Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Gastroenterol Clin Biol ; 34 Suppl 1: S62-70, 2010 Sep.
Artigo em Francês | MEDLINE | ID: mdl-20889007

RESUMO

This review summarizes the probiotic mechanisms of action of Saccharomyces boulardii (S. boulardii) against inflammatory and non-inflammatory diarrheal conditions. S. boulardii is distributed in lyophilized form in many countries and used for the prevention of diarrhea in children and adults, including Clostridium difficile (C. difficile) associated infection. The main mechanisms of action of S. boulardii include inhibition of activities of bacterial pathogenic products, trophic effects on the intestinal mucosa, as well as modification of host signaling pathways involved in inflammatory and non-inflammatory intestinal diseases. S. boulardii inhibits production of pro-inflammatory cytokines by inhibiting main regulators of inflammation, including nuclear factor κB (NF-κB), and mitogen-activated protein kinases (MAP kinases), ERK1/2 and p38, but stimulates production of anti-inflammatory molecules such as peroxisome proliferator-activated receptor-gamma (PPAR-γ). Moreover, S. boulardii suppresses bacterial infection by inhibiting adhesion and/or overgrowth of bacteria, produces a serine protease that cleaves C. difficile toxin A, and stimulates antibody production against this toxin. Furthermore, S. boulardii may interfere with pathogenesis of Inflammatory Bowel Disease (IBD) by acting on T cells and acts in diarrheal conditions by improving the fecal biostructure in patients with diarrhea. These diverse mechanisms exerted by S. boulardii provide molecular clues for its effectiveness in diarrheal diseases and intestinal inflammatory conditions with an inflammatory component.


Assuntos
Diarreia/prevenção & controle , Probióticos/uso terapêutico , Saccharomyces , Adesão Celular/efeitos dos fármacos , Citocinas/biossíntese , Diarreia/dietoterapia , Diarreia/microbiologia , Humanos , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , PPAR gama/biossíntese , Serina Proteases/efeitos dos fármacos
2.
Gut ; 58(1): 34-40, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18824554

RESUMO

OBJECTIVE: Melanin-concentrating hormone (MCH) is a hypothalamic orexigenic neuropeptide that regulates energy balance. However, the distribution of MCH and its receptor MCHR1 in tissues other than brain suggested additional, as yet unappreciated, roles for this neuropeptide. Based on previous paradigms and the presence of MCH in the intestine as well as in immune cells, its potential role in gut innate immune responses was examined. METHODS: In human intestinal xenografts grown in mice, changes in the expression of MCH and its receptors following treatment with Clostridium difficile toxin A, the causative agent of antibiotic-associated diarrhoea in hospitalised patients, were examined. In colonocytes, the effect of C difficile toxin A treatment on MCHR1 expression, and of MCH on interleukin 8 (IL8) expression was examined. MCH-deficient mice and immunoneutralisation approaches were used to examine the role of MCH in the pathogenesis of C difficile toxin A-mediated acute enteritis. RESULTS: Upregulation of MCH and MCHR1 expression was found in the human intestinal xenograft model, and of MCHR1 in colonocytes following exposure to toxin A. Treatment of colonocytes with MCH resulted in IL8 transcriptional upregulation, implying a link between MCH and inflammatory pathways. In further support of this view, MCH-deficient mice developed attenuated toxin A-mediated intestinal inflammation and secretion, as did wild-type mice treated with an antibody against MCH or MCHR1. CONCLUSION: These findings signify MCH as a mediator of C difficile-associated enteritis and possibly of additional gut pathogens. MCH may mediate its proinflammatory effects at least in part by acting on epithelial cells in the intestine.


Assuntos
Toxinas Bacterianas/toxicidade , Enterotoxinas/toxicidade , Hormônios Hipotalâmicos/fisiologia , Ileíte/microbiologia , Melaninas/fisiologia , Hormônios Hipofisários/fisiologia , Animais , Colo/metabolismo , Colo/transplante , Células Epiteliais/metabolismo , Humanos , Hormônios Hipotalâmicos/genética , Hormônios Hipotalâmicos/imunologia , Ileíte/metabolismo , Ileíte/patologia , Ileíte/prevenção & controle , Masculino , Melaninas/genética , Melaninas/imunologia , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Hormônios Hipofisários/genética , Hormônios Hipofisários/imunologia , RNA Mensageiro/genética , Receptores de Somatostatina/genética , Receptores de Somatostatina/imunologia , Receptores de Somatostatina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transplante Heterólogo , Regulação para Cima
3.
J Clin Invest ; 82(5): 1516-24, 1988 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-3141478

RESUMO

Toxin A of Clostridium difficile causes severe inflammatory enterocolitis in man and animals that appears to be mediated in part by acute inflammatory cells that migrate into the toxin A-exposed mucosa. To determine the direct effects of toxin A on intestinal epithelial permeability and structure in the absence of other modulating factors, we used cultured monolayers of a human intestinal epithelial cell line (T84). A toxin A concentration of 7 x 10(-1) micrograms/ml (3 x 10(-9) M) nearly abolished monolayer transepithelial resistance within 6-8 h. This marked permeability defect occurred while the monolayers were still confluent. Dual sodium-mannitol flux studies localized the permeability defect to the intercellular tight junction. Cytotoxicity assays and morphological evaluation using Nomarski optics and electron microscopy failed to demonstrate any evidence of cell damage at the time the maximum resistance response was observed. Fluorescent staining for F actin, however, revealed a marked decrease in fluorescent intensity in toxin-treated monolayers versus controls. These data show that toxin A can directly affect the barrier function of this model intestinal epithelium and initially does so by selectively enhancing tight junction permeability. Furthermore, cytoskeletal structure is markedly altered over the same time course, although the integrity of individual cells is maintained. Because the cytoskeleton of intestinal epithelial cells is known to be capable of regulating tight junction permeability, we speculate that the above effects of toxin A on epithelial barrier function result from alterations of the cytoskeleton.


Assuntos
Toxinas Bacterianas/farmacologia , Enterotoxinas , Intestinos/ultraestrutura , Animais , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/ultraestrutura , Epitélio/efeitos dos fármacos , Epitélio/ultraestrutura , L-Lactato Desidrogenase/metabolismo , Manitol/farmacocinética , Microscopia Eletrônica , Microscopia de Contraste de Fase , Permeabilidade , Fatores de Tempo
4.
J Clin Invest ; 81(6): 1741-5, 1988 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2838520

RESUMO

Clostridium difficile, a common enteric pathogen, mediates tissue damage and intestinal fluid secretion by release of two protein exotoxins: toxin A, an enterotoxin, and toxin B, a cytotoxin. Because toxin A elicits an intense inflammatory reaction in vivo, we studied the effects of highly purified C. difficile toxins on activation of human granulocytes. Toxin A at concentrations of 10(-7) to 10(-6) M, but not toxin B, elicited a significant chemotactic and chemokinetic response by granulocytes that was comparable with that induced by the chemotactic factor N-FMLP (10(-7) M). Neither toxin stimulated release of superoxide anion from granulocytes. Toxin A produced a rapid, transient rise in cytosolic [Ca2+]i, as measured by quin 2 fluorescence. Pertussis toxin and depletion of intra- and extracellular calcium blocked the toxin A effect on cytosolic [Ca2+]i. These findings suggest that the inflammatory effects of C. difficile toxin A in the intestine may be related to its ability to mobilize intracellular Ca2+ and elicit a chemotactic response by granulocytes.


Assuntos
Toxinas Bacterianas/farmacologia , Cálcio/metabolismo , Quimiotaxia de Leucócito , Clostridium , Granulócitos/fisiologia , Cálcio/análise , Citosol/análise , Enterotoxinas/farmacologia , Granulócitos/análise , Granulócitos/metabolismo , Granulócitos/ultraestrutura , Humanos , Superóxidos/metabolismo
5.
J Clin Invest ; 88(1): 119-25, 1991 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1905325

RESUMO

The purpose of this study was to characterize the surface receptor for toxin A, the enterotoxin from Clostridium difficile, on rabbit intestinal brush borders (BB) and on rat basophilic leukemia (RBL) cells. Purified toxin A was radiolabeled using a modified Bolton-Hunter method to sp act 2 microCi/micrograms, with retention of full biologic activity. 3H-Toxin A bound specifically to a single class of receptors on rabbit BB and on RBL cells with dissociation constants of 5.4 x 10(-8) and 3.5 x 10(-8) M, respectively. RBL cells were highly sensitive to toxin A (cell rounding) and had 180,000 specific binding sites per cell, whereas IMR-90 fibroblasts were far less sensitive to toxin A and lacked detectable specific binding sites. Exposure of BB to trypsin or chymotrypsin significantly reduced 3H-toxin A specific binding. Preincubation of BB with Bandeirea simplicifolia (BS-1) lectin also reduced specific binding, and CHAPS-solubilized receptors could be immobilized with WGA-agarose. The addition of 100 nM toxin A accelerated the association of 35S-GTP gamma S with rabbit ileal BB, and preincubation of BB with the GTP analogues GTP gamma S or Gpp(NH)p, significantly reduced 3H-toxin A specific binding. Our data indicate that the membrane receptor for toxin A is a galactose and N-acetyl-glucosamine-containing glycoprotein which appears to be coupled to a G protein.


Assuntos
Toxinas Bacterianas/metabolismo , Clostridioides difficile , Enterotoxinas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Glicoproteínas de Membrana/metabolismo , Animais , Toxinas Bacterianas/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Íleo/metabolismo , Lectinas/farmacologia , Glicoproteínas de Membrana/isolamento & purificação , Microvilosidades/metabolismo , Coelhos , Ensaio Radioligante , Ratos , Trítio , Tripsina/farmacologia
6.
J Clin Invest ; 101(8): 1547-50, 1998 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9541482

RESUMO

Toxin A, a 308,000-Mr enterotoxin from Clostridium difficile, mediates antibiotic-associated diarrhea and colitis in humans. Injection of toxin A into animal intestine triggers an acute inflammatory response characterized by activation of sensory neurons and immune cells of the intestinal lamina propria, including mast cells and macrophages, and migration of circulating neutrophils in the involved intestinal segment. In this study we show that mice genetically deficient in the neurokinin-1 receptor are protected from the secretory and inflammatory changes as well as from epithelial cell damage induced by toxin A. The protective effect of neurokinin-1R deletion correlates with diminished intestinal levels of the cytokine TNF-alpha and its mRNA and the leukocyte enzyme myeloperoxidase. These results demonstrate a major requirement for substance P receptors in the pathogenesis of acute inflammatory diarrhea.


Assuntos
Clostridioides difficile/patogenicidade , Enterocolite Pseudomembranosa/etiologia , Receptores da Neurocinina-1/metabolismo , Animais , Toxinas Bacterianas/toxicidade , Enterocolite Pseudomembranosa/metabolismo , Enterocolite Pseudomembranosa/patologia , Enterotoxinas/toxicidade , Humanos , Íleo/efeitos dos fármacos , Íleo/metabolismo , Íleo/patologia , Técnicas In Vitro , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores da Neurocinina-1/deficiência , Receptores da Neurocinina-1/genética , Substância P/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
7.
J Clin Invest ; 103(6): 843-9, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10079105

RESUMO

The neuropeptide neurotensin mediates several intestinal functions, including chloride secretion, motility, and cellular growth. However, whether this peptide participates in intestinal inflammation is not known. Toxin A, an enterotoxin from Clostridium difficile, mediates pseudomembranous colitis in humans. In animal models, toxin A causes an acute inflammatory response characterized by activation of sensory neurons and intestinal nerves and immune cells of the lamina propria. Here we show that neurotensin and its receptor are elevated in the rat colonic mucosa following toxin A administration. Pretreatment of rats with the neurotensin receptor antagonist SR-48, 692 inhibits toxin A-induced changes in colonic secretion, mucosal permeability, and histologic damage. Exposure of colonic explants to toxin A or neurotensin causes mast cell degranulation, which is inhibited by SR-48,692. Because substance P was previously shown to mediate mast cell activation, we examined whether substance P is involved in neurotensin-induced mast cell degranulation. Our results show that neurotensin-induced mast cell degranulation in colonic explants is inhibited by the substance P (neurokinin-1) receptor antagonist CP-96,345, indicating that colonic mast activation in response to neurotensin involves release of substance P. We conclude that neurotensin plays a key role in the pathogenesis of C. difficile-induced colonic inflammation and mast cell activation.


Assuntos
Toxinas Bacterianas , Colite/metabolismo , Enterotoxinas/farmacologia , Neurotensina/farmacologia , Receptores de Neurotensina/metabolismo , Animais , Compostos de Bifenilo/farmacologia , Degranulação Celular , Colite/induzido quimicamente , Colite/etiologia , Técnicas In Vitro , Secreções Intestinais/efeitos dos fármacos , Mastócitos/efeitos dos fármacos , Antagonistas dos Receptores de Neurocinina-1 , Pirazóis/farmacologia , Quinolinas/farmacologia , Ratos , Receptor Cross-Talk , Receptores de Neurotensina/antagonistas & inibidores
8.
J Clin Invest ; 94(5): 1919-26, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7962537

RESUMO

Clostridium difficile toxin A (Tx-A) mediates secretion and inflammation in experimental enterocolitis. Intravital video microscopy was used to define the mechanisms that underlie the inflammatory reactions elicited by direct exposure of the microvasculature to Tx-A. Leukocyte adherence and emigration, leukocyte-platelet aggregation, and extravasation of FITC-albumin were monitored in rat mesenteric venules exposed to Tx-A. Significant increases in leukocyte adherence and emigration (LAE) and albumin leakage were noted within 15-30 min of Tx-A exposure. These responses were accompanied by mast cell degranulation and the formation of platelet-leukocyte aggregates. The Tx-A-induced increases in LAE and albumin leakage were significantly attenuated by pretreatment with either monoclonal antibodies (mAbs) directed against the leukocyte adhesion glycoproteins, CD11/CD18, intercellular adhesion molecule-1, and P-selectin (but not E-selectin) or with sialyl Lewis x, a counter-receptor for P-selectin. The mast cell stabilizer, lodoxamide, an H1- (but not an H2-) receptor antagonist, and diamine oxidase (histaminase) were also effective in reducing the LAE and albumin leakage elicited by Tx-A. The platelet-leukocyte aggregation response was blunted by an mAb against P-selectin, sialyl Lewis x, and the H1-receptor antagonist. These observations indicate that Tx-A induces a leukocyte-dependent leakage of albumin from postcapillary venules. Mast cell-derived histamine appears to mediate at least part of the leukocyte-endothelial cell adhesion and platelet-leukocyte aggregation by engaging H1-receptors on endothelial cells and platelets to increase the expression of P-selectin. The adhesion glycoproteins CD11/CD18 and intercellular adhesion molecule-1 also contribute to the inflammatory responses elicited by toxin A.


Assuntos
Toxinas Bacterianas/toxicidade , Clostridioides difficile/patogenicidade , Enterotoxinas/toxicidade , Histamina/fisiologia , Vênulas/efeitos dos fármacos , Animais , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/análise , Moléculas de Adesão Celular/fisiologia , Leucócitos/efeitos dos fármacos , Leucócitos/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Albumina Sérica/metabolismo , Vênulas/fisiologia
9.
J Clin Invest ; 93(3): 1257-65, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7907603

RESUMO

Neutrophil infiltration is a prominent feature of Clostridium difficile-associated enteritis and colitis. The aim of this study was to examine the importance of neutrophil recruitment and neutrophil-mediated tissue damage in C. difficile toxin A-induced enteritis. Competitive binding experiments using purified 3H-toxin A demonstrated the presence of a single class of medium affinity receptors on rabbit neutrophils (Kd 7 x 10(-8) M). Pertussis toxin and the nonhydrolyzable GTP analog GTPgamma S both inhibited 3H-toxin A binding (by 56 and 65%, respectively), indicating that the rabbit neutrophil toxin A receptor is G protein linked. Toxin A elicited a dose-dependent (25-200 micrograms/ml) stimulation of neutrophil migration in vitro, and this functional effect was also pertussis toxin sensitive (69% inhibition). Treatment of neutrophils with R15.7, a blocking monoclonal antibody to the leuocyte adhesion molecule CD18, inhibited toxin A-stimulated neutrophil migration by 85% in vitro. Pretreatment of rabbits with R15.7 also prevented neutrophil infiltration of toxin A-exposed ileal loops in vivo as determined by histologic examination and by ileal tissue myeloperoxidase levels. Furthermore, R15.7 effected a substantial inhibition of fluid secretion (by 65%), mannitol permeability (by 66%), and histologic damage in toxin A-exposed ileal loops. Anti-CD18 (R15.7) had no inhibitory effect on cholera toxin enterotoxicity. These data demonstrate that C. difficile toxin A is a proinflammatory toxin whose enterotoxic effects are substantially dependent upon neutrophil recruitment.


Assuntos
Toxinas Bacterianas/toxicidade , Clostridioides difficile/patogenicidade , Enterite/etiologia , Enterotoxinas/toxicidade , Neutrófilos/efeitos dos fármacos , Animais , Antígenos CD/fisiologia , Antígenos CD18 , Enterite/sangue , Enterotoxinas/metabolismo , Proteínas de Ligação ao GTP/fisiologia , Masculino , Neutrófilos/metabolismo , Coelhos
10.
J Clin Invest ; 90(3): 822-9, 1992 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1325998

RESUMO

Human infants are relatively resistant to Clostridium difficile-associated diarrhea and colitis compared to adults. In that toxin A is the major cause of intestinal damage with this organism, we compared toxin A receptor binding and biological effects in newborn vs adult rabbit ileum. Purified toxin A (M(r) 308 kD) was labeled with tritium or biotin with full retention of biologic activity. Appearance of specific toxin A brush border (BB) binding was strongly age dependent with minimal [3H]toxin A specific binding at 2 and 5 d of life, followed by gradual increase in binding to reach adult levels at 90 d. Absence of toxin A binding sites in newborn and presence in adult rabbits was confirmed by immunohistochemical studies using biotinylated toxin A. Toxin A (50 ng to 20 micrograms/ml) inhibited protein synthesis in 90-d-old rabbit ileal loops in a dose-dependent fashion. In contrast, inhibition of protein synthesis in 5-d-old rabbit ileum occurred only at the highest toxin A doses (5 and 20 micrograms/ml) and at all doses tested was significantly less than the adult rabbit ileum. In addition, toxin A (5 micrograms/ml) caused severe mucosal damage in adult rabbit ileal explants but had no discernable morphologic effect on 5-d-old rabbit intestine. Our data indicate that newborn rabbit intestine lacks BB receptors for toxin A. The absence of the high-affinity BB receptor for toxin A in the newborn period may explain lack of biologic responsiveness to purified toxin, and the absence of disease in human infants infected with this pathogen.


Assuntos
Animais Recém-Nascidos/metabolismo , Toxinas Bacterianas/toxicidade , Clostridioides difficile/metabolismo , Enterotoxinas/toxicidade , Guanilato Ciclase , Íleo/efeitos dos fármacos , Receptores de Superfície Celular/análise , Receptores de Peptídeos , Fatores Etários , Animais , Sítios de Ligação , Enterotoxinas/metabolismo , Íleo/metabolismo , Íleo/patologia , Microvilosidades/metabolismo , Biossíntese de Proteínas , Coelhos , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase
11.
J Clin Invest ; 105(8): 1147-56, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10772660

RESUMO

Clostridium difficile toxin A causes acute neutrophil infiltration and intestinal mucosal injury. In cultured cells, toxin A inactivates Rho proteins by monoglucosylation. In monocytes, toxin A induces IL-8 production and necrosis by unknown mechanisms. We investigated the role of mitogen-activated protein (MAP) kinases in these events. In THP-1 monocytic cells, toxin A activated the 3 main MAP kinase cascades within 1 to 2 minutes. Activation of p38 was sustained, whereas stimulation of extracellular signal-regulated kinases and c-Jun NH(2)-terminal kinase was transient. Rho glucosylation became evident after 15 minutes. IL-8 gene expression was reduced by 70% by the MEK inhibitor PD98059 and abrogated by the p38 inhibitor SB203580 or by overexpression of dominant-negative mutants of the p38-activating kinases MKK3 and MKK6. SB203580 also blocked monocyte necrosis and IL-1beta release caused by toxin A but not by other toxins. Finally, in mouse ileum, SB203580 prevented toxin A-induced neutrophil recruitment by 92% and villous destruction by 90%. Thus, in monocytes exposed to toxin A, MAP kinase activation appears to precede Rho glucosylation and is required for IL-8 transcription and cell necrosis. p38 MAP kinase also mediates intestinal inflammation and mucosal damage induced by toxin A.


Assuntos
Toxinas Bacterianas/farmacologia , Clostridioides difficile/imunologia , Enterite/imunologia , Enterotoxinas/farmacologia , Interleucina-8/biossíntese , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/imunologia , Animais , Toxinas Bacterianas/metabolismo , Linhagem Celular , Clostridioides difficile/metabolismo , Enterite/enzimologia , Enterite/microbiologia , Enterocolite Pseudomembranosa/enzimologia , Enterocolite Pseudomembranosa/imunologia , Enterocolite Pseudomembranosa/microbiologia , Enterotoxinas/metabolismo , Ativação Enzimática , Regulação da Expressão Gênica/efeitos dos fármacos , Glicosilação , Interleucina-8/genética , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/imunologia , Monócitos/metabolismo , Monócitos/patologia , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno , Proteínas rho de Ligação ao GTP/imunologia , Proteínas rho de Ligação ao GTP/metabolismo
12.
J Clin Invest ; 95(5): 2004-11, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7738167

RESUMO

Toxin A but not toxin B, appears to mediate intestinal damage in animal models of Clostridium difficile enteritis. The purpose of this study was to investigate the electrophysiologic and morphologic effects of purified C. difficile toxins A and B on human colonic mucosa in Ussing chambers. Luminal exposure of tissues to 16-65 nM of toxin A and 0.2-29 nM of toxin B for 5 h caused dose-dependent epithelial damage. Potential difference, short-circuit current and resistance decreased by 76, 58, and 46%, respectively, with 32 nM of toxin A and by 76, 55, and 47%, respectively, with 3 nM of toxin B, when compared with baseline (P < 0.05). 3 nM of toxin A did not cause electrophysiologic changes. Permeability to [3H]mannitol increased 16-fold after exposure to 32 nM of toxin A and to 3 nM of toxin B when compared with controls (P < 0.05). Light and scanning electron microscopy after exposure to either toxin revealed patchy damage and exfoliation of superficial epithelial cells, while crypt epithelium remained intact. Fluorescent microscopy of phalloidin-stained sections showed that both toxins caused disruption and condensation of cellular F-actin. Our results demonstrate that the human colon is approximately 10 times more sensitive to the damaging effects of toxin B than toxin A, suggesting that toxin B may be more important than toxin A in the pathogenesis of C. difficile colitis in man.


Assuntos
Proteínas de Bactérias , Toxinas Bacterianas/toxicidade , Colo , Enterotoxinas/toxicidade , Mucosa Intestinal/efeitos dos fármacos , Actinas/efeitos dos fármacos , Actinas/metabolismo , Transporte Biológico/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Clostridioides difficile , Citotoxinas/toxicidade , Eletrofisiologia , Epitélio/efeitos dos fármacos , Epitélio/fisiologia , Epitélio/ultraestrutura , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Cinética , Manitol/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Microscopia Eletrônica de Varredura , Fatores de Tempo
13.
J Clin Invest ; 98(3): 641-9, 1996 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8698855

RESUMO

The intestinal effects of Clostridium difficile toxin A are inidated by toxin binding to luminal enterocyte receptors. We reported previously that the rabbit ileal brush border (BB) receptor is a glycoprotein with an alpha-d-galactose containing trisaccharide in the toxin-binding domain (1991. J. Clin. Invest. 88:119-125). In this study we characterized the rabbit ileal BB receptor for this toxin. Purified toxin receptor peptides of 19 and 24 amino acids showed 100% homology with rabbit sucrase-isomaltase (SI). Guinea pig receptor antiserum reacted in Western blots with rabbit SI and with the purified toxin receptor. Antireceptor IgG blocked in vitro binding of toxin A to rabbit ileal villus cell BB. Furthermore, anti-SI IgG inhibited toxin A-induced secretion (by 78.1%, P < 0.01), intestinal permeability (by 80.8%, P < 0.01), and histologic injury (P < 0.01) in rabbit ileal loops in vivo. Chinese hamster ovary cells transfected with SI cDNA showed increased intracellular calcium increase in response to native toxin (holotoxin) or to a recombinant 873-amino acid peptide representing the receptor binding domain of toxin A. These data suggest that toxin A binds specifically to carbohydrate domains on rabbit ileal SI, and that such binding is relevant to signal transduction mechanisms that mediate in vitro and in vivo toxicity.


Assuntos
Toxinas Bacterianas , Clostridioides difficile/patogenicidade , Enterotoxinas/metabolismo , Intestino Delgado/metabolismo , Complexo Sacarase-Isomaltase/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cobaias , Masculino , Dados de Sequência Molecular , Coelhos , Transfecção
14.
Neurogastroenterol Motil ; 28(3): 410-22, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26670691

RESUMO

BACKGROUND: Irritable bowel syndrome (IBS) is a stress-sensitive disorder. Environmental factors including stress can trigger epigenetic changes, which have not been well-studied in IBS. We performed a pilot study investigating genome-wide DNA methylation of IBS patients and healthy controls (HCs) to identify potential epigenetic markers and associated pathways. Additionally, we investigated relationships of epigenetic changes in selected genes with clinical traits. METHODS: Twenty-seven IBS patients (59% women; 10 IBS-diarrhea, 8 IBS-constipation, 9 IBS-mixed) and 23 age- and sex-matched HCs were examined. DNA methylation from peripheral blood mononuclear cells (PBMCs) was measured using HM450 BeadChip, and representative methylation differences were confirmed by bisulphite sequencing. Gene expression was measured using quantitative PCR. Gastrointestinal (GI) and non-GI symptoms were measured using validated questionnaires. Associations were tested using non-parametric methods. KEY RESULTS: Genome-wide DNA methylation profiling of IBS patients compared with HCs identified 133 differentially methylated positions (DMPs) (mean difference ≥10%; p < 0.05). These genes were associated with gene ontology terms including glutathione metabolism related to oxidative stress and neuropeptide hormone activity. Validation by sequencing confirmed differential methylation of subcommissural organ (SCO)-Spondin (SSPO), glutathione-S-transferases mu 5 (GSTM5), and tubulin polymerization promoting protein genes. Methylation of two promoter CpGs in GSTM5 was associated with epigenetic silencing. Epigenetic changes in SSPO gene were positively correlated with hospital anxiety and depression scores in IBS patients (r > 0.4 and false discovery rate <0.05). CONCLUSIONS & INFERENCES: This study is the first to comprehensively explore the methylome of IBS patients. We identified DMPs in novel candidate genes which could provide new insights into disease mechanisms; however, these preliminary findings warrant confirmation in larger, independent studies.


Assuntos
Metilação de DNA , Síndrome do Intestino Irritável/genética , Leucócitos Mononucleares/metabolismo , Estresse Fisiológico/genética , Adulto , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Projetos Piloto , Reação em Cadeia da Polimerase
15.
Br J Pharmacol ; 136(2): 271-9, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12010776

RESUMO

1. The role of substance P and its high affinity neurokinin-1 receptor in colitis has not been fully elucidated. We assessed the participation of neurokinin-1 receptor in colitis using the 2,4,6,-trinitrobenzensulphonic acid and dextran sulphate-induced animal models of colitis and genetically-engineered, neurokinin-1 receptor-deficient mice. 2. Clinical signs, macroscopic and histologic damage associated with 2,4,6,-trinitrobenzensulphonic acid (12 days) and dextran sulphate (5 days) colitis were more severe in neurokinin-1 deficient than in wild-type mice, while immunoreactivities for epidermal growth factor and its receptor were similar in the colon of both mice strains before and after colitis. 3. Substance P, dose-dependently induced intestinal fibroblast proliferation and enhanced epidermal growth factor-induced proliferation in intestinal fibroblasts isolated from wild-type, but not from neurokinin-1 receptor deficient mice. 4. Substance P-induced intestinal fibroblast proliferation required the presence of epidermal growth factor receptor with kinase activity. Furthermore, substance P induced epidermal growth factor tyrosine phosphorylation and activation in normal intestinal fibroblasts. 5. Our results indicate that in mice lacking the neurokinin - 1 receptor, substance P plays a protective role in prolonged experimental colitis.


Assuntos
Colite/metabolismo , Receptores ErbB/fisiologia , Receptores da Neurocinina-1/fisiologia , Animais , Colite/induzido quimicamente , Colite/patologia , Colite/prevenção & controle , Relação Dose-Resposta a Droga , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Receptores da Neurocinina-1/deficiência , Substância P/farmacologia
16.
Ann N Y Acad Sci ; 915: 347-56, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11193598

RESUMO

Clostridium difficile is the primary agent responsible for many patients with antibiotic-associated diarrhea and almost all patients with pseudomembranous colitis following antibiotic therapy. C. difficile infection is the most frequent form of colitis in hospitals and nursing homes and affects millions of patients in the United States and abroad. The first event in the pathogenesis of C. difficile infection involves alterations of the indigenous colonic microflora by antibiotics, followed by colonization with C. difficile. C. difficile causes diarrhea and colitis by releasing two high molecular weight protein exotoxins, toxin A and toxin B, with potent cytotoxic and enterotoxic properties. Evidence presented here indicates that C. difficile toxins compromise the epithelial cell barrier by at least two pathophysiologic pathways, one involving disaggregation of actin microfilaments in colonocytes via glucosylation of the Rho family of proteins leading to epithelial cell destruction and opening of the tight junctions, whereas the other appears to involve early release of proinflammatory cytokines from intestinal epithelial cells probably via activation of MAP kinases. We speculate that cytokines released from intestinal epithelial cells in response to toxin A exposure will diffuse into the lamina propria and activate macrophages, enteric nerves, and sensory neurons to release SP, CGRP, and NT, which, in turn, interact with immune and inflammatory cells and amplify the inflammatory response. Dissection of this inflammatory cascade may help us understand the pathophysiology of inflammatory diarrhea caused by this important pathogen.


Assuntos
Proteínas de Bactérias , Toxinas Bacterianas/metabolismo , Clostridioides difficile , Enterocolite Pseudomembranosa/metabolismo , Enterotoxinas/metabolismo , Absorção Intestinal/fisiologia , Animais , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia
17.
Ann N Y Acad Sci ; 840: 635-48, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9629290

RESUMO

Previous studies showed that exposure of experimental animals to immobilization stress increases colonic motility and that these effects are mediated by release of corticotropin-releasing factor (CRF), Studies from our laboratory showed that 30-min immobilization stress of rats caused several not previously described colonic responses to stress, including increased colonic mucin and prostaglandin E2 (PGE2) secretion, increased colonic mucosal levels of cyclooxygenase-2 (COX-2) mRNA, and degranulation of colonic mast cells. These stress-associated colonic changes were reproduced by intravenous or intracerebral injection of CRF in conscious, nonstressed rats. Furthermore, pretreatment of rats with the CRF antagonist alpha-helical CRF9-41, hexamethonium, or the mast cell stabilizer lodoxamide inhibited our observed colon responses to immobilization stress. Our results indicate that CRF released during immobilization stress increases colonic transit via a neuronal pathway and stimulates colonic mucin release via activation of neurons and colonic mast cells. These results provide support for an important role for CRF in stress-mediated colonic responses and a link between the nervous and the immune systems.


Assuntos
Hormônio Liberador da Corticotropina/fisiologia , Sistema Imunitário/fisiopatologia , Intestinos/fisiopatologia , Sistema Nervoso/fisiopatologia , Neurotensina/fisiologia , Estresse Fisiológico/fisiopatologia , Animais , Humanos
18.
Eur J Gastroenterol Hepatol ; 8(11): 1041-7, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8944363

RESUMO

Clostridium difficile is now regarded as a major enteric pathogen in hospitals and nursing-home facilities. The pathophysiology of this pathogen involves alterations of the indigenous colonic flora by antibiotics, ingestion of spores and colonization by C. difficile, followed by release of its toxins. Although most of the research on the intestinal effects of C. difficile had been focused on its enterotoxin or toxin A, recent results indicate that toxin B, the cytotoxin of C. difficile, is also active in human colon. The cloning and sequencing of the toxin A and toxin B gene and the identification of the GTP-binding protein Rho as their intracellular target represent major advances in our understanding of the mode of action of these toxins. An important characteristic of C. difficile infection is the dramatic inflammation seen in pseudomembranous colitis. Recent studies indicate that an interplay between lamina propria neuroimmune cells and intestinal epithelial cells may be central in pathogenesis of this toxin-mediated inflammatory response.


Assuntos
Toxinas Bacterianas/efeitos adversos , Clostridioides difficile/classificação , Enterocolite Pseudomembranosa/fisiopatologia , Diarreia/etiologia , Diarreia/fisiopatologia , Enterocolite Pseudomembranosa/patologia , Humanos , Mucosa Intestinal/patologia
19.
J Pediatr Surg ; 33(7): 991-6; discussion 996-7, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9694083

RESUMO

BACKGROUND/PURPOSE: Previous studies have shown that intestinal crypt cell transplantation using biodegradable scaffolds can generate stratified epithelium reminiscent of embryonic gut. The authors propose to tissue engineer small intestine on biodegradable scaffolds by transplanting intestinal epithelial organoid units, which maintain the epithelial mesenchymal cell-cell interaction necessary for epithelial survival, proliferation, and differentiation. METHODS: Intestinal epithelial organoid units were isolated from neonatal Lewis rats by enzyme digestion and differential sedimentation. Organoid units were seeded on to tubular scaffolds made of nonwoven polyglycolic acid (PGA) sprayed with 5% polylactic acid (PLA). Polymers either were coated (28 constructs) or noncoated (33 constructs) with collagen type I. A total of 61 organoid unit polymer constructs were implanted into 61 animals. Animals were killed and constructs harvested at 2, 6, 7, 8, 9, 10, 12, and 14 weeks. RESULTS: Histological analysis showed formation of neomucosa characterized by columnar epithelium with goblet, and paneth cells were evident in 47 of the 61 constructs. The outer walls were composed of fibrovascular tissue, degradable polymer, extracellular matrix, and smooth muscle-like cells. Immunofluorescent microscopy showed apical staining of brush border enzymes, sucrase and lactase, and basolateral staining for laminin, indicating the establishment of cell polarity. Electrophysiology of Ussing-chambered neomucosa and adult ileal mucosa exhibited similar transepithelial resistance. CONCLUSION: These results suggest that intestinal crypt cells heterotopically transplanted as epithelial organoid units on PGA-PLA tubular scaffolds can survive, reorganize, and regenerate complex composite tissue resembling small intestine demonstrating organ morphogenesis, cytodifferentiation, and phenotypic maturation.


Assuntos
Membrana Basal/fisiologia , Mucosa Intestinal/transplante , Microvilosidades/enzimologia , Organoides/transplante , Transplante Isogênico/métodos , Animais , Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Eletrofisiologia , Feminino , Sobrevivência de Enxerto/fisiologia , Imuno-Histoquímica , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Masculino , Microvilosidades/ultraestrutura , Ácido Poliglicólico , Próteses e Implantes , Ratos , Ratos Endogâmicos Lew , Síndrome do Intestino Curto/cirurgia
20.
Mucosal Immunol ; 7(6): 1492-503, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24850426

RESUMO

Intestinal fibrostenosis is among the hallmarks of severe Crohn's disease. Patients with certain TNFSF15 (gene name for TL1A) variants over-express TL1A and have a higher risk of developing strictures in the small intestine. In addition, sustained Tl1a expression in mice leads to small and large intestinal fibrostenosis under colitogenic conditions. The aim of this study was to determine whether established murine colonic fibrosis could be reversed with Tl1a antibody (Ab). Treatment with neutralizing Tl1a Ab reversed colonic fibrosis back to the original pre-inflamed levels, potentially as a result of lowered expression of connective tissue growth factor, Il31Ra, transforming growth factor ß1 and insulin-like growth factor-1. In addition, blocking Tl1a function by either neutralizing Tl1a Ab or deletion of death domain receptor 3 (Dr3) reduced the number of fibroblasts and myofibroblasts, the primary cell types that mediate tissue fibrosis. Primary intestinal myofibroblasts expressed Dr3 and functionally responded to direct Tl1a signaling by increasing collagen and Il31Ra expression. These data demonstrated a direct role for TL1A-DR3 signaling in tissue fibrosis and that modulation of TL1A-DR3 signaling could inhibit gut fibrosis.


Assuntos
Colo/imunologia , Doença de Crohn/imunologia , Transdução de Sinais/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Colo/patologia , Doença de Crohn/genética , Doença de Crohn/patologia , Fibrose , Humanos , Camundongos , Camundongos Knockout , Miofibroblastos/imunologia , Miofibroblastos/patologia , Receptores de Interleucina/genética , Receptores de Interleucina/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa