Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
mBio ; 12(6): e0283321, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34724811

ABSTRACT

Shigella spp. are highly adapted pathogens that cause bacillary dysentery in human and nonhuman primates. An unusual feature of Shigella pathogenesis is that this organism invades the colonic epithelia from the basolateral pole. Therefore, it has evolved the ability to disrupt the intestinal epithelial barrier to reach the basolateral surface. We have shown previously that the secreted serine protease A (SepA), which belongs to the family of serine protease autotransporters of Enterobacteriaceae, is responsible for the initial destabilization of the intestinal epithelial barrier that facilitates Shigella invasion. However, the mechanisms used by SepA to regulate this process remain unknown. To investigate the protein targets cleaved by SepA in the intestinal epithelium, we incubated a sample of homogenized human colon with purified SepA or with a catalytically inactive mutant of this protease. We discovered that SepA targets an array of 18 different proteins, including alpha-1 antitrypsin (AAT), a major circulating serine proteinase inhibitor in humans. In contrast to other serine proteases, SepA cleaved AAT without forming an inhibiting complex, which resulted in the generation of a neutrophil chemoattractant. We demonstrated that the products of the AAT-SepA reaction induce a mild but significant increase in neutrophil transepithelial migration in vitro. Moreover, the presence of AAT during Shigella infection stimulated neutrophil migration and dramatically enhanced the number of bacteria invading the intestinal epithelium in a SepA-dependent manner. We conclude that by cleaving AAT, SepA releases a chemoattractant that promotes neutrophil migration, which in turn disrupts the intestinal epithelial barrier to enable Shigella invasion. IMPORTANCEShigella is the second leading cause of diarrheal death globally. In this study, we identified the host protein targets of SepA, Shigella's major protein secreted in culture. We demonstrated that by cleaving AAT, a serine protease inhibitor important to protect surrounding tissue at inflammatory sites, SepA releases a neutrophil chemoattractant that enhances Shigella invasion. Moreover, SepA degraded AAT without becoming inhibited by the cleaved product, and SepA catalytic activity was enhanced at higher concentrations of AAT. Activation of SepA by an excess of AAT may be physiologically relevant at the early stages of Shigella infection, when the amount of synthesized SepA is very low compared to the concentration of AAT in the intestinal lumen. This observation may also help to explain the adeptness of Shigella infectivity at low dose, despite the requirement of reaching the basolateral side to invade and colonize the colonic epithelium.


Subject(s)
Bacterial Proteins/metabolism , Chemotactic Factors/metabolism , Dysentery, Bacillary/metabolism , Epithelial Cells/microbiology , Neutrophils/cytology , Shigella/enzymology , alpha 1-Antitrypsin/metabolism , Bacterial Proteins/genetics , Cell Movement , Chemotactic Factors/genetics , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/physiopathology , Epithelial Cells/metabolism , Humans , Intestines/cytology , Intestines/metabolism , Intestines/microbiology , Neutrophils/metabolism , Shigella/classification , Shigella/genetics , alpha 1-Antitrypsin/genetics
2.
Braz J Microbiol ; 52(4): 1745-1753, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34251610

ABSTRACT

Historically, clinical microbiological laboratories have often relied on isolation of pure cultures and phenotypic testing to identify microorganisms. These clinical tests are often based on specific biochemical reactions, growth characteristics, colony morphology, and other physiological aspects. The features used for identification in clinical laboratories are highly conserved and specific for a given group of microbes. We speculate that these features might be the result of evolutionary selection and thus may reflect aspects of the life cycle of the organism and pathogenesis. Indeed, several of the metabolic pathways targeted by diagnostic tests in some cases may represent mechanisms for host colonization or pathogenesis. Examples include, but are not restricted to, Staphylococcus aureus, Pseudomonas aeruginosa, Klebsiella pneumoniae, Salmonella enterica, Shigella spp., and enteroinvasive Escherichia coli (EIEC). Here, we provide an overview of how some common tests reflect molecular mechanisms of bacterial pathogenesis.


Subject(s)
Bacterial Infections , Dysentery, Bacillary , Host Adaptation , Bacteria/immunology , Bacterial Infections/diagnosis , Bacterial Infections/microbiology , Bacterial Infections/pathology , Dysentery, Bacillary/diagnosis , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/physiopathology , Humans , Laboratories, Clinical
3.
Infect Genet Evol ; 85: 104532, 2020 11.
Article in English | MEDLINE | ID: mdl-32911076

ABSTRACT

Shigella is an intracellular bacterial pathogen that causes bacterial dysentery called shigellosis. The assessment of pro- and anti-inflammatory mediators produced by immune cells against this bacteria are vital in identifying the effectiveness of the immune reaction in protecting the host. In Malaysia, Shigella is ranked as the third most common bacteria causing diarrheal disease among children below 5 years old. In the present study, we aim to examine the differential cytokine gene expressions of macrophages in response to two types of clinical strains of Shigella flexneri 2a (S. flexneri 2a) isolated from patients admitted in Hospital Universiti Sains Malaysia, Kelantan, Malaysia. THP-1-derived macrophages, as the model of human macrophages, were infected separately with S. flexneri 2a mild (SH062) and virulence (SH057) strains for 6, 12, and 24 h, respectively. The gene expression level of inflammatory mediators was identified using real-time quantitative polymerase chain reaction (RT-qPCR). The production of nitric oxide (NO) by the macrophages was measured by using a commercialized NO assay kit. The ability of macrophages to kill the intracellular bacteria was assessed by intracellular killing assay. Induction of tumor necrosis factor-alpha (TNFα), interleukin (IL)-1ß, IL-6, IL-12, inducible NO synthase (iNOS), and NO, confirmed the pro-inflammatory reaction of the THP-1-derived macrophages in response to S. flexneri 2a, especially against the SH507 strain. The SH057 also induced a marked increase in the expression levels of the anti-inflammatory cytokine mRNAs at 12 h and 24 h post-infection. In the intracellular killing assay, both strains showed less viable, indicating the generation of pro-inflammatory cytokines in the presence of iNOS and NO was crucial in the stimulation of macrophages for the host defense against shigellosis. Transcription analysis of THP-1-derived macrophages in this study identifies differentially expressed cytokine genes that correlated with the virulence factor of S. flexneri 2a.


Subject(s)
Cytokines/genetics , Cytokines/metabolism , Dysentery, Bacillary/genetics , Dysentery, Bacillary/physiopathology , Macrophages/microbiology , Shigella flexneri/genetics , Virulence Factors/genetics , Virulence/genetics , Animals , Child, Preschool , Disease Models, Animal , Dysentery, Bacillary/epidemiology , Female , Gene Expression Regulation, Bacterial , Genes, Bacterial , Genetic Variation , Genotype , Humans , Infant , Infant, Newborn , Malaysia/epidemiology , Male , Shigella flexneri/pathogenicity
4.
Lancet Glob Health ; 8(1): e101-e112, 2020 01.
Article in English | MEDLINE | ID: mdl-31734154

ABSTRACT

BACKGROUND: Diarrhoea, a global cause of child mortality and morbidity, is linked to adverse consequences including childhood stunting and death from other diseases. Few studies explore how diarrhoeal mortality varies subnationally, especially by cause, which is important for targeting investments. Even fewer examine indirect effects of diarrhoeal morbidity on child mortality. We estimated the subnational distribution of mortality, morbidity, and childhood stunting attributable to enterotoxigenic Escherichia coli (ETEC) and shigella infection in children younger than 5 years from 11 eastern and central African countries. These pathogens are leading causes of diarrhoea in young children and have been linked to increased childhood stunting. METHODS: We combined proxy indicators of morbidity and mortality risk from the most recent Demographic and Health Surveys with published relative risks to estimate the potential distribution of diarrhoeal disease risk. To estimate subnational burden, we used country-specific or WHO region-specific morbidity and mortality estimates and distributed them subnationally by three indices that integrate relevant individual characteristics (ie, underweight, probability of receiving oral rehydration treatment of diarrhoea, and receiving vitamin A supplementation) and household characteristics (ie, type of drinking water and sanitation facilities). FINDINGS: Characterising ETEC and shigella subnational estimates of indirect morbidity (infection-attributable stunting) and indirect mortality (stunting-related deaths from other infectious diseases) identified high-risk areas that could be missed by traditional metrics. Burundi and Democratic Republic of the Congo had the highest ETEC-associated and shigella-associated mortality and stunting rates. Mozambique, Democratic Republic of the Congo, and Zimbabwe had the greatest subnational heterogeneity in most ETEC and shigella mortality measures. Inclusion of indirect ETEC and shigella mortality in burden estimates resulted in a 20-30% increase in total ETEC and shigella mortality rates in some subnational areas. INTERPRETATION: Understanding the indirect mortality and morbidity of diarrhoeal pathogens on a subnational level will strengthen disease control strategies and could have important implications for the relative impact and cost-effectiveness of new enteric vaccines. Because our methods rely on publicly available data, they could be employed for national planning. FUNDING: Bill & Melinda Gates Foundation.


Subject(s)
Dysentery, Bacillary/epidemiology , Dysentery, Bacillary/mortality , Escherichia coli Infections/epidemiology , Escherichia coli Infections/mortality , Growth Disorders/epidemiology , Risk Assessment/statistics & numerical data , Africa/epidemiology , Cause of Death , Child, Preschool , Dysentery, Bacillary/physiopathology , Escherichia coli Infections/physiopathology , Female , Growth Disorders/physiopathology , Humans , Infant , Infant, Newborn , Male , Mortality
5.
Proc Natl Acad Sci U S A ; 116(27): 13582-13591, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31209035

ABSTRACT

Intracellular trafficking pathways in eukaryotic cells are essential to maintain organelle identity and structure, and to regulate cell communication with its environment. Shigella flexneri invades and subverts the human colonic epithelium by the injection of virulence factors through a type 3 secretion system (T3SS). In this work, we report the multiple effects of two S. flexneri effectors, IpaJ and VirA, which target small GTPases of the Arf and Rab families, consequently inhibiting several intracellular trafficking pathways. IpaJ and VirA induce large-scale impairment of host protein secretion and block the recycling of surface receptors. Moreover, these two effectors decrease clathrin-dependent and -independent endocytosis. Therefore, S. flexneri infection induces a global blockage of host cell intracellular transport, affecting the exchange between cells and their external environment. The combined action of these effectors disorganizes the epithelial cell polarity, disturbs epithelial barrier integrity, promotes multiple invasion events, and enhances the pathogen capacity to penetrate into the colonic tissue in vivo.


Subject(s)
Dysentery, Bacillary/physiopathology , Intestinal Mucosa/microbiology , Shigella flexneri , Biological Transport , Caco-2 Cells , Cell Polarity , Colon/metabolism , Colon/microbiology , Colon/pathology , Colon/physiopathology , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/pathology , Endocytosis , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology
6.
Vaccine ; 37(34): 4814-4822, 2019 08 07.
Article in English | MEDLINE | ID: mdl-30981626

ABSTRACT

Well-established, validated and clinically meaningful primary and secondary endpoints are critical in advancing vaccines through proof of principal studies, licensure and pre-qualification. To that end, the field of vaccine development for Shigella, enterotoxigenic Escherichia coli (ETEC) as well as other enteric pathogens would benefit greatly from a focused review of clinical endpoints and the use of common endpoints across the field to enable study-to-study comparisons as well as comparative assessments between vaccine candidates. A workshop was conducted to review clinical endpoints from controlled human challenge studies, field studies in naïve adult travelers and pediatric studies in low-middle income countries and to develop a consensus on clinical endpoints for future vaccine trials. Following sequential presentations on different study designs (CHIM, travelers' efficacy and pediatric efficacy), workshop participants broke into three simultaneous workgroups focused on those study designs to discuss a number of topics key to clinical endpoints specific to each study design. Previously utilized endpoints were reviewed with an eye towards potentially novel endpoints for future studies and consideration of the disease parameters and spectrum of disease targeted for prevention. The strength of support among workshop participants for the use of various endpoints is summarized as are recommendations for additional endpoints to be considered in future studies. It is anticipated that this report will facilitate endpoint determination in future efficacy trials of vaccine candidates.


Subject(s)
Diarrhea/prevention & control , Dysentery, Bacillary/prevention & control , Endpoint Determination/methods , Escherichia coli Infections/prevention & control , Escherichia coli Vaccines/administration & dosage , Shigella Vaccines/administration & dosage , Adult , Child, Preschool , Clinical Trials as Topic , Congresses as Topic , Developing Countries , Diarrhea/immunology , Diarrhea/physiopathology , Dysentery, Bacillary/immunology , Dysentery, Bacillary/physiopathology , Enterotoxigenic Escherichia coli/drug effects , Enterotoxigenic Escherichia coli/immunology , Enterotoxigenic Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Infections/physiopathology , Escherichia coli Vaccines/biosynthesis , Humans , Immunization , Immunogenicity, Vaccine , Models, Immunological , Shigella/drug effects , Shigella/immunology , Shigella/pathogenicity , Shigella Vaccines/biosynthesis , Travel
7.
mBio ; 9(3)2018 06 26.
Article in English | MEDLINE | ID: mdl-29946048

ABSTRACT

Emergency granulopoiesis is a hematopoietic program of stem cell-driven neutrophil production used to counteract immune cell exhaustion following infection. Shigella flexneri is a Gram-negative enteroinvasive pathogen controlled by neutrophils. In this study, we use a Shigella-zebrafish (Danio rerio) infection model to investigate emergency granulopoiesis in vivo We show that stem cell-driven neutrophil production occurs in response to Shigella infection and requires macrophage-independent signaling by granulocyte colony-stimulating factor (Gcsf). To test whether emergency granulopoiesis can function beyond homoeostasis to enhance innate immunity, we developed a reinfection assay using zebrafish larvae that have not yet developed an adaptive immune system. Strikingly, larvae primed with a sublethal dose of Shigella are protected against a secondary lethal dose of Shigella in a type III secretion system (T3SS)-dependent manner. Collectively, these results highlight a new role for emergency granulopoiesis in boosting host defense and demonstrate that zebrafish larvae can be a valuable in vivo model to investigate innate immune memory.IMPORTANCEShigella is an important human pathogen of the gut. Emergency granulopoiesis is the enhanced production of neutrophils by hematopoietic stem and progenitor cells (HSPCs) upon infection and is widely considered a homoeostatic mechanism for replacing exhausted leukocytes. In this study, we developed a Shigella-zebrafish infection model to investigate stem cell-driven emergency granulopoiesis. We discovered that zebrafish initiate granulopoiesis in response to Shigella infection, via macrophage-independent signaling of granulocyte colony-stimulating factor (Gcsf). Strikingly, larvae primed with a sublethal dose of Shigella are protected against a secondary lethal dose of Shigella in a type III secretion system (T3SS)-dependent manner. Taken together, we show that zebrafish infection can be used to capture Shigella-mediated stem cell-driven granulopoiesis and provide a new model system to study stem cell biology in vivo Our results also highlight the potential of manipulating stem cell-driven granulopoiesis to boost innate immunity and combat infectious disease.


Subject(s)
Coinfection/immunology , Disease Models, Animal , Dysentery, Bacillary/microbiology , Leukopoiesis , Neutrophils/immunology , Shigella flexneri/physiology , Animals , Coinfection/microbiology , Coinfection/physiopathology , Dysentery, Bacillary/immunology , Dysentery, Bacillary/physiopathology , Female , Humans , Larva/immunology , Larva/microbiology , Macrophages/immunology , Male , Neutrophils/cytology , Zebrafish/immunology , Zebrafish/microbiology
8.
Infect Immun ; 86(8)2018 08.
Article in English | MEDLINE | ID: mdl-29844234

ABSTRACT

Shigella flexneri disseminates within the colonic mucosa by displaying actin-based motility in the cytosol of epithelial cells. Motile bacteria form membrane protrusions that project into adjacent cells and resolve into double-membrane vacuoles (DMVs) from which the bacteria escape, thereby achieving cell-to-cell spread. During dissemination, S. flexneri is targeted by LC3-dependent autophagy, a host cell defense mechanism against intracellular pathogens. The S. flexneri type III secretion system effector protein IcsB was initially proposed to counteract the recruitment of the LC3-dependent autophagy machinery to cytosolic bacteria. However, a recent study proposed that LC3 was recruited to bacteria in DMVs formed during cell-to-cell spread. To resolve the controversy and clarify the role of autophagy in S. flexneri infection, we tracked dissemination using live confocal microscopy and determined the spatial and temporal recruitment of LC3 to bacteria. This approach demonstrated that (i) LC3 was exclusively recruited to wild-type or icsB bacteria located in DMVs and (ii) the icsB mutant was defective in cell-to-cell spread due to failure to escape LC3-positive as well as LC3-negative DMVs. Failure of S. flexneri to escape DMVs correlated with late LC3 recruitment, suggesting that LC3 recruitment is the consequence and not the cause of DMV escape failure. Inhibition of autophagy had no positive impact on the spreading of wild-type or icsB mutant bacteria. Our results unambiguously demonstrate that IcsB is required for DMV escape during cell-to-cell spread, regardless of LC3 recruitment, and do not support the previously proposed notion that autophagy counters S. flexneri dissemination.


Subject(s)
Autophagy , Bacterial Proteins/metabolism , Epithelial Cells/microbiology , Host-Pathogen Interactions , Microtubule-Associated Proteins/metabolism , Shigella flexneri/growth & development , Vacuoles/microbiology , Bacterial Proteins/genetics , Cell Line , Dysentery, Bacillary/physiopathology , Humans , Intravital Microscopy , Microscopy, Confocal , Mutation , Protein Binding , Spatio-Temporal Analysis , Virulence Factors/genetics , Virulence Factors/metabolism
9.
Methods ; 127: 12-22, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28522322

ABSTRACT

Macropinocytosis is the uptake of extracellular fluid within vesicles of varying size that takes place during numerous cellular processes in a large variety of cells. A growing number of pathogens, including viruses, parasites, and bacteria are known to induce macropinocytosis during their entry into targeted host cells. We have recently discovered that the human enteroinvasive, bacterial pathogen Shigella causes in situ macropinosome formation during its entry into epithelial cells. These infection-associated macropinosomes are not generated to ingest the bacteria, but are instead involved in Shigella's intracellular niche formation. They make contacts with the phagocytosed shigellae to promote vacuolar membrane rupture and their cytosolic release. Here, we provide an overview of the different imaging approaches that are currently used to analyze macropinocytosis during infectious processes with a focus on Shigella entry. We detail the advantages and disadvantages of genetically encoded reporters as well as chemical probes to trace fluid phase uptake. In addition, we report how such reporters can be combined with ultrastructural approaches for correlative light electron microscopy either in thin sections or within large volumes. The combined imaging techniques introduced here provide a detailed characterization of macropinosomes during bacterial entry, which, apart from Shigella, are relevant for numerous other ones, including Salmonella, Brucella or Mycobacteria.


Subject(s)
Bacteriological Techniques/methods , Dysentery, Bacillary/diagnostic imaging , Endosomes/ultrastructure , Host-Pathogen Interactions , Pinocytosis , Biomarkers , Dysentery, Bacillary/physiopathology , Endosomes/microbiology , Humans , Microscopy, Electron/methods , Shigella
10.
Mol Microbiol ; 99(4): 658-73, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26481305

ABSTRACT

Shigella sonnei is a bacterial pathogen and causative agent of bacillary dysentery. It deploys a type III secretion system to inject effector proteins into host epithelial cells and macrophages, an essential step for tissue invasion and immune evasion. Although the arsenal of bacterial effectors and their cellular targets have been studied extensively, little is known about the prerequisites for deployment of type III secreted proteins during infection. Here, we describe a novel S. sonnei adhesin, SSO1327 which is a multivalent adhesion molecule (MAM) required for invasion of epithelial cells and macrophages and for infection in vivo. The S. sonnei MAM mediates intimate attachment to host cells, which is required for efficient translocation of type III effectors into host cells. SSO1327 is non-redundant to IcsA; its activity is independent of type III secretion. In contrast to the up-regulation of IcsA-dependent and independent attachment and invasion by deoxycholate in Shigella flexneri, deoxycholate negatively regulates IcsA and MAM in S. sonnei resulting in reduction in attachment and invasion and virulence attenuation in vivo. A strain deficient for SSO1327 is avirulent in vivo, but still elicits a host immune response.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Dysentery, Bacillary/microbiology , Shigella sonnei/genetics , Shigella sonnei/pathogenicity , Adhesins, Bacterial/genetics , Adhesins, Bacterial/isolation & purification , Animals , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , DNA-Binding Proteins/genetics , Deoxycholic Acid/metabolism , Disease Models, Animal , Dysentery, Bacillary/physiopathology , Epithelial Cells/microbiology , Guinea Pigs , HeLa Cells , Humans , Keratoconjunctivitis/microbiology , Larva/microbiology , Macrophages/microbiology , Moths , Shigella flexneri/metabolism , Transcription Factors/genetics , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism , Up-Regulation , Virulence
11.
PLoS One ; 9(11): e112738, 2014.
Article in English | MEDLINE | ID: mdl-25405985

ABSTRACT

Shigella flexneri is a human pathogen that triggers its own entry into intestinal cells and escapes primary vacuoles to gain access to the cytosolic compartment. As cytosolic and motile bacteria encounter the cell cortex, they spread from cell to cell through formation of membrane protrusions that resolve into secondary vacuoles in adjacent cells. Here, we examined the roles of the Type 3 Secretion System (T3SS) in S. flexneri dissemination in HT-29 intestinal cells infected with the serotype 2a strain 2457T. We generated a 2457T strain defective in the expression of MxiG, a central component of the T3SS needle apparatus. As expected, the ΔmxiG strain was severely affected in its ability to invade HT-29 cells, and expression of mxiG under the control of an arabinose inducible expression system (ΔmxiG/pmxiG) restored full infectivity. In this experimental system, removal of the inducer after the invasion steps (ΔmxiG/pmxiG (Ara withdrawal)) led to normal actin-based motility in the cytosol of HT-29 cells. However, the time spent in protrusions until vacuole formation was significantly increased. Moreover, the number of formed protrusions that failed to resolve into vacuoles was also increased. Accordingly, the ΔmxiG/pmxiG (Ara withdrawal) strain failed to trigger tyrosine phosphorylation in membrane protrusions, a signaling event that is required for the resolution of protrusions into vacuoles. Finally, the ΔmxiG/pmxiG (Ara withdrawal) strain failed to escape from the formed secondary vacuoles, as previously reported in non-intestinal cells. Thus, the T3SS system displays multiple roles in S. flexneri dissemination in intestinal cells, including the tyrosine kinase signaling-dependent resolution of membrane protrusions into secondary vacuoles, and the escape from the formed secondary vacuoles.


Subject(s)
Bacterial Proteins/metabolism , Cell Surface Extensions/physiology , Dysentery, Bacillary/physiopathology , Shigella flexneri/metabolism , Type III Secretion Systems/metabolism , Vacuoles/metabolism , Bacterial Proteins/genetics , DNA Primers/genetics , Dysentery, Bacillary/metabolism , Fluorescent Antibody Technique , HT29 Cells , Humans , Phosphorylation , Protein-Tyrosine Kinases/metabolism
12.
J Health Popul Nutr ; 32(4): 580-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25895190

ABSTRACT

Shigellosis presents with varied clinical features are dictated by the species involved, virulence factors of the strain, and the host immune status. We studied the species, virulence genes, and antibiotic susceptibility pattern of the Shigella strains isolated from 33 children aged less than 12 years, with clinical features of shigellosis. Identification and antibiotic sensitivity of Shigella species were done using disc diffusion and E-test. Multiplex PCR was done for the detection of virulence genes (ipaH, ial, set1A, set1B, sen, and stx) and ESBL genes. Parents of the children were interviewed using structured questionnaire to assess the severity of the disease; 26 (79%) of the isolates were Shigella flexneri. Ciprofloxacin and ceftriaxone resistance was seen in 23 (69%) and 3 (9%) Shigella isolates respectively. Two ceftriaxone-resistant strains were found to harbour blaCTX gene and the third blaTEM gene. Virulence gene ipaH was detected in 100% of strains while ial, sen, setlA, and setlB were detected in 85%, 61%, 48%, and 48% respectively.


Subject(s)
Dysentery, Bacillary/diagnosis , Dysentery, Bacillary/microbiology , Anti-Bacterial Agents , Ceftriaxone/pharmacology , Child , Child, Preschool , Ciprofloxacin/pharmacology , Drug Resistance, Bacterial/genetics , Dysentery, Bacillary/physiopathology , Feces/microbiology , Female , Humans , Infant , Male , Microbial Sensitivity Tests , Shigella/genetics , Shigella/isolation & purification , Shigella flexneri/genetics , Shigella flexneri/isolation & purification , Virulence Factors/genetics
13.
Expert Rev Vaccines ; 12(1): 43-55, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23256738

ABSTRACT

The burden of dysentery due to shigellosis among children in the developing world is still a major concern. A safe and efficacious vaccine against this disease is a priority, since no licensed vaccine is available. This review provides an update of vaccine achievements focusing on subunit vaccine strategies and the forthcoming strategies surrounding this approach. In particular, this review explores several aspects of the pathogenesis of shigellosis and the elicited immune response as being the basis of vaccine requirements. The use of appropriate Shigella antigens, together with the right adjuvants, may offer safety, efficacy and more convenient delivery methods for massive worldwide vaccination campaigns.


Subject(s)
Dysentery, Bacillary/prevention & control , Shigella Vaccines , Shigella/immunology , Child , Child, Preschool , Dysentery, Bacillary/immunology , Dysentery, Bacillary/physiopathology , Humans , Shigella/classification , Shigella Vaccines/adverse effects , Shigella Vaccines/immunology , Vaccines, Subunit/adverse effects , Vaccines, Subunit/immunology
14.
Cell Host Microbe ; 11(4): 325-36, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22520461

ABSTRACT

Bacteria move between cells in the epithelium using a sequential pseudopodium-mediated process but the underlying mechanisms remain unclear. We show that during cell-to-cell movement, Shigella-containing pseudopodia target epithelial tricellular junctions, the contact point where three epithelial cells meet. The bacteria-containing pseudopodia were engulfed by neighboring cells only in the presence of tricellulin, a protein essential for tricellular junction integrity. Shigella cell-to-cell spread, but not pseudopodium protrusion, also depended on phosphoinositide 3-kinase, clathrin, Epsin-1, and Dynamin-2, which localized beneath the plasma membrane of the engulfing cell. Depleting tricellulin, Epsin-1, clathrin, or Dynamin-2 expression reduced Shigella cell-to-cell spread, whereas AP-2, Dab2, and Eps15 were not critical for this process. Our findings highlight a mechanism for Shigella dissemination into neighboring cells via targeting of tricellular junctions and a noncanonical clathrin-dependent endocytic pathway.


Subject(s)
Clathrin/metabolism , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/physiopathology , Endocytosis , Epithelial Cells/microbiology , Shigella/physiology , Tight Junctions/metabolism , Animals , Cell Line , Dysentery, Bacillary/metabolism , Epithelial Cells/metabolism , Humans , MARVEL Domain Containing 2 Protein , Membrane Proteins/metabolism , Tight Junctions/microbiology
15.
Foodborne Pathog Dis ; 9(5): 457-64, 2012 May.
Article in English | MEDLINE | ID: mdl-22506961

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) cause nonbloody (NBD) and bloody diarrhea (BD), and hemolytic uremic syndrome (HUS). Cattle have been described as their main reservoir. STEC O157:H7 is recognized as the predominant serotype in clinical infections, but much less is known about the dominant subtypes in humans and animals or their genetic relatedness. The aims of this study were to compare the STEC O157 subtypes found in sporadic human infections with those in the bovine reservoir using stx-genotyping, phage typing, and XbaI-pulsed-field gel electrophoresis (PFGE), and correlate the subtypes with the severity of clinical manifestations. The 280 STEC O157:H7 strains collected included in this study were isolated from HUS (n=122), BD (n=69), and NBD (n=30) cases, and healthy carriers (n=5), and from bovines (n=54) in the abattoirs. The stx-genotyping showed that stx2/stx(2c(vh-a)) was predominant in human (76.1%) and in bovine strains (55.5%), whereas the second more important genotype was stx2 (20.8%) in human and stx(2c(vh-a)) (16.7%) in cattle strains. In human strains, PT4 (37.6%), PT49 (24.3%), and PT2 (18.6%) were the most frequent PTs (80.5%). In bovine isolates, PT2 (26%), PT39 (16.7%), and PT4 and PT49 (11.1% each) were predominant. By XbaI-PFGE, all 280 strains yielded 148 patterns with 75% similarity, and 169 strains were grouped in 37 clusters. Identical PT-PFGE-stx profile combinations were detected in strains of both origins: PT4-AREXH01.0011-stx2/stx(2c(vh-a)) (12 humans and one bovine), PT4-AREXH01.0543-stx2/stx(2c(vh-a)) (one human and four bovines), PT2-AREXH01.0076-stx2/stx(2c(vh-a)) (one human and four bovines), PT49-AREXH01.0175-stx2/stx(2c(vh-a)) (seven humans and one bovine), and PT49-AREXH01.0022-stx2/stx(2c(vh-a)) (seven humans and one bovine). No correlation was found among the stx-genotypes, the phage type, and the clinical symptoms.


Subject(s)
Cattle/microbiology , Disease Reservoirs/microbiology , Dysentery, Bacillary/microbiology , Escherichia coli Infections/microbiology , Escherichia coli O157/isolation & purification , Hemolytic-Uremic Syndrome/microbiology , Abattoirs , Amplified Fragment Length Polymorphism Analysis , Animals , Argentina/epidemiology , Bacteriophage Typing , Carrier State/microbiology , Dysentery, Bacillary/epidemiology , Dysentery, Bacillary/physiopathology , Electrophoresis, Gel, Pulsed-Field , Endemic Diseases , Escherichia coli Infections/epidemiology , Escherichia coli Infections/physiopathology , Escherichia coli O157/classification , Escherichia coli O157/metabolism , Gastrointestinal Hemorrhage/etiology , Genotyping Techniques , Hemolytic-Uremic Syndrome/epidemiology , Hemolytic-Uremic Syndrome/physiopathology , Humans , Population Surveillance , Severity of Illness Index , Shiga Toxin 2/genetics , Shiga Toxin 2/metabolism
16.
Cell Host Microbe ; 9(6): 449-50, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21669393

ABSTRACT

The type III secretion system-dependent epithelial invasion and dissemination of Shigella is stimulated by ATP released through hemichannels. Romero et al. (2011) show that prior to epithelial contact, Shigella is captured by nanometer-thin micropodial extensions at a distance from the cell surface, in a process involving ATP and connexin-mediated signaling.


Subject(s)
Dysentery, Bacillary/microbiology , Epithelial Cells/microbiology , Host-Pathogen Interactions , Pseudopodia/microbiology , Shigella/physiology , Adenosine Triphosphate/metabolism , Dysentery, Bacillary/enzymology , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/physiopathology , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Pseudopodia/enzymology , Pseudopodia/metabolism , Shigella/genetics
17.
Cell Host Microbe ; 9(6): 508-19, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21669399

ABSTRACT

Shigella, the causative agent of bacillary dysentery in humans, invades epithelial cells, using a type III secretory system (T3SS) to inject bacterial effectors into host cells and remodel the actin cytoskeleton. ATP released through connexin hemichanels on the epithelial membrane stimulates Shigella invasion and dissemination in epithelial cells. Here, we show that prior to contact with the cell body, Shigella is captured by nanometer-thin micropodial extensions (NMEs) at a distance from the cell surface, in a process involving the T3SS tip complex proteins and stimulated by ATP- and connexin-mediated signaling. Upon bacterial contact, NMEs retract, bringing bacteria in contact with the cell body, where invasion occurs. ATP stimulates Erk1/2 activation, which controls actin retrograde flow in NMEs and their retraction. These findings reveal previously unappreciated facets of interaction of an invasive bacterium with host cells and a prominent role for Erk1/2 in the control of filopodial dynamics.


Subject(s)
Adenosine Triphosphate/metabolism , Dysentery, Bacillary/enzymology , Dysentery, Bacillary/microbiology , Epithelial Cells/microbiology , Host-Pathogen Interactions , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pseudopodia/microbiology , Shigella/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/physiopathology , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Pseudopodia/enzymology , Pseudopodia/metabolism , Shigella/genetics , Signal Transduction
18.
Cell Death Dis ; 2: e122, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21368893

ABSTRACT

Modulation of death is a pathogen strategy to establish residence and promote survival in host cells and tissues. Shigella spp. are human pathogens that invade colonic mucosa, where they provoke lesions caused by their ability to manipulate the host cell responses. Shigella spp. induce various types of cell death in different cell populations. However, they are equally able to protect host cells from death. Here, we have investigated on the molecular mechanisms and cell effectors governing the balance between survival and death in epithelial cells infected with Shigella. To explore these aspects, we have exploited both, the HeLa cell invasion assay and a novel ex vivo human colon organ culture model of infection that mimics natural conditions of shigellosis. Our results definitely show that Shigella induces a rapid intrinsic apoptosis of infected cells, via mitochondrial depolarization and the ensuing caspase-9 activation. Moreover, for the first time we identify the eukaryotic stress-response factor growth arrest and DNA damage 45α as a key player in the induction of the apoptotic process elicited by Shigella in epithelial cells, revealing an unexplored role of this molecule in the course of infections sustained by invasive pathogens.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , Dysentery, Bacillary/metabolism , Epithelial Cells/cytology , Mitochondria/metabolism , Nuclear Proteins/metabolism , Shigella flexneri/physiology , Cell Cycle Proteins/genetics , Cell Death , Colon/cytology , Colon/metabolism , Colon/microbiology , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/physiopathology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , HeLa Cells , Humans , In Vitro Techniques , Nuclear Proteins/genetics , Shigella flexneri/genetics
19.
Gut ; 60(4): 473-84, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21139062

ABSTRACT

BACKGROUND: Enteric glial cells (EGCs) are important regulators of intestinal epithelial barrier (IEB) functions. EGC-derived S-nitrosoglutathione (GSNO) has been shown to regulate IEB permeability. Whether EGCs and GSNO protect the IEB during infectious insult by pathogens such as Shigella flexneri is not known. METHODS: S flexneri effects were characterised using in vitro coculture models of Caco-2 cells and EGCs (or GSNO), ex vivo human colonic mucosa, and in vivo ligated rabbit intestinal loops. The effect of EGCs on S flexneri-induced changes in the invasion area and the inflammatory response were analysed by combining immunohistochemical, ELISA and PCR methods. Expression of small G-proteins was analysed by western blot. Expression of ZO-1 and localisation of bacteria were analysed by fluorescence microscopy. RESULTS: EGCs significantly reduced barrier lesions and inflammatory response induced by S flexneri in Caco-2 monolayers. The EGC-mediated effects were reproduced by GSNO, but not by reduced glutathione, and pharmacological inhibition of pathways involved in GSNO synthesis reduced EGC protecting effects. Furthermore, expression of Cdc42 and phospho-PAK in Caco-2 monolayers was significantly reduced in the presence of EGCs or GSNO. In addition, changes in ZO-1 expression and distribution induced by S flexneri were prevented by EGCs and GSNO. Finally, GSNO reduced S flexneri-induced lesions of the IEB in human mucosal colonic explants and in a rabbit model of shigellosis. CONCLUSION: These results highlight a major protective function of EGCs and GSNO in the IEB against S flexneri attack. Consequently, this study lays the scientific basis for using GSNO to reduce barrier susceptibility to infectious or inflammatory challenge.


Subject(s)
Dysentery, Bacillary/pathology , Intestinal Mucosa/innervation , Neuroglia/physiology , S-Nitrosoglutathione/metabolism , Shigella flexneri/physiology , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Translocation/physiology , Caco-2 Cells , Coculture Techniques , Colon/innervation , Colon/microbiology , Drug Evaluation, Preclinical/methods , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/physiopathology , Enteric Nervous System/physiology , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Permeability , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , S-Nitrosoglutathione/pharmacology , Shigella flexneri/drug effects , cdc42 GTP-Binding Protein/metabolism
20.
J Infect Dis ; 201(6): 903-11, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20136414

ABSTRACT

BACKGROUND: The lack of a standardized laboratory animal model that mimics key aspects of human shigellosis remains a major obstacle to addressing questions about pathogenesis, screening therapeutics, and evaluation of vaccines. METHODS: We characterized a piglet model for Shigella dysenteriae type 1. RESULTS: Piglets developed acute diarrhea, anorexia, and dehydration, which could often be fatal, with symptom severity depending on age and dose. Bacteria were apparent in the lumen and on the surface epithelium throughout the gut initially, but severe mucosal damage and bacterial cellular invasion were most profound in the colon. Detached necrotic colonocytes were present in the lumen, with inflammatory cells outpouring from damaged mucosa. High levels of interleukin (IL)-8 and IL-12 were followed by high levels of other proinflammatory cytokines. Elevated levels of tumor necrosis factor-alpha, IL-1beta, IL-6, and IL-10 were detected in feces and in gut segments from infected animals. Bacteria were present inside epithelial cells and within colonic lamina propria. In contrast, an isogenic strain lacking Shiga toxin induced similar but milder symptoms, with moderate mucosal damage and lower cytokine levels. CONCLUSION: We conclude that piglets are highly susceptible to shigellosis, providing a useful tool with which to compare vaccine candidates for immunogenicity, reactogenicity, and response to challenge; investigate the role of virulence factors; and test the efficacy of microbial agents.


Subject(s)
Disease Models, Animal , Dysentery, Bacillary/physiopathology , Gastroenteritis/microbiology , Shigella dysenteriae , Swine , Animals , Case-Control Studies , Colony Count, Microbial , Cytokines/analysis , Dysentery, Bacillary/microbiology , Euthanasia, Animal , Feces/microbiology , Gastroenteritis/physiopathology , Gastrointestinal Tract/microbiology , Interleukin-12 , Interleukin-8 , Microscopy, Electron , Shigella dysenteriae/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...