Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Cancer Res ; 74(3): 787-96, 2014 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-24351290

RÉSUMÉ

The relevance of cysteine metabolism in cancer has gained considerable interest in recent years, largely focusing on its role in generating the antioxidant glutathione. Through metabolomic profiling using a combination of high-throughput liquid and gas chromatography-based mass spectrometry on a total of 69 patient-derived glioma specimens, this report documents the discovery of a parallel pathway involving cysteine catabolism that results in the accumulation of cysteine sulfinic acid (CSA) in glioblastoma. These studies identified CSA to rank as one of the top metabolites differentiating glioblastoma from low-grade glioma. There was strong intratumoral concordance of CSA levels with expression of its biosynthetic enzyme cysteine dioxygenase 1 (CDO1). Studies designed to determine the biologic consequence of this metabolic pathway identified its capacity to inhibit oxidative phosphorylation in glioblastoma cells, which was determined by decreased cellular respiration, decreased ATP production, and increased mitochondrial membrane potential following pathway activation. CSA-induced attenuation of oxidative phosphorylation was attributed to inhibition of the regulatory enzyme pyruvate dehydrogenase. Studies performed in vivo abrogating the CDO1/CSA axis using a lentiviral-mediated short hairpin RNA approach resulted in significant tumor growth inhibition in a glioblastoma mouse model, supporting the potential for this metabolic pathway to serve as a therapeutic target. Collectively, we identified a novel, targetable metabolic pathway involving cysteine catabolism contributing to the growth of aggressive high-grade gliomas. These findings serve as a framework for future investigations designed to more comprehensively determine the clinical application of this metabolic pathway and its contributory role in tumorigenesis.


Sujet(s)
Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Cystéine/métabolisme , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Voies et réseaux métaboliques , Animaux , Tumeurs du cerveau/génétique , Lignée cellulaire tumorale , Cystéine/analogues et dérivés , Cystéine/pharmacologie , Cysteine dioxygenase/antagonistes et inhibiteurs , Cysteine dioxygenase/génétique , Cysteine dioxygenase/métabolisme , Modèles animaux de maladie humaine , Activation enzymatique/effets des médicaments et des substances chimiques , Expression des gènes , Glioblastome/génétique , Humains , Souris , Souris knockout , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Grading des tumeurs , Complexe du pyruvate déshydrogénase/métabolisme , Charge tumorale/effets des médicaments et des substances chimiques , Charge tumorale/génétique
2.
Cancer Res ; 72(22): 5878-88, 2012 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-23026133

RÉSUMÉ

Although considerable progress has been made toward understanding glioblastoma biology through large-scale genetic and protein expression analyses, little is known about the underlying metabolic alterations promoting their aggressive phenotype. We conducted global metabolomic profiling on patient-derived glioma specimens and identified specific metabolic programs differentiating low- and high-grade tumors, with the metabolic signature of glioblastoma reflecting accelerated anabolic metabolism. When coupled with transcriptional profiles, we identified the metabolic phenotype of the mesenchymal subtype to consist of accumulation of the glycolytic intermediate phosphoenolpyruvate and decreased pyruvate kinase activity. Unbiased hierarchical clustering of metabolomic profiles identified three subclasses, which we term energetic, anabolic, and phospholipid catabolism with prognostic relevance. These studies represent the first global metabolomic profiling of glioma, offering a previously undescribed window into their metabolic heterogeneity, and provide the requisite framework for strategies designed to target metabolism in this rapidly fatal malignancy.


Sujet(s)
Glioblastome/métabolisme , Gliome/métabolisme , Chromatographie gazeuse-spectrométrie de masse , Glioblastome/génétique , Glioblastome/anatomopathologie , Gliome/génétique , Gliome/anatomopathologie , Humains , Mésoderme/métabolisme , Mésoderme/anatomopathologie , Métabolomique , Grading des tumeurs , Phénotype , Phosphoénolpyruvate/métabolisme , Pyruvate kinase/métabolisme , Transduction du signal
3.
FASEB J ; 26(6): 2437-45, 2012 Jun.
Article de Anglais | MEDLINE | ID: mdl-22389438

RÉSUMÉ

Post-translational modification through protein acetylation is emerging as an important mode of cellular regulation. We have previously demonstrated the role that glucose-regulated protein 78 kDa (GRP78) acetylation and subsequent activation of the unfolded protein response (UPR) play in the antitumor activity of class I histone deacetylase (HDAC) inhibitors, which primarily target class I HDACs. In this study, we explored the contributory role these class I HDACs may play in UPR regulation. Binding studies were performed using immunoprecipitation/immunoblotting following dual-transfection with HA-tagged GRP78 and FLAG-tagged HDACs. Subcellular localization was performed using Western blot of fractionated cell lysates and confocal microscopy. Individual HDACs were inhibited using RNA interference. We identified the potential of HDACs 1, 2, and 3 to bind to GRP78. These HDACs colocalized with GRP78 in the endoplasmic reticulum (ER). Inhibition of individual HDACs resulted in GRP78 acetylation and selective activation of the UPR. Although traditionally viewed as nuclear enzymes, we demonstrate that Class I HDACs localize to the ER, bind to GRP78, and selectively activate the UPR, representing a novel mode of UPR regulation and mechanism of action of HDAC inhibitors.


Sujet(s)
Réticulum endoplasmique/métabolisme , Protéines du choc thermique/métabolisme , Histone deacetylases/métabolisme , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Chaperonne BiP du réticulum endoplasmique , Inhibiteurs de désacétylase d'histone , Humains , Interférence par ARN
4.
Neuro Oncol ; 14(1): 93-100, 2012 Jan.
Article de Anglais | MEDLINE | ID: mdl-22028388

RÉSUMÉ

A phase I study was conducted to determine the dose-limiting toxicities (DLT) and maximum tolerated dose (MTD) for the combination of vorinostat with bevacizumab and CPT-11 in recurrent glioblastoma. Vorinostat was combined with bevacizumab and CPT-11 and was escalated using a standard 3 + 3 design. Vorinostat was escalated up to 2 actively investigated doses of this compound or until the MTD was identified on the basis of DLTs. Correlative science involving proteomic profiling of serial patient plasma samples was performed. Nineteen patients were treated. The MTD of vorinostat was established at 400 mg on days 1-7 and 15-21 every 28 days when combined with bevacizumab and CPT-11. Common toxicities were fatigue and diarrhea. DLTs included fatigue, hypertension/hypotension, and central nervous system ischemia. Although the MTD was established, CPT-11 dose reductions were common early in therapy. High-dose vorinostat had an improved progression-free survival and overall survival when compared with low-dose vorinostat. Serum proteomic profiling identified IGFBP-5 and PDGF-AA as markers for improved PFS and recurrence, respectively. A MTD for the combination of vorinostat with bevacizumab and CPT-11 has been established, although it has poor long-term tolerability. With the increased toxicities associated with CPT-11 coupled with its unclear clinical significance, investigating the efficacy of vorinostat combined with bevacizumab alone may represent a more promising strategy to evaluate in the context of a phase II clinical trial.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Glioblastome/traitement médicamenteux , Adulte , Sujet âgé , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/toxicité , Protocoles de polychimiothérapie antinéoplasique/toxicité , Bévacizumab , Camptothécine/administration et posologie , Camptothécine/analogues et dérivés , Camptothécine/toxicité , Survie sans rechute , Femelle , Humains , Acides hydroxamiques/administration et posologie , Acides hydroxamiques/toxicité , Protéine-5 de liaison aux IGF/analyse , Irinotécan , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Récidive tumorale locale/traitement médicamenteux , Facteur de croissance dérivé des plaquettes/analyse , Protéomique , Vorinostat
5.
PLoS One ; 7(12): e52265, 2012.
Article de Anglais | MEDLINE | ID: mdl-23284962

RÉSUMÉ

Rapidly growing tumors require efficient means to allow them to adapt to fluctuating microenvironments consisting of hypoxia, nutrient deprivation, and acidosis. The unfolded protein response (UPR) represents a defense mechanism allowing cells to respond to these adverse conditions. The chaperone protein GRP78 serves as a master UPR regulator that is aberrantly expressed in a variety of cancers, including glioma. Therefore, cancer cells may be particularly reliant upon the adaptive mechanisms offered by the UPR and targeting GRP78 may represent a unique therapeutic strategy. Here we report that diffuse expression of GRP78 protein is present in Grade III-IV, but not Grade I-II glioma. To determine the role GRP78 plays in glioblastoma tumorigenesis, we explored the anti-tumor activity of the novel fusion protein EGF-SubA, which combines EGF with the cytotoxin SubA that has been recently shown to selectively cleave GRP78. EGF-SubA demonstrated potent tumor-specific proteolytic activity and cytotoxicity in glioblastoma lines and potentiated the anti-tumor activity of both temozolomide and ionizing radiation. To determine if the tumor microenvironment influences EGF-SubA activity, we maintained cells in acidic conditions that led to both UPR activation and increased EGF-SubA induced cytotoxicity. EGF-SubA was well tolerated in mice and led to a significant tumor growth delay in a glioma xenograft mouse model. The UPR is emerging as an important adaptive pathway contributing to glioma tumorigenesis. Targeting its primary mediator, the chaperone protein GRP78, through specific, proteolytic cleavage with the immunotoxin EGF-SubA represents a novel and promising multi-targeted approach to cancer therapy.


Sujet(s)
Facteur de croissance épidermique/pharmacologie , Glioblastome/métabolisme , Subtilisines/métabolisme , Subtilisines/pharmacologie , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire tumorale , Chaperonne BiP du réticulum endoplasmique , Facteur de croissance épidermique/génétique , Facteur de croissance épidermique/métabolisme , Glioblastome/génétique , Protéines du choc thermique/génétique , Protéines du choc thermique/métabolisme , Humains , Immunotransfert , Souris , RT-PCR , Subtilisines/génétique , Analyse sur puce à tissus , Réponse aux protéines mal repliées/génétique
6.
Mol Cancer Ther ; 10(12): 2405-14, 2011 Dec.
Article de Anglais | MEDLINE | ID: mdl-21992793

RÉSUMÉ

The purpose of this study was to determine the capacity of MK-1775, a potent Wee-1 inhibitor, to abrogate the radiation-induced G(2) checkpoint arrest and modulate radiosensitivity in glioblastoma cell models and normal human astrocytes. The radiation-induced checkpoint response of established glioblastoma cell lines, glioblastoma neural stem (GNS) cells, and astrocytes were determined in vitro by flow cytometry and in vivo by mitosis-specific staining using immunohistochemistry. Mechanisms underlying MK-1775 radiosensitization were determined by mitotic catastrophe and γH2AX expression. Radiosensitivity was determined in vitro by the clonogenic assay and in vivo by tumor growth delay. MK-1775 abrogated the radiation-induced G(2) checkpoint and enhanced radiosensitivity in established glioblastoma cell lines in vitro and in vivo, without modulating radiation response in normal human astrocytes. MK-1775 appeared to attenuate the early-phase of the G(2) checkpoint arrest in GNS cell lines, although the arrest was not sustained and did not lead to increased radiosensitivity. These results show that MK-1775 can selectively enhance radiosensitivity in established glioblastoma cell lines. Further work is required to determine the role Wee-1 plays in checkpoint activation of GNS cells.


Sujet(s)
Tumeurs du cerveau/traitement médicamenteux , Points de contrôle de la phase G2 du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle de la phase G2 du cycle cellulaire/effets des radiations , Glioblastome/traitement médicamenteux , Pyrazoles/usage thérapeutique , Pyrimidines/usage thérapeutique , Animaux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Humains , Souris , Souris nude , Thérapie moléculaire ciblée , Protéines nucléaires/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacocinétique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protein-tyrosine kinases/antagonistes et inhibiteurs , Pyrazoles/pharmacocinétique , Pyrazoles/pharmacologie , Pyrimidines/pharmacocinétique , Pyrimidines/pharmacologie , Pyrimidinones , Radiotolérance/effets des médicaments et des substances chimiques , Radiosensibilisants/pharmacocinétique , Radiosensibilisants/pharmacologie , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/effets des radiations , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Methods Enzymol ; 490: 159-74, 2011.
Article de Anglais | MEDLINE | ID: mdl-21266250

RÉSUMÉ

Although the function of histone deacetylases (HDACs) have primarily been associated with influencing transcription through chromatin remodeling, the capacity of these enzymes to interface with a diverse array of biologic processes by modulating a growing list of nonhistone substrates has gained recent attention. Recent investigations have demonstrated the potential of HDACs to directly regulate the unfolded protein response (UPR) through acetylation of its central regulatory protein, Grp78. Further, this appears to be an important mechanism underlying the anti-tumor activity of HDAC inhibitors. Herein, we provide a summary of the literature supporting the role HDACs play in regulating the UPR and a detailed description of methods to allow for the study of both acetylation of nonhistone proteins and UPR pathway activation following HDAC inhibition.


Sujet(s)
Histone deacetylases/métabolisme , Réponse aux protéines mal repliées/physiologie , Acétylation , Animaux , Mort cellulaire , Lignée cellulaire , Chaperonne BiP du réticulum endoplasmique , Protéines du choc thermique/génétique , Protéines du choc thermique/métabolisme , Histone acetyltransferases/génétique , Histone acetyltransferases/métabolisme , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Histone deacetylases/génétique , Humains , Tumeurs/traitement médicamenteux , Tumeurs/enzymologie , Tumeurs/génétique , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Transduction du signal/physiologie , eIF-2 Kinase/métabolisme
8.
J Neurooncol ; 99(2): 201-7, 2010 Sep.
Article de Anglais | MEDLINE | ID: mdl-20135194

RÉSUMÉ

Histone deacetylase (HDAC) inhibitors represent a promising class of anti-cancer agents that are actively being evaluated in the context of clinical trials in solid tumors, including glioblastoma. What makes these agents particularly attractive is their capacity to enhance the activity of commonly used cytotoxics in cancer therapy, including both chemotherapy and ionizing radiation. As recent investigations suggest HDAC inhibitors may potentiate the cytotoxicity of topoisomerase inhibitors, which continue to be a commonly used class of agents in the treatment of glioblastoma, we performed preclinical studies to determine if this combination may be a promising strategy in glioblastoma. The effects of the HDAC inhibitor vorinostat and SN38, which is the active metabolite of the topoisomerase I inhibitor CPT-11, was evaluated using the clonogenic assay. Various treatment schedules were tested to determine optimum drug sequencing. Induction of DNA double strand breaks (DSBs) with the combination of vorinostat and SN38 was evaluated using the neutral comet assay, and their subsequent repair was evaluated by gammaH2AX foci kinetics using immunofluorescent cytochemistry. Vorinostat enhanced the cytotoxicity of SN38 in glioblastoma cell lines. Optimal treatment schedules involved maximal concurrent administration of agents. Pretreatment with either agent did not enhance cytotoxicity. Vorinostat potentiated SN38-induced DNA DSBs and attenuated their subsequent repair. These results indicate vorinostat enhances the cytotoxicity of SN38 in glioblastoma cell lines, suggesting this combination may be a worthwhile strategy to test in the context of a clinical trial.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Tumeurs du cerveau/traitement médicamenteux , Altération de l'ADN/effets des médicaments et des substances chimiques , Réparation de l'ADN/effets des médicaments et des substances chimiques , Glioblastome/traitement médicamenteux , Technique de Western , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Camptothécine/administration et posologie , Camptothécine/analogues et dérivés , Prolifération cellulaire/effets des médicaments et des substances chimiques , Test des comètes , ADN topoisomérases de type I/composition chimique , Synergie des médicaments , Technique d'immunofluorescence , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Humains , Acides hydroxamiques/administration et posologie , Irinotécan , Cellules cancéreuses en culture , Test clonogénique de cellules souches tumorales , Vorinostat
9.
Neoplasia ; 12(1): 80-6, 2010 Jan.
Article de Anglais | MEDLINE | ID: mdl-20072656

RÉSUMÉ

Histone deacetylase (HDAC) inhibitors represent an emerging class of anticancer agents progressing through clinical trials. Although their primary target is thought to involve acetylation of core histones, several nonhistone substrates have been identified, including heat shock protein (HSP) 90, which may contribute towards their antitumor activity. Glucose-regulated protein 78 (GRP78) is a member of the HSP family of molecular chaperones and plays a central role in regulating the unfolded protein response (UPR). Emerging data suggest that GRP78 is critical in cellular adaptation and survival associated with oncogenesis and may serve as a cancer-specific therapeutic target. On the basis of shared homology with HSP family proteins, we sought to determine whether GRP78 could serve as a molecular target of the HDAC inhibitor vorinostat. Vorinostat treatment led to GRP78 acetylation, dissociation, and subsequent activation of its client protein double-stranded RNA-activated protein-like endoplasmic reticulum kinase (PERK). Investigations in a panel of cancer cell lines identified that UPR activation after vorinostat exposure is specific to certain lines. Mass spectrometry performed on immunoprecipitated GRP78 identified lysine-585 as a specific vorinostat-induced acetylation site of GRP78. Downstream activation of the UPR was confirmed, including eukaryotic initiating factor 2alpha phosphorylation and increase in ATF4 and C/EBP homologous protein expression. To determine the biologic relevance of UPR activation after vorinostat, RNA interference of PERK was performed, demonstrating significantly decreased sensitivity to vorinostat-induced cytotoxicity. Collectively, these findings indicate that GRP78 is a biologic target of vorinostat, and activation of the UPR through PERK phosphorylation contributes toward its antitumor activity.


Sujet(s)
Antinéoplasiques/pharmacologie , Histone acetyltransferases/antagonistes et inhibiteurs , Acides hydroxamiques/pharmacologie , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Acétylation/effets des médicaments et des substances chimiques , Technique de Western , Lignée cellulaire tumorale , Chaperonne BiP du réticulum endoplasmique , Antienzymes/pharmacologie , Protéines du choc thermique/métabolisme , Histone acetyltransferases/métabolisme , Humains , Lysine/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Interférence par ARN , Vorinostat , eIF-2 Kinase/génétique , eIF-2 Kinase/métabolisme
10.
Cell Cycle ; 5(15): 1642-8, 2006 Aug.
Article de Anglais | MEDLINE | ID: mdl-16861897

RÉSUMÉ

Azurin, a member of a family of copper-containing proteins involved in electron transfer called cupredoxins, demonstrates structural features similar to the variable domains of the immunoglobulin superfamily members. An azurin-like protein called Laz with an additional N-terminal 39 amino acid peptide known as H.8 epitope is present on the surface of gonnococci and meningococci. We demonstrate that azurin, Laz and H.8-azurin can bind to the C-terminal cleavage product MSP1-19 of merozoite surface protein 1 (MSP1) of the malarial parasite Plasmodium falciparum and significantly reduce parasitemia. Azurin and Laz also bound strongly to HIV-1 gp120. Interestingly, azurin could not only bind to gp120 but also to the dendritic cell-specific adhesion receptor DC-SIGN, mimicking the functionality of the intercellular adhesion molecule ICAM-3 with which it also binds avidly. Furthermore, these three proteins significantly suppressed HIV-1 growth in peripheral blood mononuclear cells and such suppression appeared to be occurring at an entry stage in the infection process. The presence of both antimalarial and antiretroviral activity in azurin, H.8-azurin and Laz makes these proteins, or peptides derived from them, potential therapeutic agents in the treatment of malaria, HIV-1 infections or coinfections with both P. falciparum and HIV-1.


Sujet(s)
Syndrome d'immunodéficience acquise/virologie , Azurine/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/croissance et développement , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Paludisme à Plasmodium falciparum/parasitologie , Plasmodium falciparum/croissance et développement , Plasmodium falciparum/physiologie , Animaux , Antigènes bactériens/immunologie , Protéines de la membrane externe bactérienne/métabolisme , Molécules d'adhérence cellulaire/métabolisme , Érythrocytes/parasitologie , Protéine d'enveloppe gp120 du VIH/métabolisme , Humains , Immunoglobulines/composition chimique , Immunoglobulines/immunologie , Lectines de type C/métabolisme , Protéine-1 de surface du mérozoïte/métabolisme , Neisseria/composition chimique , Neisseria/immunologie , Parasitémie , Liaison aux protéines , Pseudomonas aeruginosa/composition chimique , Pseudomonas aeruginosa/immunologie , Récepteurs de surface cellulaire/métabolisme , Résonance plasmonique de surface
11.
J Clin Microbiol ; 42(9): 4111-20, 2004 Sep.
Article de Anglais | MEDLINE | ID: mdl-15364997

RÉSUMÉ

Enteroaggregative Escherichia coli (EAEC) is an important diarrheal enteropathogen defined by aggregative adherence to cultured epithelial cells. We have detected EAEC from 121 (6.6%) of 1,826 hospitalized patients admitted with diarrhea to the Infectious Diseases Hospital in Kolkata, India. Watery diarrhea was recorded significantly (P = 0.0142) more often in children. The majority of the EAEC isolates were not serotypeable (62%) and showed resistance to five or more antibiotics (76%). We studied different virulence genes and the molecular epidemiology of 121 EAEC isolates recovered from diarrheal patients. A PCR assay for detection of virulence genes, an assay for determination of clump formation in liquid culture, and a HeLa cell adherence assay were carried out to characterize the EAEC isolates. Investigations were also conducted to correlate the virulence gene profiles with diarrheal symptoms and molecular epidemiology by pulsed-field gel electrophoresis (PFGE). Two or more virulence genes were detected in 109 (90.1%) EAEC isolates. In the cluster analysis, some isolates with specific gene profiles and phenotypes formed a group or subcluster. This study highlights the comparative distributions of three fimbrial adhesins and other virulence genes among EAEC isolates. The diverse virulence gene and PFGE profiles, along with the existence of diverse serotypes and antibiograms, suggests that the EAEC isolates are genetically heterogeneous in Kolkata.


Sujet(s)
Diarrhée/microbiologie , Infections à Escherichia coli/microbiologie , Escherichia coli/isolement et purification , Escherichia coli/pathogénicité , Virulence , Séquence nucléotidique , Amorces ADN , Diarrhée/épidémiologie , Escherichia coli/classification , Infections à Escherichia coli/épidémiologie , Humains , Inde/épidémiologie , Patients hospitalisés , Tests de sensibilité microbienne , Épidémiologie moléculaire , Données de séquences moléculaires , Phylogenèse
12.
Eur J Epidemiol ; 19(5): 473-9, 2004.
Article de Anglais | MEDLINE | ID: mdl-15233322

RÉSUMÉ

Diarrhoeal infection caused by Escherichia coli is common in India with occasional outbreaks. However, association of different pathotypes of diarrhoeagenic E. coli (DEC) with the disease and its phenotypic and genotypic characteristics are not fully demonstrated. In this study, E. coli strains from sporadic cases and outbreaks of diarrhoea during 2000-2001 were confirmed as DEC by polymerase chain reaction (PCR) targeting the specific virulence genes. DEC represented by enterotoxigenic E. coli (ETEC), enteropathogenic E. coli (EPEC) and enteroaggregative E. coli (EAggEC) were mostly belonged to O serogroups 25, 86a, 114 and 146. The gene astA was frequently detected among ETEC and EAggEC than EPEC. After initial screening of 200 DEC strains with serology and antibiotic susceptibility test, 32 strains representing ETEC, EPEC, and EAggEC isolated from different areas of India were included in the pulsed-field gel electrophoresis (PFGE) analysis. Using the PFGE results, the hierarchical representation of different linkage levels between the DEC strains were determined by unweighed pair-group arithmetic mean (UPGAMA) method. Except for few strains, clonotyping by PFGE revealed no correlation between pathotypes and serogroups as well as the place of isolation of the DEC strains. The prevailing clonal diversity among the different categories of DEC strains suggests that the pathotypes of DEC belonged to diverse clones.


Sujet(s)
Diarrhée/microbiologie , Infections à Escherichia coli/microbiologie , Escherichia coli/génétique , Gènes bactériens , Analyse de regroupements , Diarrhée/épidémiologie , Épidémies de maladies , Résistance bactérienne aux médicaments , Électrophorèse en champ pulsé , Escherichia coli/effets des médicaments et des substances chimiques , Escherichia coli/pathogénicité , Infections à Escherichia coli/traitement médicamenteux , Infections à Escherichia coli/épidémiologie , Fèces/microbiologie , Humains , Inde/épidémiologie , Épidémiologie moléculaire , Réaction de polymérisation en chaîne , Virulence/génétique
13.
J Clin Microbiol ; 41(11): 5277-81, 2003 Nov.
Article de Anglais | MEDLINE | ID: mdl-14605183

RÉSUMÉ

Among Escherichia coli strains isolated from stool specimens from patients with acute diarrhea, 1.4% were found to harbor cdtB by use of enrichment cytolethal distending toxin (CDT) PCR. These isolates were identified as being enteropathogenic E. coli (EPEC). In a retrospective study using a probe hybridization assay, 6 of 138 EPEC strains were found to harbor the cdtB locus. cdtB-positive isolates mostly belong to the O86a and O127a serogroups, with the former being associated with higher expression of CDT. Pulsed-field gel electrophoresis profiles showed that the EPEC strains harboring cdtB strains are genetically diverse.


Sujet(s)
Toxines bactériennes/génétique , Diarrhée/microbiologie , Infections à Escherichia coli/microbiologie , Protéines Escherichia coli/génétique , Escherichia coli/pathogénicité , Fèces/microbiologie , Maladie aigüe , Toxines bactériennes/classification , Toxines bactériennes/isolement et purification , Séquence nucléotidique , Sang/microbiologie , Amorces ADN , Déshydratation/microbiologie , Électrophorèse en champ pulsé , Escherichia coli/classification , Escherichia coli/génétique , Escherichia coli/isolement et purification , Infections à Escherichia coli/sang , Protéines Escherichia coli/isolement et purification , Humains , Inde , Données de séquences moléculaires , Sang occulte , Phylogenèse , Réaction de polymérisation en chaîne , Sérotypie/méthodes , Vomissement/microbiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE