Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 3804, 2023 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-37365163

RESUMO

The interleukin-1 family members, IL-1ß and IL-18, are processed into their biologically active forms by multi-protein complexes, known as inflammasomes. Although the inflammasome pathways that mediate IL-1ß processing in myeloid cells have been defined, those involved in IL-18 processing, particularly in non-myeloid cells, are still not well understood. Here we report that the host defence molecule NOD1 regulates IL-18 processing in mouse epithelial cells in response to the mucosal pathogen, Helicobacter pylori. Specifically, NOD1 in epithelial cells mediates IL-18 processing and maturation via interactions with caspase-1, instead of the canonical inflammasome pathway involving RIPK2, NF-κB, NLRP3 and ASC. NOD1 activation and IL-18 then help maintain epithelial homoeostasis to mediate protection against pre-neoplastic changes induced by gastric H. pylori infection in vivo. Our findings thus demonstrate a function for NOD1 in epithelial cell production of bioactive IL-18 and protection against H. pylori-induced pathology.


Assuntos
Células Epiteliais , Infecções por Helicobacter , Interleucina-18 , Proteína Adaptadora de Sinalização NOD1 , Animais , Camundongos , Células Epiteliais/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Transdução de Sinais , Proteína Adaptadora de Sinalização NOD1/metabolismo
2.
Mucosal Immunol ; 12(3): 720-732, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30651577

RESUMO

Inflammatory bowel disease (IBD) etiology involves genetic susceptibility, environmental triggers, and the gut microbiome. Antibiotic exposure is associated with IBD, both in early life and adulthood. Here, we investigated whether Nod2-deficiency influenced response of the gut microbiota to antibiotics and subsequent colitis susceptibility. Wild-type and Nod2-/- littermate mice were treated with amoxicillin as adults or neonates, and fecal samples were collected for 16S rRNA sequencing. Five weeks after antibiotic exposure, dextran sulfate sodium (DSS) colitis was induced. Antibiotic treatment altered the microbiota of adult WT and Nod2-/- mice, but recovery was delayed in Nod2-/- mice. Neonatal antibiotic treatment significantly changed the microbiota at weaning in WT and Nod2-/- littermates; however, Nod2-/- mice maintained reduced microbial diversity 14 days after cessation of antibiotics. Although treatment of adult mice did not influence susceptibility to colitis, neonatally treated Nod2-/- mice developed a more severe colitis. Moreover, the colitis phenotype was transferable through fecal transplantation into germ-free Nod2-/- recipients, and was associated with changes in intestinal T cells and the cytokine milieu following inflammation. These data demonstrate that neonatal antibiotic exposure has long-lasting influence on the microbiota and mucosal immunity, and may explain how NOD2 contributes to the risk of intestinal inflammation.


Assuntos
Amoxicilina/efeitos adversos , Antibacterianos/efeitos adversos , Colite/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Doenças Inflamatórias Intestinais/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Amoxicilina/administração & dosagem , Animais , Animais Recém-Nascidos , Antibacterianos/administração & dosagem , Colite/genética , Modelos Animais de Doenças , Suscetibilidade a Doenças , Transplante de Microbiota Fecal , Microbioma Gastrointestinal/fisiologia , Interação Gene-Ambiente , Humanos , Doenças Inflamatórias Intestinais/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Adaptadora de Sinalização NOD2/genética , Risco
3.
Mucosal Immunol ; 10(3): 775-788, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27600308

RESUMO

Although we know a great deal about which types of dendritic cells (DCs) promote T-cell priming in the periphery, less is known about which DC subset(s) provoke antiviral responses within the gut. Here we report that conventional Zbtb46-dependent DCs were critically required for antiviral CD8+ T-cell responses against rotavirus (RV), the major cause of childhood gastroenteritis worldwide. Furthermore, we found that in adult mice, Batf3-dependent DCs were required for generating optimal RV-specific CD8+ T-cell responses. However, in contrast to mice that lack Zbtb46-dependent DCs, a significant amount of interferon gamma-producing RV-specific CD8+ T cells were still detected in the small intestine of RV-infected adult Batf3-/- mice, suggesting the existence of compensatory cross-presentation mechanisms in the absence of Batf3-dependent DCs. In contrast to adult mice, we found that Batf3-dependent DCs were absolutely required for generating RV-specific CD8+ T-cell responses in neonates. Loss of Batf3-dependent DCs also resulted in a skewed polyclonal CD4+ T-cell response in both adult and neonatal mice upon RV infection, although local and systemic RV-specific immunoglobulin A production kinetics and titers were unimpaired. Our results provide insights that inform early-life vaccination strategies against RV infection.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Gastroenterite/virologia , Intestinos/imunologia , Proteínas Repressoras/metabolismo , Infecções por Rotavirus/imunologia , Rotavirus/imunologia , Animais , Animais Recém-Nascidos , Antígenos CD/genética , Antígenos Virais/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/genética , Células Cultivadas , Criança , Apresentação Cruzada , Humanos , Imunidade Celular , Intestinos/virologia , Lectinas Tipo C/genética , Lectinas de Ligação a Manose/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Clin Pharmacol Ther ; 99(6): 570-4, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27160649

RESUMO

The human microbiota is the ecological community of microorganisms that live within our bodies. Emerging evidence has revealed that dysregulation of the host-microbe symbiotic relationship contributes to the pathogenesis of a vast number of human diseases and impacts the efficacy and toxicity of therapeutic drugs. Therefore, a deeper understanding of the human microbiota is crucial to the development of therapeutic interventions that target the microbiota and also provides fundamental insights towards understanding intersubject variability in therapeutic outcomes.


Assuntos
Microbiota , Dieta , Humanos , Microbiota/efeitos dos fármacos , Farmacologia Clínica/métodos , Farmacologia Clínica/tendências
5.
Mol Immunol ; 46(13): 2647-54, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19535145

RESUMO

The WD40 repeat containing angio-associated migratory cell protein (AAMP) was identified as a new binding partner of the human nucleotide-binding domain, leucine rich repeat containing (NLR) family member Nod2 in a yeast two-hybrid screen. Co-immunoprecipitations from human cells verified this interaction and revealed that an internal peptide of AAMP spanning three WD40 domains was sufficient for this interaction. AAMP was found to be ubiquitously expressed in different human cell-lines and exhibited a predominant cytosolic localization in epithelial cells. Functionally, using overexpression and siRNA knock-down, we showed that AAMP modulates Nod2- and Nod1-mediated NF-kappaB activation in HEK293T cells. Taken together, our data support a new function of AAMP in regulating innate immune responses initiated by the NLR protein Nod2.


Assuntos
Proteínas de Transporte/metabolismo , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Proteínas de Transporte/genética , Linhagem Celular , Células HeLa , Humanos , Immunoblotting , Luciferases/genética , Luciferases/metabolismo , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteína Adaptadora de Sinalização NOD1/genética , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/genética , Plasmídeos/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/genética , Mapeamento de Interação de Proteínas , Receptores de Antígenos de Linfócitos B/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Transdução de Sinais
6.
J Pathol ; 214(2): 136-48, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18161746

RESUMO

The field of innate immunity has undergone an enormous upheaval during the last decade. The discovery of different groups of proteins, called pattern recognition molecules (PRMs), which detect microbial components, so-called pathogen-associated molecular patterns (PAMPs) and trigger protective responses, had a huge impact on the understanding of innate immune responses. Among the PRMs, the intracellular Nod-like receptors (NLRs) have recently been identified as key mediators of inflammatory and immune responses. The NLR family is divided into subfamilies on the basis of their different signal transduction domains, and recent studies have highlighted the role of certain NLRs, including Nod1, Nod2, Nalp3, Ipaf and Naip5, in the detection of intracellular microbes and possibly 'danger signals'. In this review, we summarize the current knowledge on the function of these proteins in immunity and inflammation, with a focus on their participation in different disease pathologies.


Assuntos
Infecções Bacterianas/imunologia , Inflamação/imunologia , Proteínas Adaptadoras de Sinalização NOD/imunologia , Humanos , Imunidade Inata , Mediadores da Inflamação/imunologia , Interleucina-1beta/biossíntese , NF-kappa B/imunologia , Proteínas Adaptadoras de Sinalização NOD/genética , Polimorfismo Genético
7.
J Cell Physiol ; 209(2): 241-52, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16897777

RESUMO

With the discovery of CARD15 as susceptibility gene for Crohn's disease (CD) a first link to a potential defect in the innate immune system was made. In this work we aimed to analyze enterocyte NOD2/CARD15 expression and regulation in response to bacterial motifs and the consequences of the most common CD-specific CARD15 mutation on antibacterial responses of normal intestinal epithelial cells (IEC). Under normal conditions, IEC lines and ileal enterocytes did not express NOD2/CARD15 mRNA or protein, contrary to IEC derived from inflammatory CD sections. In vitro analyses revealed that the simple contact with non-pathogenic commensal E. Coli K12 was sufficient to induced NOD2/CARD15 mRNA and protein in human IEC (HIEC). We identified bacterial flagellin interacting with TLR5 as major motif in this regulation of NOD2/CARD15. E. Coli mutants not expressing flagellin (DeltaFliC) failed to induce CARD15. Similarly, in HIEC transfected with a plasmid encoding dominant negative TLR5, no CARD15 induction was observed after K12 contact. Isolated TLR2 or TLR4 stimulation had no or only a marginal effect on NOD2/CARD15 expression. NOD2/CARD15 negative HIEC were unresponsive to muramyl dipeptide (MDP), but once NOD2/CARD15 was induced, HIEC and Caco2 cells responded to intra or extracellular MDP presentation with the activation of the NFkB pathway. IEC transfected with the Crohn-specific CARD15 mutant (F3020insC, FS) failed to activate NFkB after MDP-challenge, in contrast to CARD15WT IEC. In response to MDP, IEC induced a massive antibacterial peptide (ABP) response, seen in the apical release of CCL20. This was completely abolished in IEC carrying CARD15FS. These data suggest a critical role of NOD2/CARD15 in the bacterial clearance of the intestinal epithelium while CD-specific mutated NOD2/CARD15 causes an impaired epithelial barrier.


Assuntos
Enterócitos/metabolismo , Enterócitos/microbiologia , Escherichia coli/metabolismo , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptor 5 Toll-Like/metabolismo , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Motivos de Aminoácidos , Peptídeos Catiônicos Antimicrobianos/metabolismo , Células CACO-2 , Células Cultivadas , Enterócitos/citologia , Células HT29 , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação/genética , Proteína Adaptadora de Sinalização NOD2 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
8.
Infect Immun ; 74(6): 3115-24, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16714539

RESUMO

Intracellular peptidoglycan (PG) recognition in human cells is mediated by the NACHT-LRR proteins Nod1 and Nod2. Elicitation of these proteins by PG motifs released from invasive bacteria triggers signaling events, resulting in the activation of the NF-kappaB pathway. In order to decipher the molecular components involved in Nod2 signal transduction, we set out to identify new interaction partners of Nod2 by using a yeast two-hybrid screen. Besides the known interaction partner RIP2, the screen identified the leucine-rich repeat (LRR)- and PDZ domain-containing family member Erbin as a binding partner of Nod2. Erbin showed a specific interaction with Nod2 in coimmunoprecipitation experiments with human HEK 293T cells. Immunofluorescence microscopy with a newly generated anti-Nod2 monoclonal antibody showed that Erbin and Nod2 partially colocalize in human cells. Subsequent analysis of the Erbin/Nod2 interaction revealed that the LRR of Erbin and the caspase activating and recruiting domains of Nod2 were necessary for this interaction. No significant interaction was observed with a Walker B box mutant of Nod2 or a Crohn's disease-associated frameshift mutant of Nod2, indicating that complex formation is dependent on the activity of the molecule. In addition, a change in the dynamics of the Erbin/Nod2 complex was observed during Shigella flexneri infection. Furthermore, ectopic expression of increasing amounts of Erbin or short hairpin RNA-mediated knockdown of Erbin showed a negative influence of Erbin on Nod2/muramyl-dipeptide-mediated NF-kappaB activation. These results implicate Erbin as a potential negative regulator of Nod2 and show that bacterial infection has an impact on Nod2/Erbin complex formation within cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Shigella flexneri/patogenicidade , Proteínas Adaptadoras de Transdução de Sinal/química , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD2 , Estrutura Terciária de Proteína
9.
Cell Death Differ ; 13(5): 798-815, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16528382

RESUMO

Innate immunity to microorganisms in mammals has gained a substantial interest during the last decade. The discovery of the Toll-like receptor (TLR) family has allowed the identification of a class of membrane-spanning receptors dedicated to microbial sensing. TLRs transduce downstream signaling via their intracellular Toll-interleukin-1 receptor (TIR) domain. More recently, the role of intracellular microbial sensors has been uncovered. These molecules include the Nod-like receptors Nod1, Nod2, Ipaf and Nalps, together with the helicase domain-containing antiviral proteins RIG-I and Mda-5. The intracellular microbial sensors lack the TIR domain, but instead transduce downstream signals via two domains also implicated in homophilic protein-protein interactions, the caspase activation and recruitment domain (CARD) and PYRIN domains. In light with these recent findings, we propose that TIR, CARD and PYRIN domains represent the three arms of innate immune detection of microorganisms in mammals.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto/metabolismo , Imunidade Inata , Transdução de Sinais , Receptores Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Biomarcadores/análise , Doenças Genéticas Inatas/imunologia , Doenças Genéticas Inatas/metabolismo , Humanos , Modelos Biológicos , Oxigenases/metabolismo , Pirina , Receptores Toll-Like/genética
10.
Med Mycol ; 44(Supplement_1): S213-S217, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-30408906

RESUMO

Aspergillus fumigatus is a human pathogen, able to cause invasive aspergillosis in immunosuppressed patients. In the immunocompetent situation inhaled conidia are easily cleared by the immune system. Knowledge of the cellular pathways involved in the innate immunity against A. fumigatus is poorly represented. Therefore, we aimed to investigate the immune response against A. fumigatus in murine alveolar macrophages in terms of MAP kinases, NF-kappaB and cytokine signalling. Our investigations revealed that in murine alveolar macrophages, MAP kinases, ERK and p38 are activated under in vitro conditions, following addition of A. fumigatus conidia. In vivo experiments, however, showed that only ERK is directly involved, because activation of p38 was negligible. Immunosuppression with corticosteroids inhibited phosphorylation of ERK and was directly accompanied with a strongly decreased level of TNF-alpha and additional cytokines. In addition, killing of A. fumigatus conidia is reduced using the ERK inhibitor. Therefore, ERK appears to be an essential MAP kinase in the defence against A. fumigatus. Activation of the transcription factor NFkappaB appeared only at late times after infection suggesting an association with the intracellular swelling of conidia.

11.
Curr Opin Immunol ; 13(4): 410-6, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11498296

RESUMO

The ability to discriminate between pathogenic and non-pathogenic bacteria is extremely important for epithelial cells lining mucosal surfaces and is particularly so in colonic epithelial cells. Accumulating evidence suggests that bacterial recognition systems used by epithelial cells are very different from those in cells of the myeloid lineage and are likely to have developed to maintain mucosal surfaces in a state of homeostasis with the normal microbial flora. Bacterial invasion of epithelial cells or breach of the epithelial barrier provides a signal to epithelial cells to initiate inflammatory responses, which are key events for the clearance of the infecting microbe. Therefore, elucidation of the mechanisms by which epithelial cells recognize bacteria and bacterial products, and of the nature of the innate immune responses that are triggered by these factors are important for our understanding of both the immunology of mucosal surfaces and bacterial pathogenesis.


Assuntos
Infecções Bacterianas/imunologia , Animais , Apoptose , Bactérias/imunologia , Bactérias/patogenicidade , Infecções Bacterianas/genética , Infecções Bacterianas/microbiologia , Infecções Bacterianas/patologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Expressão Gênica , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Modelos Biológicos , Transdução de Sinais
12.
EMBO Rep ; 2(8): 736-42, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11463746

RESUMO

Epithelial cells are refractory to extracellular lipopolysaccharide (LPS), yet when presented inside the cell, it is capable of initiating an inflammatory response. Using invasive Shigella flexneri to deliver LPS into the cytosol, we examined how this factor, once intracellular, activates both NF-kappaB and c-Jun N-terminal kinase (JNK). Surprisingly, the mode of activation is distinct from that induced by toll-like receptors (TLRs), which mediate LPS responsiveness from the outside-in. Instead, our findings demonstrate that this response is mediated by a cytosolic, plant disease resistance-like protein called CARD4/Nod1. Biochemical studies reveal enhanced oligomerization of CARD4 upon S. flexneri infection, an event necessary for NF-kappaB induction. Dominant-negative versions of CARD4 block activation of NF-kappaB and JNK by S. flexneri as well as microinjected LPS. Finally, we showed that invasive S. flexneri triggers the formation of a transient complex involving CARD4, RICK and the IKK complex. This study demonstrates that in addition to the extracellular LPS sensing system mediated by TLRs, mammalian cells also possess a cytoplasmic means of LPS detection via a molecule that is related to plant disease-resistance proteins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/metabolismo , Proteínas de Drosophila , Regulação da Expressão Gênica/fisiologia , Lipopolissacarídeos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Shigella flexneri/fisiologia , Transdução de Sinais/fisiologia , Proteínas de Transporte/genética , Linhagem Celular , Genes Reporter , Células HeLa , Humanos , Quinase I-kappa B , Interleucina-1/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno , Lipopolissacarídeos/administração & dosagem , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Microinjeções , Proteína Adaptadora de Sinalização NOD1 , Testes de Precipitina , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/genética , Proteínas/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Shigella flexneri/patogenicidade , Fator 2 Associado a Receptor de TNF , Receptores Toll-Like , Fator de Necrose Tumoral alfa/farmacologia
13.
J Immunol ; 165(2): 903-14, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10878365

RESUMO

The pathogenesis of Shigella flexneri infection centers on the ability of this organism to invade epithelial cells and initiate an intense inflammatory reaction. Because NF-kappa B is an important transcriptional regulator of genes involved in inflammation, we investigated the role of this transcription factor during S. flexneri infection of epithelial cells. Infection of HeLa cells with invasive S. flexneri induced NF-kappa B DNA-binding activity; noninvasive S. flexneri strains did not lead to this activation. The pathway leading to NF-kappa B activation by invasive S. flexneri involved the kinases, NF-kappa B-inducing kinase, I kappa B kinase-1, and I kappa B kinase-2. NF-kappa B activation was linked to inflammation, because invasive S. flexneri activated an IL-8 promoter-driven reporter gene, and the kappa B site within this promoter was indispensable for its induction. Microinjection of bacterial culture supernatants into HeLa cells suggested that LPS is responsible for NF-kappa B activation by S. flexneri infection. In conclusion, the eukaryotic transcription factor NF-kappa B was activated during S. flexneri infection of epithelial cells, which suggests a role for this transcriptional regulator in modulating the immune response during infection in vivo.


Assuntos
Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Interleucina-8/biossíntese , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Shigella flexneri/imunologia , Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Células CACO-2 , Proteínas de Ligação a DNA/metabolismo , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática/imunologia , Células Epiteliais/enzimologia , Células Epiteliais/microbiologia , Células HeLa , Humanos , Quinase I-kappa B , Imunidade Inata , Interleucina-8/genética , Interleucina-8/metabolismo , Líquido Intracelular/microbiologia , Luciferases/biossíntese , Luciferases/genética , Luciferases/metabolismo , NF-kappa B/biossíntese , Regiões Promotoras Genéticas/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Shigella flexneri/patogenicidade , Transdução de Sinais/imunologia , Virulência/imunologia
14.
Philos Trans R Soc Lond B Biol Sci ; 355(1397): 575-86, 2000 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-10874731

RESUMO

Shigella flexneri is a Gram-negative facultatively intracellular pathogen responsible for bacillary dysentery in humans. More than one million deaths occur yearly due to infections with Shigella spp. and the victims are mostly children of the developing world. The pathogenesis of Shigella centres on the ability of this organism to invade the colonic epithelium where it induces severe mucosal inflammation. Much information that we have gained concerning the pathogenesis of Shigella has been derived from the study of in vitro models of infection. Using these techniques, a number of the molecular mechanisms by which Shigella invades epithelial cells and macrophages have been identified. In vivo models of shigellosis have been hampered since humans are the only natural hosts of Shigella. However, experimental infection of macaques as well as the murine lung and rabbit ligated ileal loop models have been important in defining some of the immune and inflammatory components of the disease. In particular, the murine lung model has shed light on the development of systemic and local immune protection against Shigella infection. It would be naive to believe that any one model of Shigella infection could adequately represent the complexity of the disease in humans, and more sophisticated in vivo models are now necessary. These models require the use of human cells and tissue, but at present such models remain in the developmental stage. Ultimately, however, it is with such studies that novel treatments and vaccine candidates for the treatment and prevention of shigellosis will be designed.


Assuntos
Disenteria Bacilar/microbiologia , Shigella flexneri/patogenicidade , Adaptação Fisiológica/imunologia , Animais , Apoptose , Adesão Celular , Disenteria Bacilar/imunologia , Células Epiteliais/microbiologia , Humanos , Líquido Intracelular/microbiologia , Macrófagos/microbiologia , Coelhos , Receptores de Superfície Celular/metabolismo , Shigella flexneri/imunologia
15.
J Pharmacol Exp Ther ; 287(1): 128-36, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9765331

RESUMO

Many studies have indicated an association between bacteria and the severity of enteric secretory or inflammatory disorders. We previously showed that monolayers of human T84 epithelial cells display altered ion transport and permeability after coculture with Staphylococcus aureus enterotoxin B (SEB, a model superantigen)-activated immune cells, where interferon-gamma and tumor necrosis factor-alpha were key mediators in the pathophysiology. Here we examined whether the regulatory Th2-type cytokines, interleukin (IL)-10 and IL-4, could prevent these epithelial irregularities. T84 monolayers were cocultured with human peripheral blood mononuclear cells (PBMC) or T cell-enriched, monocyte-depleted PBMC (T + B cells) +/- SEB for 20 hr in the presence or absence of IL-10 or IL-4. Subsequently, T84 monolayers were mounted in Ussing chambers and ion transport (short-circuit current (Isc) and DeltaIsc evoked by forskolin) and permeability (ion resistance and probe fluxes) were assessed. IL-10 dose-dependently inhibited the increased T84 permeability and the reduced responsiveness to forskolin that were evoked by coculture with SEB-activated PBMC or T + B cells. Similar changes in T84 function occurred in response to conditioned medium from SEB-activated immune cells; however, addition of IL-10 to the conditioned medium did not prevent the changes in epithelial function. In contrast, when PBMC were stimulated with SEB in the presence of IL-10, the subsequent conditioned medium was less effective in evoking altered epithelial function. These data suggest that the affect of IL-10 was due to effects on the immune cells and not directly on the epithelium. In contrast to IL-10, IL-4 did not ameliorate any of the immune-mediated changes in T84 function. We conclude that IL-10 can reduce the epithelial functional changes caused by SEB-activated immune cells and this data adds further support for IL-10 immunotherapy in the treatment of intestinal secretory or inflammatory disorders.


Assuntos
Enterotoxinas/farmacologia , Interleucina-10/farmacologia , Interleucina-4/farmacologia , Mucosa Intestinal/metabolismo , Transporte de Íons/efeitos dos fármacos , Linfócitos/fisiologia , Superantígenos/farmacologia , Linhagem Celular , Citocinas/biossíntese , Células Epiteliais/metabolismo , Humanos , Mucosa Intestinal/imunologia , Muromonab-CD3/imunologia , Permeabilidade
16.
Infect Immun ; 66(4): 1680-7, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9529098

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) infection is associated with watery diarrhea and can lead to complications, including hemorrhagic colitis and the hemolytic-uremic syndrome. The mechanisms by which these organisms produce diarrheal disease remain to be elucidated. Changes in T84 epithelial cell electrophysiology were examined following EHEC infection. T84 cell monolayers infected with EHEC O157:H7 displayed a time-dependent decrease in transepithelial resistance. Increases in the transepithelial flux of both [3H]mannitol and 51Cr-EDTA accompanied the EHEC-induced decreases in T84 resistance. Altered barrier function induced by EHEC occurred at the level of the tight junction since immunofluorescent staining of the tight-junction-associated protein ZO-1 was disrupted when examined by confocal microscopy. Decreased resistance induced by EHEC involved a protein kinase C (PKC)-dependent pathway as the highly specific PKC inhibitor, CGP41251, abrogated the EHEC-induced drop in resistance. PKC activity was also increased in T84 cells infected with EHEC. Calmodulin and myosin light chain kinase played a role in EHEC-induced resistance changes as inhibition of these effector molecules partially reversed the effects of EHEC on barrier function. These studies demonstrate that intracellular signal transduction pathways activated following EHEC infection link the increases in T84 epithelial permeability induced by this pathogen.


Assuntos
Diarreia/etiologia , Escherichia coli O157/patogenicidade , Mucosa Intestinal/metabolismo , Transdução de Sinais , Calmodulina/fisiologia , Linhagem Celular , Sobrevivência Celular , Humanos , Quinase de Cadeia Leve de Miosina/fisiologia , Permeabilidade , Proteína Quinase C/fisiologia
17.
J Infect Dis ; 177(4): 955-61, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9534968

RESUMO

Helicobacter pylori, the etiologic agent of chronic-active gastritis and duodenal ulcers in humans, and Helicobacter mustelae, a gastric pathogen in ferrets, bind to phosphatidylethanolamine (PE), a constituent of host gastric mucosal cells, and to gangliotetraosylceramide (Gg4) and gangliotriaosylceramide (Gg3). The effect of a bovine colostrum concentrate (BCC) on the interaction of H. pylori and H. mustelae to their lipid receptors was examined. BCC blocked attachment of both species to Gg4, Gg3, and PE. Partial inhibition of binding was observed with native bovine and human colostra. BCC lacked detectable antibodies (by immunoblotting) to H. pylori surface proteins (adhesins). However, colostral lipid extracts contained PE and lyso-PE that bound H. pylori in vitro. These results indicate that colostrum can block the binding of Helicobacter species to select lipids and that binding inhibition is conferred, in part, by colostral PE or PE derivatives. Colostral lipids may modulate the interaction of H. pylori and other adhesin-expressing pathogens with their target tissues.


Assuntos
Colostro/imunologia , Gangliosídeo G(M2)/análogos & derivados , Glicoesfingolipídeos/metabolismo , Helicobacter pylori/metabolismo , Helicobacter/metabolismo , Fosfatidiletanolaminas/metabolismo , Adesinas Bacterianas/imunologia , Adesinas Bacterianas/metabolismo , Animais , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/imunologia , Aderência Bacteriana/imunologia , Bovinos , Cromatografia em Camada Fina , Colostro/química , Feminino , Gangliosídeo G(M2)/imunologia , Gangliosídeo G(M2)/metabolismo , Gangliosídeos , Glicoesfingolipídeos/imunologia , Helicobacter/imunologia , Helicobacter pylori/imunologia , Humanos , Immunoblotting , Metabolismo dos Lipídeos , Lipídeos/análise , Fosfatidiletanolaminas/imunologia , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo
19.
Aliment Pharmacol Ther ; 11 Suppl 3: 70-80, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9467981

RESUMO

Research efforts in inflammatory bowel disease (IBD) have been directed towards the epithelium as it has become clear that epithelial cells play a critical role in inflammatory response. Most research involving IBD employs in vitro techniques. In vitro epithelial cell studies have played and are continuing to play a major role in providing specific information relevant to IBD. Thus, such studies have provided irrefutable evidence that epithelial responses can be induced by microbes/microbial products and by immune activation. Culture experiments have provided insights into the effects of individual cytokines and other inflammatory mediators on epithelial pathophysiology, injury and repair, apoptosis, necrosis, and other processes that may be involved in IBD. Activated epithelial cells can participate in and even orchestrate immune responses, by stimulating T cells (and possibly others) and by producing cytokines that recruit specific inflammatory cells. Physiological regulation of epithelial tight junctions has been demonstrated by in vitro studies; the implication of this information for treating IBD is just beginning to be explored. It is becoming increasingly clear that epithelial processing and presentation of antigens is critical to the outcome of the immune response.


Assuntos
Citocinas/biossíntese , Células Epiteliais/imunologia , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/imunologia , Linfócitos T/imunologia , Animais , Bactérias/patogenicidade , Toxinas Bacterianas/imunologia , Linhagem Celular , Quimiocinas/biossíntese , Células Epiteliais/microbiologia , Substâncias de Crescimento/biossíntese , Humanos , Mucosa Intestinal/microbiologia
20.
Am J Physiol ; 273(6): G1349-58, 1997 12.
Artigo em Inglês | MEDLINE | ID: mdl-9435561

RESUMO

Verotoxin-producing Escherichia coli (VTEC) are pathogenic bacteria associated with diarrhea, hemorrhagic colitis, and hemolytic uremic syndrome. Verotoxins (VTs) elaborated by these organisms produce cytopathic effects on a restricted number of cell types, including endothelial cells lining the microvasculature of the bowel and the kidney. Because human intestinal epithelial cells lack the globotriaosylceramide receptor for VT binding, it is unclear how the toxin moves across the intestinal mucosa to the systemic circulation. The aims of this study were to determine the effects of VT-1 on intestinal epithelial cell function and to characterize VT-1 translocation across monolayers of T84 cells, an intestinal epithelial cell line. VT-1 at concentrations up to 1 microgram/ml had no effect on the barrier function of T84 monolayers as assessed by measuring transmonolayer electrical resistance (102 +/- 8% of control monolayers). In contrast, both VT-positive and VT-negative VTEC bacterial strains lowered T84 transmonolayer resistance (45 +/- 7 and 38 +/- 6% of controls, respectively). Comparable amounts of toxin moved across monolayers of T84 cells, exhibiting high-resistance values, as monolayers with VTEC-induced decreases in barrier function, suggesting a transcellular mode of transport. Translocation of VT-1 across T84 monolayers paralleled the movement of a comparably sized protein, horseradish peroxidase. Immunoelectron microscopy confirmed transcellular transport of VT-1, since the toxin was observed within endosomes and associated with specific intracellular targets, including the Golgi network and endoplasmic reticulum. These data present a mode of VT-1 uptake by toxin-insensitive cells and suggest a general mechanism by which bacterial toxins lacking specific intestinal receptors can penetrate the intestinal epithelial barrier.


Assuntos
Toxinas Bacterianas/farmacocinética , Enterotoxinas/farmacocinética , Mucosa Intestinal/fisiologia , Animais , Toxinas Bacterianas/toxicidade , Transporte Biológico , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/ultraestrutura , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Escherichia coli/classificação , Peroxidase do Rábano Silvestre/farmacocinética , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/ultraestrutura , Potenciais da Membrana/efeitos dos fármacos , Microscopia Imunoeletrônica , Sorotipagem , Toxina Shiga I , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...