Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Pediatr Surg Int ; 40(1): 35, 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38216767

RESUMO

PURPOSE: Necrotizing enterocolitis (NEC) is a severe intestinal disease primarily affecting premature infants, marked by impaired epithelial regeneration. Breastfed infants are less susceptible to NEC than formula-fed ones, and human milk oligosaccharides (HMO) found in breast milk have prebiotic properties that can protect against NEC. However, it is unclear how HMOs influence intestinal epithelium regeneration in relation to the gut microbiota. METHODS: Broad-spectrum antibiotics were administered to pregnant dams to reduce the microbiota in offspring. NEC was induced through administration of hyperosmolar formula, lipopolysaccharide, and hypoxia from postnatal days (p) 5-9. Intestinal epithelial organoids were derived from p9 mice. HMOs were isolated from human donor breast milk and then solubilized in the formula for each feed or culture media for organoids. RESULTS: HMOs did not alter the microbiota profile in the presence of a normal or reduced microbiota. In the reduced microbiota, HMO treatment decreased NEC intestinal injury, and increased proliferation and stem cell activity. Additionally, in the complete absence of the microbiota, HMOs stimulated intestinal organoid growth. CONCLUSION: This study demonstrates that HMOs promoted intestinal epithelial regeneration independent of the gut microbiota. These findings provide further insight into the various benefits HMOs may have in the protection against NEC.


Assuntos
Enterocolite Necrosante , Doenças do Recém-Nascido , Microbiota , Lactente , Feminino , Gravidez , Recém-Nascido , Animais , Humanos , Camundongos , Leite Humano , Enterocolite Necrosante/prevenção & controle , Mucosa Intestinal , Oligossacarídeos/farmacologia , Regeneração
2.
Pediatr Res ; 91(1): 101-106, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34561550

RESUMO

BACKGROUND: Stem cell therapy has been proven to rescue intestinal injury and stimulate intestinal regeneration in necrotizing enterocolitis (NEC). Specifically, stem cells derived from amniotic fluid (AFSCs) and mesenchymal stem cells (MSCs) derived from bone marrow have shown promising results in the treatment of experimental NEC. This study aims to examine the effects of AFSCs and MSCs on the prevention of intestinal injury during experimental NEC. METHODS: Supernatants from AFSC and MSC cultures were collected to perform proteomic analysis. Prior to NEC induction, mice received intraperitoneal injections of phosphate-buffered saline (PBS), 2 × 106 AFSCs, or 2 × 106 MSCs. RESULTS: We found that AFSCs grew faster than MSCs. Proteomic analysis indicated that AFSCs are primarily involved in cell development and growth, while MSCs are involved in immune regulation. Administering AFSCs before NEC induction decreased NEC severity and mucosal inflammation. Intestinal proliferation and endogenous stem cell activation were increased after AFSC administration. However, administering MSCs before NEC induction had no beneficial effects. CONCLUSIONS: This study demonstrated that AFSCs and MSCs have different protein release profiles. AFSCs can potentially be used as a preventative strategy for neonates at risk of NEC, while MSCs cannot be used. IMPACT: AFSCs and MSCs have distinct protein secretory profiles, and AFSCs are primarily involved in cell development and growth, while MSCs are involved in immune regulation. AFSCs are unique in transiently enhancing healthy intestinal epithelial cell growth, which offers protection against the development of experimental NEC. The prevention of NEC via the administration of AFSCs should be evaluated in infants at great risk of developing NEC or in infants with early signs of NEC.


Assuntos
Líquido Amniótico/citologia , Transplante de Células-Tronco , Animais , Enterocolite Necrosante , Humanos , Recém-Nascido , Camundongos
3.
Cell Death Dis ; 10(10): 743, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31582728

RESUMO

Necrotizing enterocolitis (NEC) is a devastating neonatal disease characterized by acute intestinal injury. Intestinal stem cell (ISC) renewal is required for gut regeneration in response to acute injury. The Wnt/ß-catenin pathway is essential for intestinal renewal and ISC maintenance. We found that ISC expression, Wnt activity and intestinal regeneration were all decreased in both mice with experimental NEC and in infants with acute active NEC. Moreover, intestinal organoids derived from NEC-injured intestine of both mice and humans failed to maintain proliferation and presented more differentiation. Administration of Wnt7b reversed these changes and promoted growth of intestinal organoids. Additionally, administration of exogenous Wnt7b rescued intestinal injury, restored ISC, and reestablished intestinal epithelial homeostasis in mice with NEC. Our findings demonstrate that during NEC, Wnt/ß-catenin signaling is decreased, ISC activity is impaired, and intestinal regeneration is defective. Administration of Wnt resulted in the maintenance of intestinal epithelial homeostasis and avoidance of NEC intestinal injury.


Assuntos
Enterocolite Necrosante/fisiopatologia , Intestinos/fisiopatologia , Regeneração/fisiologia , Via de Sinalização Wnt , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Enterocolite Necrosante/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Intestinos/efeitos dos fármacos , Intestinos/patologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Organoides/efeitos dos fármacos , Organoides/metabolismo , Proteínas Proto-Oncogênicas/administração & dosagem , Proteínas Proto-Oncogênicas/farmacologia , Regeneração/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Análise de Sobrevida , Proteínas Wnt/administração & dosagem , Proteínas Wnt/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética
4.
Diabetes ; 64(7): 2537-49, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25735732

RESUMO

Obesity and diabetes are characterized by increased inflammation reflecting disordered control of innate immunity. We reveal a local intestinal intraepithelial lymphocyte (IEL)-GLP-1 receptor (GLP-1R) signaling network that controls mucosal immune responses. Glp1r expression was enriched in intestinal IEL preparations and copurified with markers of Tαß and Tγδ IELs, the two main subsets of intestinal IELs. Exendin-4 increased cAMP accumulation in purified IELs and reduced the production of cytokines from activated IELs but not from splenocytes ex vivo. These actions were mimicked by forskolin, absent in IELs from Glp1r(-/-) mice, and attenuated by the GLP-1R agonist exendin (9-39) consistent with a GLP-1R-dependent mechanism of action. Furthermore, Glp1r(-/-) mice exhibited dysregulated intestinal gene expression, an abnormal representation of microbial species in feces, and enhanced sensitivity to intestinal injury following administration of dextran sodium sulfate. Bone marrow transplantation using wild-type C57BL/6 donors normalized expression of multiple genes regulating immune function and epithelial integrity in Glp1r(-/-) recipient mice, whereas acute exendin-4 administration robustly induced the expression of genes encoding cytokines and chemokines in normal and injured intestine. Taken together, these findings define a local enteroendocrine-IEL axis linking energy availability, host microbial responses, and mucosal integrity to the control of innate immunity.


Assuntos
Mucosa Intestinal/imunologia , Linfócitos/imunologia , Receptores de Glucagon/agonistas , Receptores de Glucagon/fisiologia , Animais , Citocinas/análise , Exenatida , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/farmacologia , Transdução de Sinais , Peçonhas/farmacologia
5.
Shock ; 43(5): 483-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25608140

RESUMO

Intestinal epithelia regulate barrier integrity when challenged by inflammation, oxidative stress, and microbes. Transforming growth factor-ß1 (TGF-ß1) is a cytokine with known beneficial effects on intestinal epithelia, including barrier enhancement, after exposure to proinflammatory cytokines and infectious agents. The aim of this study was to determine whether TGF-ß1 directly protects intestinal epithelia during hypoxia-reoxygenation (HR). Intestinal epithelial monolayers (T84, Caco-2) were exposed to either hypoxia (1% O2, 1 h) or oxidative stress (hydrogen peroxide, 1 mM), followed by normoxic atmosphere for different time points in the absence and presence of varying concentrations of TGF-ß1. Transepithelial electrical resistance (TER) assessed barrier function, with RNA extracted for reverse transcription polymerase chain reaction analysis of GPx-1, HIF-1, heme-oxygenase-1 (HO-1), and NOX-1. In some experiments, intestinal epithelia were exposed to enterohemorrhagic Escherichia coli (EHEC) O157:H7 during the reoxygenation period and TER recorded 7 h after the infectious challenge. Hypoxia-reoxygenation significantly decreased TER in intestinal epithelia compared with normoxic controls. Transforming growth factor-ß1 pretreatment ameliorated HR-induced epithelial barrier dysfunction in T84 (at 1 - 3 h) and Caco-2 (1 h) monolayers. Transforming growth factor-ß1 preserved barrier integrity for up to 16 h after challenge with hydrogen peroxide. In TGF-ß1-treated epithelial monolayers, only HO-1 mRNA significantly increased after HR (P < 0.05 vs. normoxic controls). The EHEC-induced epithelial barrier dysfunction was significantly worsened by intestinal HR (P < 0.05 vs. normoxia-EHEC-infected cells), but this was not protected by TGF-ß1 pretreatment. Transforming growth factor-ß1 preserves loss of epithelial barrier integrity caused by the stress of HR via a mechanism that may involve the upregulation of HO-1 transcription. Targeted treatment with TGF-ß could lead to novel therapies in enteric diseases characterized by HR injury.


Assuntos
Mucosa Intestinal/efeitos dos fármacos , Estresse Oxidativo , Fator de Crescimento Transformador beta1/farmacologia , Células CACO-2 , Hipóxia Celular , Citocinas/metabolismo , Impedância Elétrica , Escherichia coli Êntero-Hemorrágica/patogenicidade , Células Epiteliais/efeitos dos fármacos , Epitélio/metabolismo , Expressão Gênica , Heme Oxigenase-1/metabolismo , Humanos , Peróxido de Hidrogênio/química , Enteropatias/metabolismo , Mucosa Intestinal/patologia , Oxigênio/química , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia , Fatores de Tempo
6.
Inflamm Bowel Dis ; 21(2): 297-306, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25590952

RESUMO

BACKGROUND: Adherent-invasive Escherichia coli (AIEC) colonization has been strongly implicated in the pathogenesis of Crohn's disease. Environmental triggers such as vitamin D deficiency have emerged as key factors in the pathogenesis of inflammatory bowel diseases. The aim of this study was to investigate the effects of 1,25(OH)2D3 on AIEC infection-induced changes in vivo and in vitro. METHODS: Barrier function was assessed in polarized epithelial Caco-2-bbe cells grown in medium with or without vitamin D and challenged with AIEC strain LF82. Weaned C57BL/6 mice were fed either a vitamin D-sufficient or -deficient diet for 5 weeks and then infected with AIEC, in the absence and presence of low-dose dextran sodium sulphate. Disease severity was assessed by histological analysis and in vivo intestinal permeability assay. Presence of invasive bacteria was assessed by transmission electron microscopy. RESULTS: Caco-2-bbe cells incubated with 1,25(OH)2D3 were protected against AIEC-induced disruption of transepithelial electrical resistance and tight-junction protein redistribution. Vitamin D-deficient C57BL/6 mice given a course of 2% dextran sodium sulphate exhibited pronounced epithelial barrier dysfunction, were more susceptible to AIEC colonization, and showed exacerbated colonic injury. Transmission electron microscopy of colonic tissue from infected mice demonstrated invasion of AIEC and fecal microbiome analysis revealed shifts in microbial communities. CONCLUSIONS: These data show that vitamin D is able to mitigate the deleterious effects of AIEC on the intestinal mucosa, by maintaining intestinal epithelial barrier homeostasis and preserving tight-junction architecture. This study highlights the association between vitamin D status, dysbiosis, and Crohn's disease.


Assuntos
Calcitriol/farmacologia , Colite/etiologia , Doença de Crohn/etiologia , Modelos Animais de Doenças , Infecções por Escherichia coli/etiologia , Escherichia coli/patogenicidade , Mucosa Intestinal/patologia , Deficiência de Vitamina D/fisiopatologia , Animais , Apoptose , Aderência Bacteriana/efeitos dos fármacos , Western Blotting , Células CACO-2 , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Colite/metabolismo , Colite/patologia , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Sulfato de Dextrana/farmacologia , Condutividade Elétrica , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Feminino , Imunofluorescência , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Humanos , Técnicas Imunoenzimáticas , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deficiência de Vitamina D/complicações
7.
J Nutr ; 144(11): 1725-33, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25143376

RESUMO

BACKGROUND: Few studies have focused on the ability of prebiotics to prevent pathogen-induced cellular changes or alter the composition of the intestinal microbiota in complimentary relevant cell and animal models of inflammatory bowel disease. OBJECTIVE: The objective of this study was to determine if pretreatment with inulin and a short-chain fructo-oligosaccharide (sc-FOS) prevents enterohemorrhagic Escherichia coli (EHEC) O157:H7 infection in Caco2-bbe epithelial cells and what effect 10% wt:v sc-FOS or inulin has on C57BL/6 mice under sham conditions or pretreatment with prebiotics before Citrobacter rodentium infection (10(8) colony-forming units). METHODS: Actin rearrangement and tight junction protein (zona occludin-1) were examined with immunofluorescence. Barrier function was assessed by a fluorescent probe and by measuring transepithelial electrical resistance (TER). Alterations in cytokine gene expression and microbiome were assessed with quantitative reverse transcriptase-polymerase chain reaction and fluorescence in situ hybridization. Short-chain fatty acids (SCFAs) were measured by GC. RESULTS: sc-FOS added to monolayers altered actin polymerization without affecting TER or permeability to a fluorescein isothiocyanate (FITC) probe, whereas inulin increased TER (P < 0.005) and altered actin arrangement without affecting FITC permeability. Neither prebiotic attenuated EHEC-induced decreases in barrier function. Prebiotics increased interleukin 10 (Il10) and transforming growth factor-ß (Tgfß) cytokine responses alone (P < 0.05) or with EHEC O157:H7 infection (P < 0.05) in vitro. Increases in tumor necrosis factor-α (Tnfα) (P < 0.05) and decreases in chemokine CXC motif ligand 8 (Cxcl8) (P < 0.05) expression were observed with prebiotic treatment prior to EHEC infection. No differences were noted in barrier function or cytokine responses in the absence or presence of C. rodentium in vivo. Alterations in microbiome were evident at 6 d and 10 d postinfection in treatment groups, but a change in C. rodentium load was not observed. Inulin and sc-FOS (P < 0.05) increased fecal SCFAs in the absence of infection. CONCLUSION: This study provides new insights as to how prebiotics act in complementary in vitro and in vivo models of intestinal injury.


Assuntos
Infecções por Enterobacteriaceae/complicações , Escherichia coli O157 , Inflamação/tratamento farmacológico , Inulina/farmacologia , Oligossacarídeos/farmacologia , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Células CACO-2 , Citrobacter rodentium , Colite/tratamento farmacológico , Colite/microbiologia , Fezes/microbiologia , Feminino , Humanos , Inulina/química , Camundongos , Camundongos Endogâmicos C57BL , Oligossacarídeos/química , Prebióticos
8.
Infect Immun ; 82(4): 1648-56, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24491575

RESUMO

Enterohemorrhagic Escherichia coli serotype O157:H7 causes outbreaks of diarrhea, hemorrhagic colitis, and the hemolytic-uremic syndrome. E. coli O157:H7 intimately attaches to epithelial cells, effaces microvilli, and recruits F-actin into pedestals to form attaching and effacing lesions. Lipid rafts serve as signal transduction platforms that mediate microbe-host interactions. The aims of this study were to determine if protein kinase C (PKC) is recruited to lipid rafts in response to E. coli O157:H7 infection and what role it plays in attaching and effacing lesion formation. HEp-2 and intestine 407 tissue culture epithelial cells were challenged with E. coli O157:H7, and cell protein extracts were then separated by buoyant density ultracentrifugation to isolate lipid rafts. Immunoblotting for PKC was performed, and localization in lipid rafts was confirmed with an anti-caveolin-1 antibody. Isoform-specific PKC small interfering RNA (siRNA) was used to determine the role of PKC in E. coli O157:H7-induced attaching and effacing lesions. In contrast to uninfected cells, PKC was recruited to lipid rafts in response to E. coli O157:H7. Metabolically active bacteria and cells with intact lipid rafts were necessary for the recruitment of PKC. PKC recruitment was independent of the intimin gene, type III secretion system, and the production of Shiga toxins. Inhibition studies, using myristoylated PKCζ pseudosubstrate, revealed that atypical PKC isoforms were activated in response to the pathogen. Pretreating cells with isoform-specific PKC siRNA showed that PKCζ plays a role in E. coli O157:H7-induced attaching and effacing lesions. We concluded that lipid rafts mediate atypical PKC signal transduction responses to E. coli O157:H7. These findings contribute further to the understanding of the complex array of microbe-eukaryotic cell interactions that occur in response to infection.


Assuntos
Aderência Bacteriana/fisiologia , Infecções por Escherichia coli/microbiologia , Escherichia coli O157/fisiologia , Proteína Quinase C/fisiologia , Células Cultivadas , Células Epiteliais/microbiologia , Escherichia coli O157/patogenicidade , Humanos , Immunoblotting , Microdomínios da Membrana/fisiologia , Transdução de Sinais/fisiologia , Fatores de Virulência/fisiologia
9.
Infect Immun ; 80(7): 2307-15, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22526675

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) serotype O157:H7 is a food-borne pathogen that causes significant morbidity and mortality in developing and industrialized nations. EHEC infection of host epithelial cells is capable of inhibiting the gamma interferon (IFN-γ) proinflammatory pathway through the inhibition of Stat-1 phosphorylation, which is important for host defense against microbial pathogens. The aim of this study was to determine the bacterial factors involved in the inhibition of Stat-1 tyrosine phosphorylation. Human HEp-2 and Caco-2 epithelial cells were challenged directly with either EHEC or bacterial culture supernatants and stimulated with IFN-γ, and then the protein extracts were analyzed by immunoblotting. The data showed that IFN-γ-mediated Stat-1 tyrosine phosphorylation was inhibited by EHEC secreted proteins. Using two-dimensional difference gel electrophoresis, EHEC Shiga toxins were identified as candidate inhibitory factors. EHEC Shiga toxin mutants were then generated and complemented in trans, and mutant culture supernatant was supplemented with purified Stx to confirm their ability to subvert IFN-γ-mediated cell activation. We conclude that while other factors are likely involved in the suppression of IFN-γ-mediated Stat-1 tyrosine phosphorylation, E. coli-derived Shiga toxins represent a novel mechanism by which EHEC evades the host immune system.


Assuntos
Escherichia coli O157/imunologia , Escherichia coli O157/patogenicidade , Evasão da Resposta Imune , Interferon gama/imunologia , Fator de Transcrição STAT1/imunologia , Toxinas Shiga/imunologia , Toxinas Shiga/toxicidade , Western Blotting , Linhagem Celular , Eletroforese em Gel Bidimensional , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Humanos , Interferon gama/antagonistas & inibidores , Fosforilação , Proteoma/análise , Fator de Transcrição STAT1/metabolismo
10.
J Infect Dis ; 206(1): 99-109, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22430833

RESUMO

BACKGROUND: Probiotics prevent disease induced by Citrobacter rodentium, a murine-specific enteric pathogen. Whether probiotics can be used to interrupt the infectious process following initiation of infection was determined. METHODS: C57BL/6 adult and neonatal mice were challenged with C. rodentium, and a probiotic mixture containing Lactobacillus helveticus and Lactobacillus rhamnosus was provided 1 week before bacterial challenge, concurrently with infection, or 3 days and 6 days after infection. Mice were sacrificed 10 days after infection, and disease severity was assessed by histological analysis and in vivo intestinal permeability assay. Inflammatory pathways and the composition of the fecal microbiome were assessed in adult mice. RESULTS: Preadministration and coadministration of probiotics ameliorated C. rodentium-induced barrier dysfunction, epithelial hyperplasia, and binding of the pathogen to host colonocytes in adults, with similar findings in neonatal mice. Upregulated tumor necrosis factor α and interferon γ transcripts were suppressed in the pretreated probiotic group, whereas interleukin 17 transcription was suppressed with probiotics given up to 3 days after infection. Probiotics promoted transcription of interleukin 10 and FOXP3, and increased follicular T-regulatory cells in pretreatment mice. C. rodentium infection resulted in an altered fecal microbiome, which was normalized with probiotic intervention. CONCLUSIONS: This study provides evidence that probiotics can prevent illness and treat disease in an animal model of infectious colitis.


Assuntos
Citrobacter rodentium/crescimento & desenvolvimento , Colite/terapia , Infecções por Enterobacteriaceae/terapia , Lacticaseibacillus rhamnosus , Lactobacillus helveticus , Probióticos/farmacologia , Animais , Citrobacter rodentium/metabolismo , Colite/metabolismo , Colite/microbiologia , Colo/metabolismo , Colo/microbiologia , Infecções por Enterobacteriaceae/metabolismo , Infecções por Enterobacteriaceae/microbiologia , Fezes/microbiologia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Hiperplasia/metabolismo , Hiperplasia/microbiologia , Hiperplasia/prevenção & controle , Hiperplasia/terapia , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Metagenoma , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/microbiologia , Fator de Necrose Tumoral alfa/metabolismo
11.
Microbiology (Reading) ; 155(Pt 2): 531-540, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19202101

RESUMO

Enterohaemorrhagic Escherichia coli (EHEC) O157:H7 inhibits interferon (IFN)-gamma-stimulated tyrosine phosphorylation of signal transducer and activator of transcription (STAT)-1 in epithelial cells. We determined the effects of probiotics on EHEC-mediated disruption of IFN-gamma-stimulated STAT-1 activation in epithelial cell lines. Confluent Intestine 407, HEp-2 and Caco-2 epithelial cells were pre-treated (3 h) with either probiotics or surface-layer proteins derived from Lactobacillus helveticus R0052 prior to infection with EHEC O157:H7 strain CL56 (m.o.i. 100:1, 6 h, 37 degrees C in 5% CO2). Subsequently, cells were washed and stimulated with human recombinant IFN-gamma (50 ng ml(-1), 0.5 h, 37 degrees C) followed by whole-cell protein extraction and immunoblotting for tyrosine-phosphorylated STAT-1. Relative to uninfected cells, STAT-1-activation was reduced after EHEC O157:H7 infection. Pre-incubation with the probiotic L. helveticus R0052 followed by EHEC infection abrogated pathogen-mediated disruption of IFN-gamma-STAT-1 signalling. As determined using Transwell inserts, probiotic-mediated protection was independent of epithelial cell contact. In contrast, pre-incubation with boiled L. helveticus R0052, an equal concentration of viable Lactobacillus rhamnosus R0011, or surface-layer proteins (0.14 mg ml(-1)) did not restore STAT-1 signalling in EHEC-infected cells. The viable probiotic agent L. helveticus R0052 prevented EHEC O157:H7-mediated subversion of epithelial cell signal transduction responses.


Assuntos
Regulação para Baixo , Infecções por Escherichia coli/metabolismo , Escherichia coli O157/fisiologia , Interferon gama/metabolismo , Probióticos/química , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Proteínas de Bactérias/farmacologia , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/prevenção & controle , Humanos , Lactobacillus helveticus/química , Lactobacillus helveticus/fisiologia , Fosforilação/efeitos dos fármacos , Probióticos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tirosina/metabolismo
12.
Microb Pathog ; 45(5-6): 377-85, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18930803

RESUMO

Attaching-effacing lesion-inducing Escherichia albertii and the related, but non-attaching-effacing organism, Hafnia alvei, are both implicated as enteric pathogens in humans. However, effects of these bacteria on epithelial cells are not well-characterized. Related enteropathogens, including enterohemorrhagic Escherichia coli O157:H7, decrease epithelial barrier function by disrupting intercellular tight junctions in polarized epithelia. Therefore, this study assessed epithelial barrier function and tight junction protein distribution in polarized epithelia following bacterial infections. Polarized epithelial (MDCK-I and T84) cells grown on filter supports were infected apically with E. coli O157:H7, E. albertii, and H. alvei for 16h at 37 degrees C. All strains decreased transepithelial electrical resistance and increased permeability to a dextran probe in a host cell-dependent manner. Immunofluorescence microscopy showed that both E. coli O157:H7 and E. albertii, but not H. alvei, caused a redistribution of the tight junction protein zona occludens-1. In contrast to E. coli O157:H7, E. albertii and H. alvei did not redistribute claudin-1. Western blotting of whole cell protein extracts demonstrated that each bacterium caused differential changes in tight junction protein expression, dependent on the host cell. These findings demonstrate that E. albertii and H. alvei are candidate enteric pathogens that have both strain-specific and host epithelial cell-dependent effects.


Assuntos
Infecções por Enterobacteriaceae/microbiologia , Escherichia/patogenicidade , Hafnia alvei/patogenicidade , Intestinos/microbiologia , Junções Íntimas/microbiologia , Animais , Aderência Bacteriana , Linhagem Celular , Permeabilidade da Membrana Celular , Claudina-1 , Cães , Infecções por Enterobacteriaceae/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Escherichia/fisiologia , Escherichia coli O157/patogenicidade , Hafnia alvei/fisiologia , Humanos , Mucosa Intestinal/metabolismo , Proteínas de Membrana/metabolismo , Junções Íntimas/metabolismo
13.
Cell Microbiol ; 9(2): 356-67, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16925785

RESUMO

Adherence of intestinal pathogens, including Escherichia coli O157:H7, to human intestinal epithelial cells is a key step in pathogenesis. Probiotic bacteria, including Lactobacillus helveticus R0052 inhibit the adhesion of E. coli O157:H7 to epithelial cells, a process which may be related to specific components of the bacterial surface. Surface-layer proteins (Slps) are located in a paracrystalline layer outside the bacterial cell wall and are thought to play a role in tissue adherence. However, the ability of S-layer protein extract derived from probiotic bacteria to block adherence of enteric pathogens has not been investigated. Human epithelial (HEp-2 and T84) cells were treated with S-layer protein extract alone, infected with E. coli O157:H7, or pretreated with S-layer protein extract prior to infection to determine their importance in the inhibition of pathogen adherence. The effects of S-layer protein extracts were characterized by phase-contrast and immunofluorescence microscopy and measurement of the transepithelial electrical resistance of polarized monolayers. Pre-treatment of host epithelial cells with S-layer protein extracts prior to E. coli O157:H7 infection decreased pathogen adherence and attaching-effacing lesions in addition to preserving the barrier function of monolayers. These in vitro studies indicate that a non-viable constituent derived from a probiotic strain may prove effective in interrupting the infectious process of an intestinal pathogen.


Assuntos
Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/farmacologia , Células Epiteliais/efeitos dos fármacos , Escherichia coli O157/efeitos dos fármacos , Lactobacillus helveticus/química , Proteínas da Membrana Bacteriana Externa/isolamento & purificação , Proteínas de Bactérias/farmacologia , Linhagem Celular , Células Epiteliais/microbiologia , Escherichia coli O157/metabolismo
14.
Infect Immun ; 73(8): 5183-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16041036

RESUMO

The aim of this study was to determine if probiotics reduce epithelial injury following exposure to Escherichia coli O157:H7 and E. coli O127:H6. The pretreatment of intestinal (T84) cells with lactic acid-producing bacteria reduced the pathogen-induced drop in transepithelial electrical resistance. These findings demonstrate that probiotics prevent epithelial injury induced by attaching-effacing bacteria.


Assuntos
Aderência Bacteriana/fisiologia , Citoesqueleto/metabolismo , Células Epiteliais/metabolismo , Escherichia coli O157/metabolismo , Probióticos/metabolismo , Actinina/metabolismo , Células Epiteliais/microbiologia , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/terapia , Humanos , Lactobacillus acidophilus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA