Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Curr Top Microbiol Immunol ; 357: 105-36, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21983749

RESUMO

The kidneys are the major organs affected in diarrhea-associated hemolytic uremic syndrome (D(+)HUS). The pathophysiology of renal disease in D(+)HUS is largely the result of the interaction between bacterial virulence factors such as Shiga toxin and lipopolysaccharide and host cells in the kidney and in the blood circulation. This chapter describes in detail the current knowledge of how these bacterial toxins may lead to kidney disease and renal failure. The toxin receptors expressed by specific blood and resident renal cell types are also discussed as are the actions of the toxins on these cells.


Assuntos
Síndrome Hemolítico-Urêmica/complicações , Síndrome Hemolítico-Urêmica/etiologia , Insuficiência Renal/etiologia , Toxina Shiga/metabolismo , Fatores de Virulência/metabolismo , Células Sanguíneas/metabolismo , Fibrinólise , Glicolipídeos/metabolismo , Síndrome Hemolítico-Urêmica/fisiopatologia , Humanos , Inflamação/metabolismo , Rim/citologia , Rim/metabolismo , Rim/fisiopatologia , Lipopolissacarídeos/metabolismo , Insuficiência Renal/fisiopatologia , Insuficiência Renal/terapia , Esfingolipídeos/metabolismo , Trombose/etiologia
2.
Infect Immun ; 77(3): 959-69, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19124603

RESUMO

Hemolytic-uremic syndrome (HUS) caused by Shiga toxin-producing Escherichia coli infection is a leading cause of pediatric acute renal failure. Bacterial toxins produced in the gut enter the circulation and cause a systemic toxemia and targeted cell damage. It had been previously shown that injection of Shiga toxin 2 (Stx2) and lipopolysaccharide (LPS) caused signs and symptoms of HUS in mice, but the mechanism leading to renal failure remained uncharacterized. The current study elucidated that murine cells of the glomerular filtration barrier were unresponsive to Stx2 because they lacked the receptor glycosphingolipid globotriaosylceramide (Gb(3)) in vitro and in vivo. In contrast to the analogous human cells, Stx2 did not alter inflammatory kinase activity, cytokine release, or cell viability of the murine glomerular cells. However, murine renal cortical and medullary tubular cells expressed Gb(3) and responded to Stx2 by undergoing apoptosis. Stx2-induced loss of functioning collecting ducts in vivo caused production of increased dilute urine, resulted in dehydration, and contributed to renal failure. Stx2-mediated renal dysfunction was ameliorated by administration of the nonselective caspase inhibitor Q-VD-OPH in vivo. Stx2 therefore targets the murine collecting duct, and this Stx2-induced injury can be blocked by inhibitors of apoptosis in vivo.


Assuntos
Apoptose/fisiologia , Epitélio/patologia , Túbulos Renais Coletores/patologia , Toxina Shiga II/metabolismo , Animais , Antígenos Glicosídicos Associados a Tumores/metabolismo , Western Blotting , Células Cultivadas , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Epitélio/imunologia , Epitélio/metabolismo , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Imunofluorescência , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Túbulos Renais Coletores/imunologia , Túbulos Renais Coletores/metabolismo , Lipopolissacarídeos/imunologia , Camundongos , Toxina Shiga II/imunologia , Veias Umbilicais
3.
Cell Microbiol ; 10(7): 1468-77, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18331592

RESUMO

Shiga toxins (Stxs) and ricin initiate damage to host cells by cleaving a single adenine residue on the alpha-sarcin loop of the 28S ribosomal RNA. This molecular insult results in a cascade of intracellular events termed the ribotoxic stress response (RSR). Although Stxs and ricin have been shown to cause the RSR, the mitogen-activated protein kinase kinase kinase (MAP3K) that transduces the signal from intoxicated ribosomes to activate SAPKinases has remained elusive. We show in vitro that DHP-2 (7-[3-fluoro-4-aminophenyl-(4-(2-pyridin-2-yl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl))]-quinoline), a zipper sterile-alpha-motif kinase (ZAK)-specific inhibitor, blocks Stx2/ricin-induced SAPKinase activation. Treatment of cells with DHP-2 also blocks Stx2/ricin-mediated upregulation of the proinflammatory cytokine interleukin-8 and results in a modest but statistically significant improvement in cell viability following Stx2/ricin treatment. Finally we show that siRNA directed against the N-terminus of ZAK diminishes Stx2/Ricin-induced SAPKinase activation. Together, these data demonstrate that a ZAK isoform(s) is the MAP3Kinase that transduces the RSR. Therefore, ZAKalpha and/or beta isoforms may act as potential therapeutic target(s) for treating Stx/ricin-associated illnesses. Furthermore, a small molecule inhibitor like DHP-2 may prove valuable in preventing the Stx/ricin-induced proinflammatory and/or apoptotic effects that are thought to contribute to pathogenesis by Stx-producing Escherichia coli and ricin.


Assuntos
Citocinas/metabolismo , Regulação da Expressão Gênica , Proteínas Quinases/metabolismo , Ricina/metabolismo , Toxina Shiga II/metabolismo , Idoso , Animais , Linhagem Celular , Criança , Chlorocebus aethiops , Citocinas/genética , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Interleucina-8/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinases , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases/genética , Pirazóis/metabolismo , Quinolinas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Escherichia coli Shiga Toxigênica/metabolismo , Escherichia coli Shiga Toxigênica/patogenicidade , Células Vero , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Infect Immun ; 76(3): 1115-21, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18086809

RESUMO

Escherichia coli O157:H7 Shiga toxin 2 (Stx2), one of the causative agents of hemolytic-uremic syndrome, is toxic to endothelial cells, including primary cultured human umbilical vein endothelial cells (HUVEC). This sensitivity of cells to Stx2 can be increased with either lipopolysaccharide (LPS) or tumor necrosis factor alpha (TNF-alpha). The goal of the present study was to identify the intracellular signaling pathway(s) by which LPS and TNF-alpha sensitize HUVEC to the cytotoxic effects of Stx2. To identify these pathways, specific pharmacological inhibitors and small interfering RNAs were tested with cell viability endpoints. A time course and dose response experiment for HUVEC exposure to LPS and TNF-alpha showed that a relatively short exposure to either agonist was sufficient to sensitize the cells to Stx2 and that both agonists stimulated intracellular signaling pathways within a short time. Cell viability assays indicated that the p38 mitogen-activated protein kinase (MAPK) inhibitors SB202190 and SB203580 and the general protein synthesis inhibitor cycloheximide inhibited both the LPS and TNF-alpha sensitization of HUVEC to Stx2, while all other inhibitors tested did not inhibit this sensitization. Additionally, SB202190 reduced the cellular globotriaosylceramide content under LPS- and TNF-alpha-induced conditions. In conclusion, our results show that LPS and TNF-alpha induction of Stx2 sensitivity in HUVEC is mediated through a pathway that includes p38 MAPK. These results indicate that inhibition of p38 MAPK in endothelial cells may protect a host from the deleterious effects of Stx2.


Assuntos
Células Endoteliais/microbiologia , Escherichia coli O157/fisiologia , Lipopolissacarídeos/metabolismo , Toxina Shiga II/toxicidade , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Sobrevivência Celular , Células Cultivadas , Cicloeximida/farmacologia , Relação Dose-Resposta a Droga , Células Endoteliais/química , Células Endoteliais/imunologia , Inibidores Enzimáticos/farmacologia , Inativação Gênica , Humanos , Imidazóis/farmacologia , Piridinas/farmacologia , Fatores de Tempo , Triexosilceramidas/análise , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
5.
Infect Immun ; 76(8): 3679-89, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18541659

RESUMO

Shiga toxin 1 (Stx1) and Stx2 produced by Escherichia coli O157 are known to be cytotoxic to Vero and HeLa cells by inhibiting protein synthesis and by inducing apoptosis. In the present study, we have demonstrated that 10 ng/ml Stx2 induced DNA fragmentation in human brain microvascular endothelial cells (HBMEC), with cleavage activation of caspase-3, -6, -8, and -9. A microarray approach used to search for apoptotic potential signals in response to Stx2 revealed that Stx2 treatment induced a marked upregulation of C/EBP homologous protein (CHOP)/growth arrest and DNA damage-inducible protein 153 (GADD153). Increased CHOP expression was dependent on enzymatically active Stx1. Knockdown of CHOP mRNA reduced the activation of caspase-3 and prevented apoptotic cell death. These results suggest that Stx2-induced apoptosis is mediated by CHOP in HBMEC and involves activation of both the intrinsic and extrinsic pathways of apoptosis.


Assuntos
Apoptose , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/microbiologia , Toxina Shiga II/toxicidade , Fator de Transcrição CHOP/biossíntese , Caspases/metabolismo , Sobrevivência Celular , Células Cultivadas , Cromatina/ultraestrutura , Fragmentação do DNA , Perfilação da Expressão Gênica , Inativação Gênica , Humanos , Microscopia Eletrônica de Transmissão , Análise de Sequência com Séries de Oligonucleotídeos , Sintaxina 1/genética , Sintaxina 1/metabolismo
6.
Lab Invest ; 88(11): 1178-91, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18779782

RESUMO

Effects in the liver of fatal intoxication with the binary toxin ricin are unclear. We report a robust neutrophil influx into the liver of C57BL/6 mice after lethal parenteral ricin challenge, occurring in peri-portal and centro-lobular hepatic areas within 2 h, followed by the abrupt disappearance of hepatic macrophages/Kupffer cells. Chemokine profiles determined by microarray, ribonuclease protection assays, northern blotting, and enzyme-linked immunosorbent assays showed rapid (2 h) upregulation and persistence of those for neutrophils (CXCL1/KC, CXCL2/MIP-2) and monocytes (CCL2/MCP-1). Red blood cell pooling (8-12 h), loss of hepatocyte glycogen (8-48 h) associated with progressive hypoglycemia, fibrin deposition (24-48 h), and death (72-96 h) followed. Monoclonal antibody to ricin A chain, administered intravenously, blunted hypoglycemia, and abrogated death. This outcome was observed when anti-ricin antibody was given before toxin exposure as well as when administered approximately 10 h after toxin exposure. Targeting antibody to specific amino-acid sequences on the ricin A chain (HAEL and QXXWXXA) was critical to the therapeutic effect. Re-emergence of liver macrophages/Kupffer cells and replenishment of glycogen in previously depleted hepatocytes preceded full recovery of the host. These data identify critical events for liver injury and healing in ricin intoxication, as well as a new means and specific targets for post-exposure therapeutic intervention.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Especificidade de Anticorpos/imunologia , Substâncias para a Guerra Química/intoxicação , Ricina/imunologia , Ricina/intoxicação , Animais , Substâncias para a Guerra Química/química , Modelos Animais de Doenças , Epitopos/imunologia , Glicogênio/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Hipoglicemia/induzido quimicamente , Hipoglicemia/prevenção & controle , Células de Kupffer/fisiologia , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Intoxicação/tratamento farmacológico , Intoxicação/patologia , Intoxicação/fisiopatologia , Recuperação de Função Fisiológica/fisiologia , Ricina/química
7.
BMC Infect Dis ; 8: 141, 2008 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-18937852

RESUMO

BACKGROUND: The pathophysiology of sepsis is due in part to early systemic inflammation. Here we describe molecular and cellular responses, as well as survival, in A 2A adenosine receptor (AR) agonist treated and untreated animals during experimental sepsis. METHODS: Sepsis was induced in mice by intraperitoneal inoculation of live bacteria (Escherichia coli or Staphylococcus aureus) or lipopolysaccharide (LPS). Mice inoculated with live bacteria were treated with an A 2A AR agonist (ATL313) or phosphate buffered saline (PBS), with or without the addition of a dose of ceftriaxone. LPS inoculated mice were treated with ATL313 or PBS. Serum cytokines and chemokines were measured sequentially at 1, 2, 4, 8, and 24 hours after LPS was administered. In survival studies, mice were followed until death or for 7 days. RESULTS: There was a significant survival benefit in mice infected with live E. coli (100% vs. 20%, p = 0.013) or S. aureus (60% vs. 20%, p = 0.02) when treated with ATL313 in conjunction with an antibiotic versus antibiotic alone. ATL313 also improved survival from endotoxic shock when compared to PBS treatment (90% vs. 40%, p = 0.005). The serum concentrations of TNF-alpha, MIP-1 alpha, MCP-1, IFN-gamma, and IL-17 were decreased by ATL313 after LPS injection (p < 0.05). Additionally, ATL313 increased the concentration of IL-10 under the same conditions (p < 0.05). Circulating white blood cell concentrations were higher in ATL313 treated animals (p < 0.01). CONCLUSION: Further studies are warranted to determine the clinical utility of ATL313 as a novel treatment for sepsis.


Assuntos
Anti-Inflamatórios/uso terapêutico , Inflamação/tratamento farmacológico , Piperidinas/uso terapêutico , Receptor A2A de Adenosina/efeitos dos fármacos , Sepse/tratamento farmacológico , Animais , Ceftriaxona/uso terapêutico , Citocinas/sangue , Modelos Animais de Doenças , Infecções por Escherichia coli/tratamento farmacológico , Feminino , Leucócitos/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/tratamento farmacológico , Análise de Sobrevida
8.
Anesth Analg ; 102(4): 1108-13, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16551907

RESUMO

Lidocaine attenuates cell injury induced by ischemic-reperfusion and inflammation, although the protective mechanisms are not understood. We hypothesized that lidocaine and other amide local anesthetics protect against endothelial cell injury through activation of the mitochondrial adenosine triphosphate-sensitive potassium (mitoK(ATP)) channels. We determined the effects of amide local anesthetics (lidocaine, ropivacaine, and bupivacaine), ester local anesthetics (tetracaine and procaine), one amide analog (YWI), and two non-amide local anesthetic analogs (JDA and ICM) on viability of human microvascular endothelial cells after exposure to lipopolysaccharide (LPS) in the absence or presence of the mitoK(ATP) channel antagonist 5-hydroxydecaonate. Flavoprotein fluorescence was used to investigate the effects of local anesthetics on diazoxide-induced activation of mitoK(ATP) channels. Lidocaine, ropivacaine, bupivicaine, YWI, JDA, and ICM attenuated by 60% to 70% the decrease in cell viability caused by LPS. Amide local anesthetics and YWI protection was inhibited by 5-hydroxydecaonate, whereas the protection induced by JDA and ICM was not. Tetracaine and procaine did not protect against LPS-induced injury. The amide local anesthetics and the amide analog (YWI) enhanced diazoxide-induced flavoprotein fluorescence by 5% to 20%, whereas ester local anesthetics decreased diazoxide-induced flavoprotein fluorescence by 5% to 60% and the non-amide local anesthetic analogs had no effect. In conclusion, amide local anesthetics and the amide analog (YWI) attenuate LPS-induced cell injury, in part, through activation of mitoK(ATP) channels. In contrast, tetracaine and procaine had no protective effects and inhibited activation of mitoK(ATP) channels. The non-amide local anesthetic analogs induced protection but through mechanisms independent of mitoK(ATP) channels.


Assuntos
Anestésicos Locais/farmacologia , Citoproteção/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Lipopolissacarídeos/efeitos adversos , Mitocôndrias/efeitos dos fármacos , Canais de Potássio/fisiologia , Trifosfato de Adenosina/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Citoproteção/fisiologia , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Humanos , Mitocôndrias/fisiologia
9.
Thromb Haemost ; 94(5): 1019-27, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16366002

RESUMO

Platelet and monocyte activation may contribute to hemolytic anemia, thrombocytopenia and renal failure associated with the hemolytic uremic syndrome (HUS) caused by Escherichia coli O157:H7. Since Shiga toxins (Stxs) and lipopolysaccharide (LPS) from this bacterium are implicated in the pathogenesis of HUS, we examined whether stimulation of THP-1 human monocytic cells by Shiga toxin 2 (Stx2) and LPS can lead to the activation of platelet function. We now show that Stx2 causedTHP-1 cells to release the chemokines IL-8, MDC, and RANTES and that the presence of LPS further stimulated this release. IL-8 was produced in greatest amount and was an effective co-agonist for inducing platelet aggregation. Primary human monocytes also released large amounts of IL-8 in response to LPS and Stx2. Factors released byTHP-1 cells exposed to Stx2 and LPS activated platelet function as evidenced by increased aggregation, serotonin secretion, P-selectin exposure and by the formation of stable platelet-monocyte aggregates. Our data therefore show that monocytes exposed to E.coli-derived Stx2 and LPS release factors which activate platelet function.


Assuntos
Lipopolissacarídeos/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Agregação Plaquetária/fisiologia , Toxina Shiga II/farmacologia , Plaquetas/fisiologia , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Quimiocina CCL22 , Quimiocina CCL5/metabolismo , Quimiocinas CC/metabolismo , Grânulos Citoplasmáticos/fisiologia , Humanos , Interleucina-8/metabolismo , Leucemia , Monócitos/citologia
10.
Nutrition ; 20(10): 934-41, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15474885

RESUMO

OBJECTIVE: In this study, we postulated the beneficial role of oral alanyl-glutamine, a more stable glutamine derivative to decrease 5-fluorouracil (5-FU)-induced mucositis in mice. METHODS: We measured different morphologic parameters to assess structural changes over time in the small bowel, including crypt depth, villus height, villus area, mitotic and apoptotic indices at the crypt level using terminal deoxyuridine triphosphate nick end labeling, and hematoxylin-eosin staining of ileal tissue. In addition, we analyzed the effect of different alanyl-glutamine concentrations on animal weight curves after 5-FU treatment. RESULTS: Neither glutamine nor alanyl- glutamine prevented the 5-FU intestinal structural damage or apoptosis in crypt enterocytes at 24 h after 5-FU challenge. However, we found that alanyl-glutamine, but not glutamine, speeds intestinal recovery when compared with 5-FU-treated controls (P < 0.05), predominantly by enhancing mitotic activity and crypt length. CONCLUSION: Our findings provide important data to support clinical studies of oral alanyl-glutamine in 5-FU-induced mucositis.


Assuntos
Dipeptídeos/administração & dosagem , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Animais , Antimetabólitos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fluoruracila/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C
11.
Microbiol Spectr ; 2(3)2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25530918

RESUMO

Shiga toxin (Stx) is the primary cause of severe host responses including renal and central nervous system (CNS) disease in Shiga toxin-producing E. coli (STEC) infections. The interaction of Stx with different eukaryotic cell types is described. Host responses to Stx and bacterial lipopolysaccharide (LPS) are compared as related to the features of the STEC-associated Hemolytic Uremic Syndrome (HUS). Data derived from animal models of HUS and CNS disease, in vivo, and eukaryotic cells, in vitro, are evaluated in relation to HUS disease of humans.


Assuntos
Doenças do Sistema Nervoso Central/microbiologia , Infecções por Escherichia coli/microbiologia , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Síndrome Hemolítico-Urêmica/microbiologia , Toxinas Shiga/metabolismo , Fatores de Virulência/metabolismo , Animais , Doenças do Sistema Nervoso Central/patologia , Técnicas Citológicas , Modelos Animais de Doenças , Infecções por Escherichia coli/patologia , Síndrome Hemolítico-Urêmica/patologia , Humanos , Virulência
12.
Clin Vaccine Immunol ; 19(12): 1932-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23035176

RESUMO

We have developed a novel vaccine against Shiga toxin (Stx)-producing Escherichia coli (STEC) infection using a recombinant Mycobacterium bovis BCG (rBCG) system. Two intraperitoneal vaccinations with rBCG expressing the Stx2 B subunit (Stx2B) resulted in an increase of protective serum IgG and mucosal IgA responses to Stx2B in BALB/c mice. When orally challenged with 10(3) CFU of STEC strain B2F1 (O91: H21), the immunized mice survived statistically significantly longer than the nonvaccinated mice. We suggest that intraperitoneal immunization with rBCG expressing Stx2B would be a potential vaccine strategy for STEC.


Assuntos
Vacina BCG/genética , Portadores de Fármacos , Vacinas contra Escherichia coli/imunologia , Vetores Genéticos , Toxina Shiga II/imunologia , Escherichia coli Shiga Toxigênica/imunologia , Animais , Vacina BCG/imunologia , Modelos Animais de Doenças , Infecções por Escherichia coli/prevenção & controle , Vacinas contra Escherichia coli/administração & dosagem , Vacinas contra Escherichia coli/genética , Feminino , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos BALB C , Toxina Shiga II/genética , Escherichia coli Shiga Toxigênica/genética , Análise de Sobrevida , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
13.
Toxins (Basel) ; 2(12): 2769-2794, 2010 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-21297888

RESUMO

Shiga toxin-producing Escherichia coli is a contaminant of food and water that in humans causes a diarrheal prodrome followed by more severe disease of the kidneys and an array of symptoms of the central nervous system. The systemic disease is a complex referred to as diarrhea-associated hemolytic uremic syndrome (D(+)HUS). D(+)HUS is characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure. This review focuses on the renal aspects of D(+)HUS. Current knowledge of this renal disease is derived from a combination of human samples, animal models of D(+)HUS, and interaction of Shiga toxin with isolated renal cell types. Shiga toxin is a multi-subunit protein complex that binds to a glycosphingolipid receptor, Gb3, on select eukaryotic cell types. Location of Gb3 in the kidney is predictive of the sites of action of Shiga toxin. However, the toxin is cytotoxic to some, but not all cell types that express Gb3. It also can cause apoptosis or generate an inflammatory response in some cells. Together, this myriad of results is responsible for D(+)HUS disease.

14.
Toxins (Basel) ; 2(8): 1997-2006, 2010 08.
Artigo em Inglês | MEDLINE | ID: mdl-20725533

RESUMO

We have shown previously that neurons in the mouse spinal cord express Gb(3). We show in this article that distribution of anti-Gb(3)-Ab reactivity occurs in many different types of neurons of different areas of the central nervous system (CNS). The immunoreactive neurons are in olfactory bulbs, cerebral cortex, hippocampus, striatum, amygdala, thalamus, hypothalamus, cerebellum, and medulla oblongata. In several different circumventricular organs where vessels do not have the blood-brain-barrier (BBB) structure, anti-Gb(3)-Ab is not positive for vessel structures, while neurons at these regions are positive. Also, within the ventricular area, ependymal cells in the third ventricle express Gb(3), as revealed by anti-Gb(3)-Ab staining and intensity analysis.

15.
Histochem Cell Biol ; 130(1): 157-64, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18365234

RESUMO

Shiga toxin-producing Escherichia coli causes hemolytic uremic syndrome, a constellation of disorders that includes kidney failure and central nervous system dysfunction. Shiga toxin binds the amphipathic, membrane-bound glycolipid globotriaosylceramide (Gb(3)) and uses it to enter host cells and ultimately cause cell death. Thus, cell types that express Gb(3) in target tissues should be recognized. The objective of this study was to determine whether immunohistologic detection of Gb(3) was affected by the method of tissue preparation. Tissue preparation included variations in fixation (immersion or perfusion) and processing (paraffin or frozen) steps; paraffin processing employed different dehydration solvents (acetone or ethanol). Perfusion-fixation in combination with frozen sections or acetone-dehydrated tissue for paraffin sections resulted in specific recognition of Gb(3) using immunohistochemical or immunofluorescent methods. In the mouse tissues studied, Gb(3) was associated with tubules in the kidney and neurons in the nervous system. On the other hand, Gb(3) localization to endothelial cells was determined to be an artifact generated due to immersion-fixation or tissue dehydration with ethanol. This finding was corroborated by glycolipid profiles from tissue subjected to dehydration; namely Gb(3) was subject to extraction by ethanol more than acetone during tissue dehydration. The results of this study show that tissue preparation is crucial to the persistence and preservation of the glycolipid Gb(3) in mouse tissue. These methods may serve as a basis for determining the localization of other amphipathic glycolipids in tissue.


Assuntos
Técnica Direta de Fluorescência para Anticorpo/métodos , Secções Congeladas/métodos , Túbulos Renais/química , Neurônios/química , Inclusão em Parafina/métodos , Triexosilceramidas/análise , Animais , Anticorpos Monoclonais/imunologia , Síndrome Hemolítico-Urêmica/metabolismo , Síndrome Hemolítico-Urêmica/microbiologia , Túbulos Renais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Toxinas Shiga/metabolismo , Escherichia coli Shiga Toxigênica/metabolismo , Triexosilceramidas/imunologia , Triexosilceramidas/metabolismo
16.
J Infect Dis ; 198(9): 1398-406, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18754742

RESUMO

Affinity-purified Shiga toxin (Stx) 2 given intraperitoneally to mice caused weight loss and hind-limb paralysis followed by death. Globotriaosylceramide (Gb(3)), the receptor for Stx2, was localized to neurons of the central nervous system (CNS) of normal mice. Gb3 was not found in astrocytes or endothelial cells of the CNS. In human cadaver CNS, we found Gb(3) in neurons and endothelial cells. Mouse Gb(3) localization was confirmed by immunoelectron microscopy. In Stx2-exposed mice, anti-Stx2-gold immunoreaction was positive in neurons. During paralysis, after Stx2 injection, multiple glial nuclei were observed surrounding motoneurons by electron microscopy. Also revealed was a lamellipodia-like process physically inhibiting the synaptic connection of motoneurons. Ca2+ imaging of cerebral astrocytic end-feet in Stx2-treated mouse brains suggested that the toxin increased neurotransmitter release from neurons. In this article, we propose that the neuron is a primary target of Stx2, affecting neuronal function and leading to paralysis.


Assuntos
Sistema Nervoso Central/metabolismo , Neurônios/metabolismo , Toxina Shiga II/toxicidade , Triexosilceramidas/metabolismo , Animais , Transporte Biológico , Cálcio/metabolismo , Sistema Nervoso Central/efeitos dos fármacos , Feminino , Imunofluorescência , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Neurônios Motores/ultraestrutura , Neuroglia/citologia , Organismos Livres de Patógenos Específicos , Redução de Peso
17.
Infect Immun ; 75(3): 1229-36, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17220320

RESUMO

The macrophage has previously been implicated in contributing to the renal inflammation associated with hemolytic-uremic syndrome (HUS). However, there is currently no in vivo model detailing the contribution of the renal macrophage to the kidney disease associated with HUS. Therefore, renal macrophage recruitment and inhibition of infiltrating renal macrophages were evaluated in an established HUS mouse model. Macrophage recruitment to the kidney was evident by immunohistochemistry 2 h after administration of purified Stx2 and peaked at 48 h postinjection. Mice administered a combination of Stx2 and lipopolysaccharide (LPS) showed increased macrophage recruitment to the kidney compared to mice treated with Stx2 or LPS alone. Monocyte chemoattractants were induced in the kidney, including monocyte chemoattractant protein 1 (MCP-1/CCL2), macrophage inflammatory protein 1alpha (MIP-1alpha/CCL3), and RANTES (CCL5), in a pattern that was coincident with macrophage infiltration as indicated by immunohistochemistry, protein, and RNA analyses. MCP-1 was the most abundant chemokine, MIP-1alpha was the least abundant, and RANTES levels were intermediate. Mice treated with MCP-1, MIP-1alpha, and RANTES neutralizing antibodies had a significant decrease in Stx2 plus LPS-induced macrophage accumulation in the kidney, indicating that these chemokines are required for macrophage recruitment. Furthermore, mice exposed to these three neutralizing antibodies had decreased fibrin deposition in their kidneys, implying that macrophages contribute to the renal damage associated with HUS.


Assuntos
Movimento Celular/imunologia , Quimiocina CCL2/fisiologia , Quimiocina CCL5/fisiologia , Síndrome Hemolítico-Urêmica/metabolismo , Rim/metabolismo , Rim/patologia , Proteínas Inflamatórias de Macrófagos/fisiologia , Macrófagos/metabolismo , Animais , Quimiocina CCL3 , Quimiocina CCL4 , Modelos Animais de Doenças , Síndrome Hemolítico-Urêmica/imunologia , Rim/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Toxina Shiga II/farmacologia
18.
Am J Pathol ; 170(2): 526-37, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17255321

RESUMO

Neutrophilia is a characteristic of hemolytic uremic syndrome caused by Shiga toxin (Stx2)-producing Escherichia coli. However, the role of neutrophils in the toxin-induced renal injury occurring in enterohemorrhagic E. coli infection remains undefined. We report the trafficking of neutrophils to the kidney of C57BL/6 mice throughout a 72-hour time course after challenge with purified E. coli Stx2 and lipopolysaccharide (LPS). Increased neutrophils were observed in the renal cortex, particularly within the glomeruli where a more than fourfold increase in neutrophils was noted within 2 hours after challenge. Using microarray analysis, an increased number of transcripts for chemoattractants CXCL1/KC (69-fold at 2 hours) and CXCL2/MIP-2 (29-fold at 2 hours) were detected. Ribonuclease protection assays, Northern blotting, enzyme-linked immunosorbent assay, and immunohistochemistry confirmed microarray results, showing that both chemokines were expressed only on the immediate periglomerular epithelium and that these events coincided with neutrophil invasion of glomeruli. Co-administration of Stx2 with LPS enhanced and prolonged the KC and MIP-2 host response (RNA and protein) induced by LPS alone. Immunoneutralization in vivo of CXCL1/KC and CXCL2/MIP-2 abrogated neutrophil migration into glomeruli by 85%. These data define the molecular basis for neutrophil migration into the kidney after exposure to virulence factors of Shiga toxin-producing E. coli O157:H7.


Assuntos
Quimiocinas CXC/biossíntese , Quimiocinas/biossíntese , Infecções por Escherichia coli/metabolismo , Escherichia coli O157 , Nefrite/metabolismo , Neutrófilos/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL1 , Quimiocina CXCL2 , Infecções por Escherichia coli/induzido quimicamente , Infecções por Escherichia coli/patologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/microbiologia , Glomérulos Renais/patologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Nefrite/induzido quimicamente , Nefrite/microbiologia , Nefrite/patologia , Infiltração de Neutrófilos , Toxina Shiga II/toxicidade
19.
J Am Soc Nephrol ; 17(12): 3404-14, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17082244

RESUMO

Hemolytic uremic syndrome (HUS), which is caused by Shiga toxin-producing Escherichia coli infection, is the leading cause of acute renal failure in children. At present, there is no complete small animal model of this disease. This study investigated a mouse model using intraperitoneal co-injection of purified Shiga toxin 2 (Stx2) plus LPS. Through microarray, biochemical, and histologic analysis, it was found to be a valid model of the human disease. Biochemical and microarray analysis of mouse kidneys revealed the Stx2 plus LPS challenge to be distinct from the effects of either agent alone. Microarrays identified differentially expressed genes that were demonstrated previously to play a role in this disease. Blood and serum analysis of these mice showed neutrophilia, thrombocytopenia, red cell hemolysis, and increased serum creatinine and blood urea nitrogen. In addition, histologic analysis and electron microscopy of mouse kidneys demonstrated glomerular fibrin deposition, red cell congestion, microthrombi formation, and glomerular ultrastructural changes. It was established that this C57BL/6 mouse is a complete model of HUS that includes the thrombocytopenia, hemolytic anemia, and renal failure that define the human disease. In addition, a time course of HUS disease progression that will be useful for identification of therapeutic targets and development of new treatments for HUS is described.


Assuntos
Síndrome Hemolítico-Urêmica/etiologia , Rim/patologia , Lipopolissacarídeos/toxicidade , Toxina Shiga II/toxicidade , Anemia Hemolítica/etiologia , Animais , Nitrogênio da Ureia Sanguínea , Creatinina/sangue , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/patologia , Rim/fisiopatologia , Leucócitos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Trombocitopenia/etiologia , Redução de Peso
20.
Infect Immun ; 74(1): 81-7, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16368960

RESUMO

Clostridium difficile is the leading cause of nosocomial bacterial diarrhea. Glutamine and its stable and highly soluble derivative alanyl-glutamine, have been beneficial in models of intestinal injury. In this study, we extend our work on the mechanisms of Clostridium difficile toxin A (TxA)-induced apoptosis in human intestinal epithelial T84 cells and evaluate the effects of glutamine and alanyl-glutamine on TxA-induced apoptosis in vitro and disruption of ileal mucosa in vivo. T84 cells were incubated with TxA (100 ng/ml) in medium with or without glutamine or alanyl-glutamine (3 to 100 mM). Apoptosis was evaluated by DNA fragmentation in vitro and the terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end-labeling method in vivo. Caspase and Bid involvement were investigated by Western blotting. Ligated rabbit ileal loops were used for the evaluation of intestinal secretion, mucosal disruption, and apoptosis. TxA induced caspases 6, 8, and 9 prior to caspase 3 activation in T84 cells and induced Bid cleavage by a caspase-independent mechanism. Glutamine or alanyl-glutamine significantly reduced TxA-induced apoptosis of T84 cells by 47% and inhibited activation of caspase 8. Both glutamine and alanyl-glutamine reduced TxA-induced ileal mucosal disruption and secretion. Altogether, we further delineated the apoptosis-signaling cascade induced by TxA in T84 cells and demonstrated the protective effects of glutamine and alanyl-glutamine. Glutamine and alanyl-glutamine inhibited the apoptosis of T84 cells by preventing caspase 8 activation and reduced TxA-induced intestinal secretion and disruption.


Assuntos
Apoptose/fisiologia , Clostridioides difficile/fisiologia , Dipeptídeos/fisiologia , Enterotoxinas/fisiologia , Glutamina/fisiologia , Animais , Apoptose/efeitos dos fármacos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Toxinas Bacterianas , Caspases/metabolismo , Linhagem Celular , Clostridioides difficile/patogenicidade , Dipeptídeos/farmacologia , Glutamina/farmacologia , Humanos , Íleo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA