , Humanos , Microscopía Electrónica de Rastreo , Sistemas de Secreción Tipo IV/genética , Ureasa/genética , Factores de Virulencia/genética
7.
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
8.
Nat Commun ; 5: 4497, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25187177

RESUMEN

Persistent colonization of the gastric mucosa by Helicobacter pylori (Hp) elicits chronic inflammation and aberrant epithelial cell proliferation, which increases the risk of gastric cancer. Here we examine the ability of microRNAs to modulate gastric cell proliferation in response to persistent Hp infection and find that epigenetic silencing of miR-210 plays a key role in gastric disease progression. Importantly, DNA methylation of the miR-210 gene is increased in Hp-positive human gastric biopsies as compared with Hp-negative controls. Moreover, silencing of miR-210 in gastric epithelial cells promotes proliferation. We identify STMN1 and DIMT1 as miR-210 target genes and demonstrate that inhibition of miR-210 expression augments cell proliferation by activating STMN1 and DIMT1. Together, our results highlight inflammation-induced epigenetic silencing of miR-210 as a mechanism of induction of chronic gastric diseases, including cancer, during Hp infection.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Infecciones por Helicobacter/genética , Metiltransferasas/metabolismo , MicroARNs/metabolismo , Estatmina/metabolismo , Neoplasias Gástricas/genética , Animales , Ciclo Celular/genética , Proliferación Celular , Enfermedad Crónica , Metilación de ADN , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/patología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Perfilación de la Expresión Génica , Gerbillinae , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Helicobacter pylori/fisiología , Humanos , Masculino , Metiltransferasas/genética , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Cultivo Primario de Células , Regiones Promotoras Genéticas , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Estatmina/genética , Neoplasias Gástricas/complicaciones , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología
9.
Cell Host Microbe ; 13(5): 570-583, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23684308

RESUMEN

Caspase-mediated inflammatory cell death acts as an intrinsic defense mechanism against infection. Bacterial pathogens deploy countermeasures against inflammatory cell death, but the mechanisms by which they do this remain largely unclear. In a screen for Shigella flexneri effectors that regulate cell death during infection, we discovered that Shigella infection induced acute inflammatory, caspase-4-dependent epithelial cell death, which is counteracted by the bacterial OspC3 effector. OspC3 interacts with the caspase-4-p19 subunit and inhibits its activation by preventing caspase-4-p19 and caspase-4-p10 heterodimerization by depositing the conserved OspC3 X1-Y-X2-D-X3 motif at the putative catalytic pocket of caspase-4. Infection of guinea pigs with a Shigella ospC3-deficient mutant resulted in enhanced inflammatory cell death and associated symptoms, correlating with decreased bacterial burdens. Salmonella Typhimurium and enteropathogenic Escherichia coli infection also induced caspase-4-dependent epithelial death. These findings highlight the importance of caspase-4-dependent innate immune responses and demonstrate that Shigella delivers a caspase-4-specific inhibitor to delay epithelial cell death and promote infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Caspasas Iniciadoras/metabolismo , Muerte Celular , Inhibidores Enzimáticos/metabolismo , Células Epiteliales/microbiología , Interacciones Huésped-Patógeno , Shigella flexneri/patogenicidad , Animales , Proteínas Bacterianas/genética , Línea Celular , ADN Bacteriano/química , ADN Bacteriano/genética , Modelos Animales de Enfermedad , Disentería Bacilar/inmunología , Disentería Bacilar/microbiología , Disentería Bacilar/patología , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Técnicas de Inactivación de Genes , Cobayas , Humanos , Datos de Secuencia Molecular , Unión Proteica , Mapeo de Interacción de Proteínas , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Análisis de Secuencia de ADN , Shigella flexneri/genética , Shigella flexneri/inmunología , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
10.
J Mol Biol ; 425(15): 2623-31, 2013 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-23542009

RESUMEN

Ubc13 is a ubiquitin-conjugating enzyme that plays a key role in the nuclear factor-κB signal transduction pathway in human diseases. The Shigella flexneri effector OspI affects inflammatory responses by catalyzing the deamidation of a specific glutamine residue at position 100 in Ubc13 during infection. This modification prevents the activation of the TNF (tumor necrosis factor) receptor-associated factor 6, leading to modulation of the diacylglycerol-CBM (CARD-Bcl10-Malt1) complex-TNF receptor-associated factor 6-nuclear factor-κB signaling pathway. To elucidate the structural basis of OspI function, we determined the crystal structures of the catalytically inert OspI C62A mutant and its complex with Ubc13 at resolutions of 3.0 and 2.96Å, respectively. The structure of the OspI-Ubc13 complex revealed that the interacting surfaces between OspI and Ubc13 are a hydrophobic surface and a complementary charged surface. Furthermore, we predict that the complementary charged surface of OspI plays a key role in substrate specificity determination.


Asunto(s)
Shigella flexneri/química , Shigella flexneri/metabolismo , Enzimas Ubiquitina-Conjugadoras/química , Enzimas Ubiquitina-Conjugadoras/metabolismo , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estructura Cuaternaria de Proteína , Electricidad Estática
12.
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
13.
J Cell Biol ; 195(6): 931-42, 2011 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-22123830

RESUMEN

Host cell death is an intrinsic immune defense mechanism in response to microbial infection. However, bacterial pathogens use many strategies to manipulate the host cell death and survival pathways to enhance their replication and survival. This manipulation is quite intricate, with pathogens often suppressing cell death to allow replication and then promoting it for dissemination. Frequently, these effects are exerted through modulation of the mitochondrial pro-death, NF-κB-dependent pro-survival, and inflammasome-dependent host cell death pathways during infection. Understanding the molecular details by which bacterial pathogens manipulate cell death pathways will provide insight into new therapeutic approaches to control infection.


Asunto(s)
Apoptosis/inmunología , Infecciones Bacterianas/inmunología , Interacciones Huésped-Patógeno/inmunología , Fagocitos/inmunología , Animales , Infecciones Bacterianas/patología , Humanos , Inflamasomas/inmunología , Ratones , Mitocondrias/inmunología , Necrosis , Fagocitos/microbiología
14.
J Biol Chem ; 286(34): 29964-72, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21757722

RESUMEN

Population genetic analyses of bacterial genes whose products interact with host tissues can give new understanding of infection and disease processes. Here we show that strains of the genetically diverse gastric pathogen Helicobacter pylori from Amerindians from the remote Peruvian Amazon contain novel alleles of cagA, a major virulence gene, and reveal distinctive properties of their encoded CagA proteins. CagA is injected into the gastric epithelium where it hijacks pleiotropic signaling pathways, helps Hp exploit its special gastric mucosal niche, and affects the risk that infection will result in overt gastroduodenal diseases including gastric cancer. The Amerindian CagA proteins contain unusual but functional tyrosine phosphorylation motifs and attenuated CRPIA motifs, which affect gastric epithelial proliferation, inflammation, and bacterial pathogenesis. Amerindian CagA proteins induced less production of IL-8 and cancer-associated Mucin 2 than did those of prototype Western or East Asian strains and behaved as dominant negative inhibitors of action of prototype CagA during mixed infection of Mongolian gerbils. We suggest that Amerindian cagA is of relatively low virulence, that this may have been selected in ancestral strains during infection of the people who migrated from Asia into the Americas many thousands of years ago, and that such attenuated CagA proteins could be useful therapeutically.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Evolución Molecular , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Helicobacter pylori/patogenicidad , Factores de Virulencia/metabolismo , Alelos , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Femenino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Gerbillinae , Infecciones por Helicobacter/genética , Helicobacter pylori/genética , Humanos , Indígenas Sudamericanos , Interleucina-8/genética , Interleucina-8/metabolismo , Masculino , Datos de Secuencia Molecular , Mucina 2/genética , Mucina 2/metabolismo , Perú , Fosforilación , Transducción de Señal/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiología , Factores de Virulencia/genética
15.
Curr Opin Immunol ; 23(4): 448-55, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21763117

RESUMEN

The intestinal mucosa is equipped with multiple innate immune defense systems that sense bacterial infection, transmit alarm signals to the immune system, defeat intruding bacteria, and renew damaged and aging epithelial cells. Nevertheless, mucosal bacterial pathogens have versatile pathogenic mechanisms that modulate the host inflammatory and immune responses, manipulate host cell death and survival signal pathways, and renovate the injured epithelium. These properties enable pathogens to adapt to the intestinal mucosal environment, exploit cellular and immune functions, and facilitate infection. Here we review current topics on host defense mechanisms against bacterial infection and the countermeasures that Shigella use to evade the innate immune system.


Asunto(s)
Disentería Bacilar/inmunología , Interacciones Huésped-Patógeno , Inmunidad Innata/inmunología , Mucosa Intestinal/microbiología , Shigella/fisiología , Actinas/fisiología , Animales , Péptidos Catiónicos Antimicrobianos/fisiología , Autofagia , Citoplasma/microbiología , Disentería Bacilar/microbiología , Células Epiteliales/microbiología , Células Epiteliales/patología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inflamación , Macrófagos/microbiología , Macrófagos/patología , Modelos Biológicos , Necrosis , Neutrófilos/fisiología , Procesamiento Proteico-Postraduccional , Septinas/fisiología , Shigella/inmunología , Shigella/ultraestructura , Células Th17/fisiología
16.
Curr Opin Microbiol ; 14(1): 16-23, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20934372

RESUMEN

Although the intestinal epithelium is equipped with multiple defense systems that sense bacterial components, transmit alarms to the immune system, clear the bacteria, and renew the injured epithelial lining, mucosal bacterial pathogens are capable of efficiently colonizing the intestinal epithelium, because they have evolved systems that modulate the inflammatory and immune responses of the host and exploit the harmful environments as replicative niches. In this review we highlight current topics concerning Shigella's tactics that interfere with the innate immune systems.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Shigella/fisiología , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Regulación de la Expresión Génica , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Shigella/inmunología , alfa-Defensinas/genética , alfa-Defensinas/metabolismo
17.
J Biol Chem ; 283(49): 33858-64, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18849341

RESUMEN

FLN29 was identified as an interferon (IFN)-inducible gene, and it has been shown to suppress Toll-like receptor 4-mediated NF-kappaB activation by binding to TRAF6. To elucidate the physiological roles of FLN29, we generated FLN29-deficient mice. FLN29 deficiency resulted in hyper-response to LPS both in vivo and in vitro, demonstrating the negative regulatory role of FLN29 in TLR4 signaling. Furthermore, we found that FLN29(-/-) mice exhibited increased susceptibility to poly(I:C)-induced septic shock compared with WT mice. FLN29(-/-) fibroblasts were highly resistant to vesicular stomatitis virus infection, and these cells produced more IFN-beta than WT cells did in response to not only intracellular poly(I:C) but also overexpression of IPS-1. Forced expression of FLN29 inhibited the IPS-1-dependent activation of both NF-kappaB and IRF3. We also found that FLN29 could interact with TRIF, IPS-1, TRAF3, and TRAF6. Together, these results suggest that FLN29, in addition to playing a negative regulatory role in the TLR4 signaling pathway, negatively regulates the RIG-I-like helicase signaling pathway at the level of IPS-1/TRAF6 and IPS-1/TRAF3 complexes.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/química , Humanos , Factor 3 Regulador del Interferón/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Lipopolisacáridos/metabolismo , Proteínas de la Membrana/química , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/química , Receptores de Superficie Celular , Receptores Inmunológicos , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Vesiculovirus/metabolismo
18.
Gastroenterology ; 131(1): 179-93, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16831601

RESUMEN

BACKGROUND & AIMS: A recent study has suggested that the methylation silencing of the suppressor of cytokine signaling-3 (SOCS3), a negative regulator of interleukin-6-related cytokines, could be involved in hepatocellular carcinoma (HCC). However, the roles of SOCS3 in hepatocellular carcinogenesis and hepatitis have not been established. We investigated the effect of deleting the SOCS3 gene on the development of hepatitis and HCC in hepatitis C virus-infected patients and mouse models. METHODS: The expression of SOCS genes in HCC and non-HCC regions of patient samples was determined by real-time reverse-transcription polymerase chain reaction and immunoblotting. The conditional knockout approach in mice was used to determine the hepatocyte-specific roles of SOCS3. To generate a liver-specific deletion, floxed SOCS3 (SOCS3(fl/fl)) mice were crossed with albumin-Cre transgenic mice. Hepatitis and HCC were induced by administering concanavalin A and diethylnitrosamine, respectively. RESULTS: SOCS3 expression was reduced in the HCC regions compared with the non-HCC regions. Carcinogen-induced hepatic tumor development was enhanced by deletion of the SOCS3 gene, which was associated with higher levels of the targets of signal transducers and activators of transcription (ie, B-cell lymphoma-XL, B-cell lymphoma-2, C-myelocytomatosis, cyclin D1, and vascular endothelial growth factor). In the concanavalin A-mediated hepatitis model, deletion of the SOCS3 gene in the hepatocytes protected against liver injury through suppression of interferon-gamma signaling and induction of the antiapoptotic protein Bcl-XL. CONCLUSIONS: Deletion of the SOCS3 gene in hepatocytes promotes the activation of STAT3, resistance to apoptosis, and an acceleration of proliferation, resulting in enhanced hepatitis-induced hepatocarcinogenesis.


Asunto(s)
Carcinoma Hepatocelular/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/complicaciones , Eliminación de Gen , Hepatocitos/patología , Neoplasias Hepáticas/genética , ARN Neoplásico/genética , Proteínas Supresoras de la Señalización de Citocinas/genética , Adulto , Anciano , Animales , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Concanavalina A/toxicidad , Femenino , Hepatocitos/efectos de los fármacos , Humanos , Inmunohistoquímica , Técnicas In Vitro , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/patología , Masculino , Ratones , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 3 Supresora de la Señalización de Citocinas
19.
J Exp Med ; 203(6): 1391-7, 2006 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-16717119

RESUMEN

Approximately 20% of human cancers are estimated to develop from chronic inflammation. Recently, the NF-kappaB pathway was shown to play an essential role in promoting inflammation-associated cancer, but the role of the JAK/STAT pathway, another important signaling pathway of proinflammatory cytokines, remains to be investigated. Suppressor of cytokine signaling-1 (SOCS1) acts as an important physiological regulator of cytokine responses, and silencing of the SOCS1 gene by DNA methylation has been found in several human cancers. Here, we demonstrated that SOCS1-deficient mice (SOCS1-/- Tg mice), in which SOCS1 expression was restored in T and B cells on a SOCS1-/- background, spontaneously developed colorectal carcinomas carrying nuclear beta-catenin accumulation and p53 mutations at 6 months of age. However, interferon (IFN)gamma-/- SOCS1-/- mice and SOCS1-/- Tg mice treated with anti-IFNgamma antibody did not develop such tumors. STAT3 and NF-kappaB activation was evident in SOCS1-/- Tg mice, but these were not sufficient for tumor development because these are also activated in IFNgamma-/- SOCS1-/- mice. However, colons of SOCS1-/- Tg mice, but not IFNgamma-/- SOCS1-/- mice, showed hyperactivation of STAT1, which resulted in the induction of carcinogenesis-related enzymes, cyclooxygenase-2 and inducible nitric oxide synthase. These data strongly suggest that SOCS1 is a unique antioncogene which prevents chronic inflammation-mediated carcinogenesis by regulation of the IFNgamma/STAT1 pathways.


Asunto(s)
Neoplasias del Colon/inmunología , Neoplasias Colorrectales/inmunología , Interferón gamma/toxicidad , Factor de Transcripción STAT1/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/deficiencia , Animales , Proteínas Portadoras/genética , Neoplasias del Colon/genética , Neoplasias Colorrectales/genética , Interferón gamma/deficiencia , Interferón gamma/genética , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética
20.
Biochem Biophys Res Commun ; 344(3): 1023-30, 2006 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-16643855

RESUMEN

Toll-like receptors (TLRs) play an important role as a sensor of microbial pathogens in the innate immune response. TLRs transmit signals through the recruitment of adaptor proteins including tumor necrosis factor-associated factor 6 (TRAF6), which mediates the activation of IkappaB kinase (IKK). TIFA (TRAF-interacting protein with a forkhead-associated (FHA) domain) has been shown to bind to TRAF6 and activate IKK by promoting the oligomerization and ubiquitin-ligase activity of TRAF6. FHA domains preferentially bind to phospho-threonine residues in their targets. Here, we identified a novel zinc finger protein, ZCCHC11, that interacts with TIFA from phosphoproteins of a macrophage cell line, RAW 264.7, by using affinity purification with GST-TIFA and mass spectrometric analysis. By a search of the EST database, we found a 200kDa full-length form (ZCCHC11L). ZCCHC11L was mostly located to the nucleus, but translocated into the cytoplasm in response to LPS and bound to TIFA. Overexpression and knockdown by siRNA indicated that ZCCHC11 functions as a negative regulator of TLR-mediated NF-kappaB activation. The N-terminal region (ZCCHC11S) including C2H2-type [corrected] Zn-finger motif was sufficient for suppression of NF-kappaB. We propose that ZCCHC11 is a unique TLR signal regulator, which interacts with TIFA after LPS treatment and suppresses the TRAF6-dependent activation of NF-kappaB.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Riñón/metabolismo , Macrófagos/metabolismo , Transducción de Señal/fisiología , Fracciones Subcelulares/metabolismo , Receptores Toll-Like/metabolismo , Animales , Línea Celular , Humanos , Ratones , Dedos de Zinc/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA