Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 16(6): e0253242, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34129624

RESUMO

Streptococcus agalactiae (group B streptococcus or GBS) is a commensal bacterium that can frequently behave as a pathogen, particularly in the neonatal period and in the elderly. The gut is a primary site of GBS colonization and a potential port of entry during neonatal infections caused by hypervirulent clonal complex 17 (CC17) strains. Here we studied the interactions between the prototypical CC17 BM110 strain and polarized enterocytes using the Caco-2 cell line. GBS could adhere to and invade these cells through their apical or basolateral surfaces. Basolateral invasion was considerably more efficient than apical invasion and predominated under conditions resulting in weakening of cell-to-cell junctions. Bacterial internalization occurred by a mechanism involving caveolae- and lipid raft-dependent endocytosis and actin re-organization, but not clathrin-dependent endocytosis. In the first steps of Caco-2 invasion, GBS colocalized with the early endocytic marker EEA-1, to later reside in acidic vacuoles. Taken together, these data suggest that CC17 GBS selectively adheres to the lateral surface of enterocytes from which it enters through caveolar lipid rafts using a classical, actin-dependent endocytic pathway. These data may be useful to develop alternative preventive strategies aimed at blocking GBS invasion of the intestinal barrier.


Assuntos
Enterócitos/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/patogenicidade , Aderência Bacteriana , Células CACO-2/microbiologia , Endocitose , Humanos , Junções Intercelulares/microbiologia , Microscopia de Fluorescência , Streptococcus agalactiae/fisiologia , Virulência
2.
J Clin Lab Anal ; 35(2): e23627, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33070380

RESUMO

Epithelial cells are typically connected through different types of cell junctions that are localized from the apical membrane to the basal surface. In this way, epithelium cells form the first barrier against pathogenic microorganisms and prevent their entry into internal organs and the circulatory system. Recent studies demonstrate that bacterial pathogens disrupt epithelial cell junctions through targeting junctional proteins by secreted virulence factors. In this review, we discuss the diverse strategies used by common bacterial pathogens, including Pseudomonas aeruginosa, Helicobacter pylori, and enteropathogenic Escherichia coli, to disrupt epithelial cell junctions during infection. We also discuss the potential of targeting the pathogenic mechanisms in the treatment of pathogen-associated diseases.


Assuntos
Infecções Bacterianas/microbiologia , Infecções Bacterianas/patologia , Interações Hospedeiro-Patógeno , Junções Intercelulares/microbiologia , Fatores de Virulência/fisiologia , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/patogenicidade , Humanos , Junções Intercelulares/patologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/patogenicidade
3.
mBio ; 11(2)2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32291306

RESUMO

RepA is a bacterial protein that builds intracellular amyloid oligomers acting as inhibitory complexes of plasmid DNA replication. When carrying a mutation enhancing its amyloidogenesis (A31V), the N-terminal domain (WH1) generates cytosolic amyloid particles that are inheritable within a bacterial lineage. Such amyloids trigger in bacteria a lethal cascade reminiscent of mitochondrial impairment in human cells affected by neurodegeneration. To fulfill all the criteria to qualify as a prion-like protein, horizontal (intercellular) transmissibility remains to be demonstrated for RepA-WH1. Since this is experimentally intractable in bacteria, here we transiently expressed in a murine neuroblastoma cell line the soluble, barely cytotoxic RepA-WH1 wild type [RepA-WH1(WT)] and assayed its response to exposure to in vitro-assembled RepA-WH1(A31V) amyloid fibers. In parallel, murine cells releasing RepA-WH1(A31V) aggregates were cocultured with human neuroblastoma cells expressing RepA-WH1(WT). Both the assembled fibers and donor-derived RepA-WH1(A31V) aggregates induced, in the cytosol of recipient cells, the formation of cytotoxic amyloid particles. Mass spectrometry analyses of the proteomes of both types of injured cells pointed to alterations in mitochondria, protein quality triage, signaling, and intracellular traffic. Thus, a synthetic prion-like protein can be propagated to, and become cytotoxic to, cells of organisms placed at such distant branches of the tree of life as bacteria and mammalia, suggesting that mechanisms of protein aggregate spreading and toxicity follow default pathways.IMPORTANCE Proteotoxic amyloid seeds can be transmitted between mammalian cells, arguing that the intercellular exchange of prion-like protein aggregates can be a common phenomenon. RepA-WH1 is derived from a bacterial intracellular functional amyloid protein, engineered to become cytotoxic in Escherichia coli Here, we have studied if such bacterial aggregates can also be transmitted to, and become cytotoxic to, mammalian cells. We demonstrate that RepA-WH1 is capable of entering naive cells, thereby inducing the cytotoxic aggregation of a soluble RepA-WH1 variant expressed in the cytosol, following the same trend that had been described in bacteria. These findings highlight the universality of one of the central principles underlying prion biology: No matter the biological origin of a given prion-like protein, it can be transmitted to a phylogenetically unrelated recipient cell, provided that the latter expresses a soluble protein onto which the incoming protein can readily template its amyloid conformation.


Assuntos
Proteínas de Bactérias/metabolismo , Junções Intercelulares/microbiologia , Príons/metabolismo , Animais , Proteínas de Bactérias/síntese química , Linhagem Celular Tumoral , Técnicas de Cocultura , Células HeLa , Humanos , Fusão de Membrana , Camundongos , Neuroblastoma , Príons/síntese química
4.
Biochim Biophys Acta Biomembr ; 1862(6): 183237, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32126234

RESUMO

Targeting the apical junctional complex during acute bacterial infections can be detrimental for the host in several aspects. First, the rupture of the epithelium or endothelium integrity is toxic in itself. In addition, extracellular bacterial pathogens or bacterial toxins can cross the body's physical barriers using the paracellular route and induce infection or intoxication of distant organs. No single strategy has been developed to disrupt junctional structures, rather each bacterium has its own method, which can be classed in one of the following three categories: (i) proteolysis/perturbation of adhesive proteins involved in tight or adherens junctions by bacterial or toxin-activated eukaryotic proteases, (ii) manipulation of host regulatory pathways leading to weakened intercellular adhesion, or (iii) delocalization of the junctional complex to open the gateway toward the subepithelial compartment. In this review, examples of each of these mechanisms are provided to illustrate how creative bacteria can be when seeking to disrupt cell-cell junctions.


Assuntos
Bactérias/patogenicidade , Junções Intercelulares/microbiologia , Animais , Infecções Bacterianas/etiologia , Toxinas Bacterianas/farmacologia , Humanos
5.
F1000Res ; 82019.
Artigo em Inglês | MEDLINE | ID: mdl-31249676

RESUMO

Chlamydia trachomatis is the most prevalent sexually transmitted bacterial pathogen and the leading cause of preventable blindness in the developing world. C. trachomatis invades the epithelium of the conjunctiva and genital tract and replicates within an intracellular membrane-bound compartment termed the inclusion. To invade and replicate in mammalian cells, Chlamydia remodels epithelial surfaces by reorganizing the cytoskeleton and cell-cell adhesions, reprograms membrane trafficking, and modulates cell signaling to dampen innate immune responses. If the infection ascends to the upper female genital tract, it can result in pelvic inflammatory disease and tissue scarring. C. trachomatis infections are associated with infertility, ectopic pregnancies, the fibrotic disorder endometriosis, and potentially cancers of the cervix and uterus. Unfortunately, the molecular mechanisms by which this clinically important human pathogen subverts host cellular functions and causes disease have remained relatively poorly understood because of the dearth of molecular genetic tools to study Chlamydiae and limitations of both in vivo and in vitro infection models. In this review, we discuss recent advances in the experimental molecular tool kit available to dissect C. trachomatis infections with a special focus on Chlamydia-induced epithelial barrier disruption by regulating the structure, function, and dynamics of epithelial cell-cell junctions.


Assuntos
Infecções por Chlamydia/genética , Infecções por Chlamydia/patologia , Chlamydia trachomatis/patogenicidade , Infertilidade/microbiologia , Animais , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Feminino , Humanos , Imunidade Inata , Junções Intercelulares/microbiologia , Junções Intercelulares/patologia , Gravidez
6.
Environ Microbiol ; 21(3): 1086-1098, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30637902

RESUMO

Pathogenic bacteria use different strategies to infect their hosts, including the simultaneous production of pore forming toxins and several virulence factors that may synergize their pathogenic effects. However, how the pathogenic bacteria are able to break out the host intestinal barrier is poorly understood. The infectious cycle of Bacillus thuringiensis (Bt) bacterium in Caenorhabditis elegans is a powerful model system to study the early stages of the infection process. Bt produces Cry pore-forming toxins during the sporulation phase that are key virulence factors involved in its pathogenesis. In this study, we show that Bt disrupts the intestinal epithelial junctions of C. elegans at early stages of infection allowing Bt bacterium to complete its life cycle in the worm. We further confirmed that the vegetative Bt cells trigger a quorum sensing response that is activated by PlcR regulator, resulting in production of different virulence factors, such as the metalloproteinases ColB and Bmp1, that besides Cry toxins are necessary to disrupt the nematode epithelial junctions causing efficient bacterial host infection and death of the nematode. Our work provides new insights into the pathogenesis of Bt and highlights the importance of breaking down host epithelial junctions for a successful infection. A similar mechanism could be used by other pathogen-host interactions since epithelial junctions are conserved structures from insects to mammals.


Assuntos
Bacillus thuringiensis/patogenicidade , Caenorhabditis elegans/microbiologia , Animais , Proteínas de Bactérias , Interações Hospedeiro-Patógeno , Junções Intercelulares/microbiologia , Mucosa Intestinal/microbiologia , Metaloproteases/metabolismo , Percepção de Quorum , Fatores de Virulência
7.
Nihon Saikingaku Zasshi ; 72(3): 213-218, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-28845032

RESUMO

Streptococcus pyogenes is a ß-hemolytic organism responsible for a wide variety of human diseases that commonly occur as self-limiting purulent diseases of the pharynx and skin. Although the occurrence of invasive infections by S. pyogenes is rare, mortality rates remain high even with progressive medical therapy. As a prerequisite for causing the severe invasive disease, S. pyogenes must invade underlying sterile tissues by translocating across the epithelial barrier. In this study, streptolysin S and SpeB were identified as the novel factors that facilitate bacterial translocation via degradation of intercellular junctions. Furthermore, we found that S. pyogenes exploits host plasminogen for acceleration of bacterial invasion into deeper tissues via tricellular tight junctions. Here, I would like to show our study on bacterial translocation across the epithelial barrier through paracellular route.


Assuntos
Translocação Bacteriana , Epitélio/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/fisiologia , Streptococcus pyogenes/patogenicidade , Proteínas de Bactérias/fisiologia , Translocação Bacteriana/genética , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Epitélio/fisiologia , Exotoxinas/fisiologia , Humanos , Junções Intercelulares/microbiologia , Junções Intercelulares/fisiologia , Plasminogênio/metabolismo , Streptococcus pyogenes/genética , Estreptolisinas/fisiologia , Junções Íntimas/microbiologia , Junções Íntimas/fisiologia
8.
PLoS Pathog ; 13(4): e1006269, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28406994

RESUMO

Colonization and disruption of the epithelium is a major infection mechanism of mucosal pathogens. The epithelium counteracts infection by exfoliating damaged cells while maintaining the mucosal barrier function. The sexually transmitted bacterium Neisseria gonorrhoeae (GC) infects the female reproductive tract primarily from the endocervix, causing gonorrhea. However, the mechanism by which GC overcome the mucosal barrier remains elusive. Using a new human tissue model, we demonstrate that GC can penetrate into the human endocervix by inducing the exfoliation of columnar epithelial cells. We found that GC colonization causes endocervical epithelial cells to shed. The shedding results from the disassembly of the apical junctions that seal the epithelial barrier. Apical junction disruption and epithelial exfoliation increase GC penetration into the endocervical epithelium without reducing bacterial adherence to and invasion into epithelial cells. Both epithelial exfoliation and junction disruption require the activation and accumulation of non-muscle myosin II (NMII) at the apical surface and GC adherent sites. GC inoculation activates NMII by elevating the levels of the cytoplasmic Ca2+ and NMII regulatory light chain phosphorylation. Piliation of GC promotes, but the expression of a GC opacity-associated protein variant, OpaH that binds to the host surface proteins CEACAMs, inhibits GC-induced NMII activation and reorganization and Ca2+ flux. The inhibitory effects of OpaH lead to reductions in junction disruption, epithelial exfoliation, and GC penetration. Therefore, GC phase variation can modulate infection in the human endocervix by manipulating the activity of NMII and epithelial exfoliation.


Assuntos
Colo do Útero/patologia , Gonorreia/microbiologia , Junções Intercelulares/microbiologia , Miosina Tipo II/metabolismo , Neisseria gonorrhoeae/patogenicidade , Aderência Bacteriana , Cálcio/metabolismo , Colo do Útero/microbiologia , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Epitélio/microbiologia , Epitélio/patologia , Feminino , Humanos , Junções Intercelulares/patologia , Mucosa/microbiologia , Mucosa/patologia
9.
Trends Microbiol ; 25(4): 316-328, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28057411

RESUMO

Helicobacter pylori colonizes the gastric mucosa in the human stomach and represents a major risk factor for peptic ulcer disease and gastric cancer. Here, we summarize our current knowledge of the complex impact of H. pylori on manipulating host signalling networks, that is, by the cag pathogenicity island (cagPAI)-encoded type IV secretion system (T4SS). We show that H. pylori infections reflect a paradigm for interspecies contact-dependent molecular communication, which includes the disruption of cell-cell junctions and cytoskeletal rearrangements, as well as proinflammatory, cell cycle-related, proliferative, antiapoptotic, and DNA damage responses. The contribution of these altered signalling cascades to disease outcome is discussed.


Assuntos
Aderência Bacteriana/fisiologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/patologia , Helicobacter pylori/patogenicidade , Estômago/patologia , Sistemas de Secreção Tipo IV/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Junções Intercelulares/microbiologia , Úlcera Péptica/microbiologia , Transdução de Sinais/fisiologia , Estômago/microbiologia , Neoplasias Gástricas/microbiologia , Fatores de Virulência/genética
10.
J Infect Chemother ; 22(4): 216-20, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26809217

RESUMO

A type IV pilus filament, mainly composed of PilA, is retracted by the driving power generated by PilT and PilU ATPases. pilA is required for injection of type III ExoS effectors into epithelial cells thereby facilitating Pseudomonas aeruginosa penetration through the epithelial barrier by impairing the defense function of tight junctions. Here, we examined whether the pilT and pilU of the P. aeruginosa PAO1 strain are required for ExoS injection into epithelial cells. We measured the quantity of ExoS injected into epithelial cells, and found that within such cells its quantity decreased by 80% (ΔpilA strain), 75% (ΔpilT strain), and 30% (ΔpilU strain) compared with the wild-type strain. pilT deficiency decreased the disruption of human epithelial colorectal adenocarcinoma (Caco-2) cell monolayers to the same extent as that of pilA and exoS deficiency, whereas pilU deficiency decreased disruption of the monolayers less than deficiency of the other genes. pilT and pilU deficiency decreased bacterial penetration of the Caco-2 cell monolayers to the same level as pilA and exoS deficiency. Our data showed that the pilU gene expression level was reduced in the PAO1 strain after adhesion to Caco-2 cell surfaces, but the expression levels of the pilA and pilT genes did not change. We conclude that P. aeruginosa injects ExoS into cells through the function of type IV pilus retraction, and that pilT makes a greater contribution to this process than pilU.


Assuntos
ADP Ribose Transferases/metabolismo , Adenosina Trifosfatases/genética , Proteínas de Bactérias/genética , Toxinas Bacterianas/metabolismo , Células Epiteliais/microbiologia , Fímbrias Bacterianas/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Células CACO-2 , Proteínas de Fímbrias/genética , Fímbrias Bacterianas/fisiologia , Humanos , Junções Intercelulares/microbiologia , Mutação , Ocludina/metabolismo
11.
PLoS One ; 10(8): e0134342, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26244560

RESUMO

Neisseria gonorrhoeae (GC) establishes infection at the mucosal surface of the human genital tract, most of which is lined with polarized epithelial cells. GC can cause localized as well as disseminated infections, leading to various complications. GC constantly change their surface structures via phase and antigenic variation, which has been implicated as a means for GC to establish infection at various anatomic locations of male and female genital tracks. However, the exact contribution of each surface molecule to bacterial infectivity remains elusive due to their phase variation. Using a GC derivative that is genetically devoid of all opa genes (MS11∆Opa), this study shows that Opa expression interferes with GC transmigration across polarized human epithelial cells. MS11∆Opa transmigrates across polarized epithelial cells much faster and to a greater extent than MS11Opa+, while adhering at a similar level as MS11Opa+. When MS11Opa+, able to phase vary Opa expression, was inoculated, only those bacteria that turn off Opa expression transmigrate across the polarized epithelial monolayer. Similar to bacteria alone or co-cultured with non-polarized epithelial cells, MS11∆Opa fails to form large microcolonies at the apical surface of polarized epithelial cells. Apical inoculation of MS11Opa+, but not MS11∆Opa, induces the recruitment of the Opa host-cell receptor carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) to the apical junction and the vicinity of bacterial adherent sites. Our results suggest that Opa expression limits gonococcal ability to invade into subepithelial tissues by forming tight interactions with neighboring bacteria and by inducing CEACAM redistribution to cell junctions.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Polaridade Celular , Células Epiteliais/microbiologia , Neisseria gonorrhoeae/fisiologia , Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/genética , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Permeabilidade da Membrana Celular , Colo/citologia , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Interações Hospedeiro-Patógeno , Humanos , Junções Intercelulares/metabolismo , Junções Intercelulares/microbiologia , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Neisseria gonorrhoeae/genética
12.
Virulence ; 6(7): 704-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26305580

RESUMO

Periodontitis is an infectious inflammatory disease that destroys the tooth-supporting tissues. It is caused by the formation of subgingival biofilms on the surface of the tooth. Characteristic bacteria associated with subgingival biofilms are the Gram-negative anaerobes Porphyromonas gingivalis, Tannerella forsythia and Treponema denticola, collectively known as the "red complex" species. Inter-epithelial junctions ensure the barrier integrity of the gingival epithelium. This may however be disrupted by the biofilm challenge. The aim of this in vitro study was to investigate the effect of subgingival biofilms on the expression of inter-epithelial junctions by gingival epithelia, and evaluate the relative role of the red complex. Multi-layered human gingival epithelial cultures were challenged with a 10-species in vitro subgingival biofilm model, or its variant without the red complex, for 3 h and 24 h. A low-density array microfluidic card platform was then used for analyzing the expression of 62 genes encoding for tight junctions, gap junctions, adherens junctions, and desmosomes. Although there was a limited effect of the biofilms on the expression of tight, adherens and gap junctions, the expression of a number of desmosomal components was affected. In particular, Desmoglein-1 displayed a limited and transient up-regulation in response to the biofilm. In contrast, Desmocollin-2, Desmoplakin and Plakoglobin were down-regulated equally by both biofilm variants, after 24 h. In conclusion, this subgingival biofilm model may down-regulate selected desmosomal junctions in the gingival epithelium, irrespective of the presence of the "red complex." In turn, this could compromise the structural integrity of the gingival tissue, favoring bacterial invasion and chronic infection.


Assuntos
Biofilmes/crescimento & desenvolvimento , Gengiva/microbiologia , Bactérias Gram-Negativas/fisiologia , Junções Intercelulares/metabolismo , Junções Intercelulares/microbiologia , Células Cultivadas , Desmocolinas/genética , Desmocolinas/metabolismo , Desmoplaquinas/genética , Desmoplaquinas/metabolismo , Regulação para Baixo , Bactérias Gram-Negativas/crescimento & desenvolvimento , Humanos , Junções Intercelulares/genética , Periodontite/microbiologia , Transcriptoma , Regulação para Cima , gama Catenina/genética , gama Catenina/metabolismo
13.
Int J Exp Pathol ; 95(4): 271-81, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24945433

RESUMO

Weil's disease, the most severe form of leptospirosis, is characterized by jaundice, haemorrhage and renal failure. The mechanisms of jaundice caused by pathogenic Leptospira remain unclear. We therefore aimed to elucidate the mechanisms by integrating histopathological changes with serum biochemical abnormalities during the development of jaundice in a hamster model of Weil's disease. In this work, we obtained three-dimensional images of infected hamster livers using scanning electron microscope together with freeze-cracking and cross-cutting methods for sample preparation. The images displayed the corkscrew-shaped bacteria, which infiltrated the Disse's space, migrated between hepatocytes, detached the intercellular junctions and disrupted the bile canaliculi. Destruction of bile canaliculi coincided with the elevation of conjugated bilirubin, aspartate transaminase and alkaline phosphatase levels in serum, whereas serum alanine transaminase and γ-glutamyl transpeptidase levels increased slightly, but not significantly. We also found in ex vivo experiments that pathogenic, but not non-pathogenic leptospires, tend to adhere to the perijunctional region of hepatocyte couplets isolated from hamsters and initiate invasion of the intercellular junction within 1 h after co-incubation. Our results suggest that pathogenic leptospires invade the intercellular junctions of host hepatocytes, and this invasion contributes in the disruption of the junction. Subsequently, bile leaks from bile canaliculi and jaundice occurs immediately. Our findings revealed not only a novel pathogenicity of leptospires, but also a novel mechanism of jaundice induced by bacterial infection.


Assuntos
Hepatócitos/microbiologia , Junções Intercelulares/microbiologia , Icterícia/etiologia , Leptospira interrogans/fisiologia , Leptospirose/complicações , Doença de Weil/complicações , Alanina Transaminase/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Aspartato Aminotransferases/metabolismo , Translocação Bacteriana/fisiologia , Bilirrubina/metabolismo , Cricetinae , Modelos Animais de Doenças , Hepatócitos/patologia , Hepatócitos/ultraestrutura , Junções Intercelulares/patologia , Junções Intercelulares/ultraestrutura , Icterícia/metabolismo , Leptospirose/metabolismo , Masculino , Mesocricetus , Doença de Weil/metabolismo
14.
J Biol Chem ; 288(19): 13317-24, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23532847

RESUMO

BACKGROUND: Group A Streptococcus (GAS) translocates across the host epithelial barrier. RESULTS: Streptococcal pyrogenic exotoxin B (SpeB) directly cleaves junctional proteins. CONCLUSION: The proteolytic efficacy of SpeB allows GAS to translocate across the epithelial barrier. SIGNIFICANCE: SpeB-mediated dysfunction of the epithelial barrier may have important implications for not only bacterial invasion but also dissemination of other virulence factors throughout intercellular spaces. Group A Streptococcus (GAS) is an important human pathogen that possesses an ability to translocate across the epithelial barrier. In this study, culture supernatants of tested GAS strains showed proteolytic activity against human occludin and E-cadherin. Utilizing various types of protease inhibitors and amino acid sequence analysis, we identified SpeB (streptococcal pyrogenic exotoxin B) as the proteolytic factor that cleaves E-cadherin in the region neighboring the calcium-binding sites within the extracellular domain. The cleaving activities of culture supernatants from several GAS isolates were correlated with the amount of active SpeB, whereas culture supernatants from an speB mutant showed no such activities. Of note, the wild type strain efficiently translocated across the epithelial monolayer along with cleavage of occludin and E-cadherin, whereas deletion of the speB gene compromised those activities. Moreover, destabilization of the junctional proteins was apparently relieved in cells infected with the speB mutant, as compared with those infected with the wild type. Taken together, our findings indicate that the proteolytic efficacy of SpeB in junctional degradation allows GAS to invade deeper into tissues.


Assuntos
Proteínas de Bactérias/metabolismo , Translocação Bacteriana , Cisteína Endopeptidases/metabolismo , Exotoxinas/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/enzimologia , Antígenos CD , Proteínas de Bactérias/genética , Células CACO-2 , Caderinas/química , Caderinas/metabolismo , Cisteína Endopeptidases/genética , Impedância Elétrica , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Epitélio/microbiologia , Epitélio/patologia , Exotoxinas/genética , Interações Hospedeiro-Patógeno , Humanos , Junções Intercelulares/metabolismo , Junções Intercelulares/microbiologia , Estrutura Terciária de Proteína , Proteólise , Deleção de Sequência , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/isolamento & purificação , Streptococcus pyogenes/fisiologia
15.
Cell Microbiol ; 15(6): 1042-57, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23279089

RESUMO

Neisseria gonorrhoeae initiates infection at the apical surface of columnar endocervical epithelial cells in the female reproductive tract. These cells provide a physical barrier against pathogens by forming continuous apical junctional complexes between neighbouring cells. This study examines the interaction of gonococci (GC) with polarized epithelial cells. We show that viable GC preferentially localize at the apical side of the cell-cell junction in polarized endometrial and colonic epithelial cells, HEC-1-B and T84. In GC-infected cells, continuous apical junctional complexes are disrupted, and the junction-associated protein ß-catenin is redistributed from the apical junction to the cytoplasm and to GC adherent sites; however, overall cellular levels remain unchanged. This redistribution of junctional proteins is associated with a decrease in the 'fence' function of the apical junction but not its 'gate' function. Disruption of the apical junction by removing calcium increases GC transmigration across the epithelial monolayer. GC inoculation induces the phosphorylation of both epidermal growth factor receptor (EGFR) and ß-catenin, while inhibition of EGFR kinase activity significantly reduces both GC-induced ß-catenin redistribution and GC transmigration. Therefore, the gonococcus is capable of weakening the apical junction and polarity of epithelial cells by activating EGFR, which facilitates GC transmigration across the epithelium.


Assuntos
Polaridade Celular/fisiologia , Células Epiteliais/microbiologia , Receptores ErbB/fisiologia , Junções Intercelulares/microbiologia , Neisseria gonorrhoeae/fisiologia , Migração Transendotelial e Transepitelial/fisiologia , Adenocarcinoma/metabolismo , Adenocarcinoma/microbiologia , Adenocarcinoma/patologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/patologia , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/microbiologia , Neoplasias do Endométrio/patologia , Células Epiteliais/fisiologia , Feminino , Humanos , Junções Intercelulares/fisiologia , Neisseria gonorrhoeae/patogenicidade , beta Catenina/metabolismo
16.
Fungal Genet Biol ; 49(10): 830-7, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22903040

RESUMO

Global amphibian declines are in part driven by the chytrid fungus Batrachochytrium dendrobatidis, causing superficial dermatomycosis with epidermal hyperplasia and hyperkeratosis in infected amphibians. The susceptibility to chytridiomycosis and the severity of epidermal lesions in amphibians with chytridiomycosis are not consistent across species or even among individuals. Severe infections cause death of the animal most likely through disturbance of ion homeostasis. The mechanism by which this superficial skin infection results in epidermal lesions has so far eluded precise definition. It was the aim of this study to unravel how B. dendrobatidis causes alterations that affect skin integrity. Exposure of Xenopus laevis skin to B. dendrobatidis zoospore supernatant using skin explants and Ussing chambers caused rapid disruption of intercellular junctions, demonstrated using histology and transmission electron microscopy. The loss of intercellular junctions led to detachment-induced cell apoptosis, or anoikis. The zoospore supernatant induced neither apoptosis nor necrosis in isolated primary keratinocytes of X. laevis. This supports the idea that the loss of cell contacts triggered apoptosis in the skin explants. Mass spectrometric analysis of the protein composition of the supernatant revealed a complex mixture, including several new virulence associated proteins, such as proteases, biofilm-associated proteins and a carotenoid ester lipase. Protease and lipase activity of the supernatant was confirmed with a protease and lipase assay. In conclusion, B. dendrobatidis zoospores produce a complex mixture of proteins that quickly disturbs epidermal intercellular junctions leading to anoikis in the anuran skin. The role of the identified proteins in this process remains to be determined.


Assuntos
Anoikis , Quitridiomicetos/patogenicidade , Esporos Fúngicos/patogenicidade , Xenopus laevis/microbiologia , Animais , Quitridiomicetos/enzimologia , Junções Intercelulares/microbiologia , Lipase/análise , Lipase/metabolismo , Espectrometria de Massas , Microscopia Eletrônica de Transmissão , Peptídeo Hidrolases/análise , Peptídeo Hidrolases/metabolismo , Proteínas/metabolismo , Proteômica , Pele/citologia , Pele/microbiologia , Esporos Fúngicos/enzimologia , Virulência , Xenopus laevis/anatomia & histologia
17.
PLoS Pathog ; 7(5): e1002050, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21589900

RESUMO

Helicobacter pylori (Hp) injects the CagA effector protein into host epithelial cells and induces growth factor-like signaling, perturbs cell-cell junctions, and alters host cell polarity. This enables Hp to grow as microcolonies adhered to the host cell surface even in conditions that do not support growth of free-swimming bacteria. We hypothesized that CagA alters host cell physiology to allow Hp to obtain specific nutrients from or across the epithelial barrier. Using a polarized epithelium model system, we find that isogenic ΔcagA mutants are defective in cell surface microcolony formation, but exogenous addition of iron to the apical medium partially rescues this defect, suggesting that one of CagA's effects on host cells is to facilitate iron acquisition from the host. Hp adhered to the apical epithelial surface increase basolateral uptake of transferrin and induce its transcytosis in a CagA-dependent manner. Both CagA and VacA contribute to the perturbation of transferrin recycling, since VacA is involved in apical mislocalization of the transferrin receptor to sites of bacterial attachment. To determine if the transferrin recycling pathway is involved in Hp colonization of the cell surface, we silenced transferrin receptor expression during infection. This resulted in a reduced ability of Hp to colonize the polarized epithelium. To test whether CagA is important in promoting iron acquisition in vivo, we compared colonization of Hp in iron-replete vs. iron-deficient Mongolian gerbils. While wild type Hp and ΔcagA mutants colonized iron-replete gerbils at similar levels, ΔcagA mutants are markedly impaired in colonizing iron-deficient gerbils. Our study indicates that CagA and VacA act in concert to usurp the polarized process of host cell iron uptake, allowing Hp to use the cell surface as a replicative niche.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Mucosa Gástrica/microbiologia , Helicobacter pylori/fisiologia , Ferro/metabolismo , Transcitose/fisiologia , Adaptação Fisiológica , Animais , Antígenos de Bactérias/genética , Aderência Bacteriana/fisiologia , Proteínas de Bactérias/genética , Células CACO-2 , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Polaridade Celular/fisiologia , Cães , Regulação para Baixo , Epitélio/metabolismo , Epitélio/microbiologia , Mucosa Gástrica/metabolismo , Gerbillinae/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/crescimento & desenvolvimento , Helicobacter pylori/metabolismo , Helicobacter pylori/patogenicidade , Humanos , Junções Intercelulares/metabolismo , Junções Intercelulares/microbiologia , Ferro/farmacologia , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Deleção de Sequência , Transdução de Sinais , Transferrina/metabolismo
18.
J Biol Chem ; 286(4): 2750-61, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21084306

RESUMO

Group A Streptococcus pyogenes (GAS) is a human pathogen that causes local suppurative infections and severe invasive diseases. Systemic dissemination of GAS is initiated by bacterial penetration of the epithelial barrier of the pharynx or damaged skin. To gain insight into the mechanism by which GAS penetrates the epithelial barrier, we sought to identify both bacterial and host factors involved in the process. Screening of a transposon mutant library of a clinical GAS isolate recovered from an invasive episode allowed identification of streptolysin S (SLS) as a novel factor that facilitates the translocation of GAS. Of note, the wild type strain efficiently translocated across the epithelial monolayer, accompanied by a decrease in transepithelial electrical resistance and cleavage of transmembrane junctional proteins, including occludin and E-cadherin. Loss of integrity of intercellular junctions was inhibited after infection with a deletion mutant of the sagA gene encoding SLS, as compared with those infected with the wild type strain. Interestingly, following GAS infection, calpain was recruited to the plasma membrane along with E-cadherin. Moreover, bacterial translocation and destabilization of the junctions were partially inhibited by a pharmacological calpain inhibitor or genetic interference with calpain. Our data indicate a potential function of SLS that facilitates GAS invasion into deeper tissues via degradation of epithelial intercellular junctions in concert with the host cysteine protease calpain.


Assuntos
Proteínas de Bactérias/metabolismo , Junções Intercelulares/metabolismo , Mucosa Respiratória/metabolismo , Infecções Estreptocócicas/enzimologia , Streptococcus pyogenes/enzimologia , Streptococcus pyogenes/patogenicidade , Estreptolisinas/metabolismo , Proteínas de Bactérias/genética , Células CACO-2 , Caderinas/metabolismo , Calpaína/metabolismo , Humanos , Junções Intercelulares/microbiologia , Faringe/metabolismo , Faringe/microbiologia , Mucosa Respiratória/microbiologia , Infecções Estreptocócicas/genética , Streptococcus pyogenes/genética , Estreptolisinas/genética
19.
PLoS Pathog ; 6(5): e1000900, 2010 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-20485518

RESUMO

Listeria monocytogenes (Lm) uses InlA to invade the tips of the intestinal villi, a location at which cell extrusion generates a transient defect in epithelial polarity that exposes the receptor for InlA, E-cadherin, on the cell surface. As the dying cell is removed from the epithelium, the surrounding cells reorganize to form a multicellular junction (MCJ) that Lm exploits to find its basolateral receptor and invade. By examining individual infected villi using 3D-confocal imaging, we uncovered a novel role for the second major invasin, InlB, during invasion of the intestine. We infected mice intragastrically with isogenic strains of Lm that express or lack InlB and that have a modified InlA capable of binding murine E-cadherin and found that Lm lacking InlB invade the same number of villi but have decreased numbers of bacteria within each infected villus tip. We studied the mechanism of InlB action at the MCJs of polarized MDCK monolayers and find that InlB does not act as an adhesin, but instead accelerates bacterial internalization after attachment. InlB locally activates its receptor, c-Met, and increases endocytosis of junctional components, including E-cadherin. We show that MCJs are naturally more endocytic than other sites of the apical membrane, that endocytosis and Lm invasion of MCJs depends on functional dynamin, and that c-Met activation by soluble InlB or hepatocyte growth factor (HGF) increases MCJ endocytosis. Also, in vivo, InlB applied through the intestinal lumen increases endocytosis at the villus tips. Our findings demonstrate a two-step mechanism of synergy between Lm's invasins: InlA provides the specificity of Lm adhesion to MCJs at the villus tips and InlB locally activates c-Met to accelerate junctional endocytosis and bacterial invasion of the intestine.


Assuntos
Proteínas de Bactérias/metabolismo , Endocitose/fisiologia , Células Epiteliais/microbiologia , Listeria monocytogenes/metabolismo , Listeriose/microbiologia , Proteínas de Membrana/metabolismo , Animais , Caderinas/metabolismo , Linhagem Celular , Polaridade Celular/fisiologia , Cães , Dinaminas/metabolismo , Células Epiteliais/metabolismo , Feminino , Junções Intercelulares/metabolismo , Junções Intercelulares/microbiologia , Rim/citologia , Camundongos , Camundongos Endogâmicos BALB C , Microvilosidades/metabolismo , Microvilosidades/microbiologia , Receptores Proteína Tirosina Quinases/metabolismo
20.
Inflamm Bowel Dis ; 16(8): 1311-21, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20232407

RESUMO

BACKGROUND: Recent insights into the pathogenesis of Crohn's disease (CD) point to an important role of the mucosal barrier and intestinal microflora that may induce a chronic inflammation after crossing the intestinal barrier. The first detected susceptibility gene for CD, NOD2, is a pattern recognition receptor (PRR) for the recognition of the bacterial cell wall component muramyldipeptide (MDP). Binding of MDP to NOD2 is followed by activation of proinflammatory pathways mainly regulated by nuclear factor kappa B (NF-kappaB). In this study we investigated whether impaired recognition of MDP via NOD2 variants is associated with increased bacterial translocation across the epithelial barrier and whether this is followed by increased or decreased NF-kappaB activation. METHODS: NOD2 variants were analyzed in 36 CD patients and 30 controls. Endotoxin was stained by immunohistochemistry in 30 intestinal biopsies from patients carrying NOD2 variants (NOD2-mut) or being NOD2 wildtype (WT). Junctional proteins were visualized by immunofluorescence and quantified by Western blotting. NF-kappaB activation was analyzed by immunohistochemistry in specimens from NOD2-WT and NOD2-mut CD and control patients. RESULTS: We demonstrated the increased presence of endotoxin in the mucosal lamina propria of CD patients carrying NOD2 variants. This was associated with an altered composition of epithelial cell-cell contacts. Patients carrying NOD2 variants displayed increased NF-kappaB activation in the mucosa. CONCLUSIONS: This study for the first time demonstrates that translocation of luminal bacteria and/or bacterial products into the intestinal mucosa is increased in patients carrying NOD2 variants, leading to higher activation of proinflammatory signaling cascades.


Assuntos
Translocação Bacteriana , Doença de Crohn/imunologia , Doença de Crohn/microbiologia , Proteína Adaptadora de Sinalização NOD2/genética , Acetilmuramil-Alanil-Isoglutamina/análise , Caderinas/análise , Claudinas/análise , Endotoxinas/análise , Humanos , Junções Intercelulares/imunologia , Junções Intercelulares/microbiologia , Interleucina-8/biossíntese , Proteínas de Membrana/análise , Mucosa/imunologia , Mucosa/microbiologia , NF-kappa B/análise , Ocludina , beta Catenina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...