Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Indian J Med Res ; 158(3): 303-310, 2023 09.
Article in English | MEDLINE | ID: mdl-37815071

ABSTRACT

Background & objectives: The study of Shigella pathogenesis at present is severely hampered by the lack of a relevant animal model that replicates human bacillary dysentery. Different Shigella serogroups cause varying severity of clinical illness. Ex vivo colonization of Shigella flexneri, S. dysenteriae and S. sonnei were characterized in human paediatric colonic pinch biopsies in the in vitro organ culture (IVOC) model to study the invasiveness of Shigella by gentamicin protection assay (GPA). Furthermore, the expression of antimicrobial peptides (AMPs) in response to different serotypes of Shigella was also studied in IVOC model. Methods: IVOC explants were inoculated with 109 colony forming units of different serotypes of Shigella and recovery of bacteria studied. Histopathological analysis was carried out to study inflammatory immune responses. GPA was done to elucidate the invasiveness of different serotypes of Shigella. Secretions of AMPs were measured by enzyme-linked immunosorbent assay (ELISA). Western blotting was performed to check the expression of AMPs and nuclear factor kappa B in IVOC explants. Results: After 24 h post-infection, the colon biopsies showed intense inflammatory reaction. In both IVOC and GPA, S. dysenteriae 1 was the most invasive as compared to S. flexneri and S. sonnei. S. sonnei was the least invasive. ELISA demonstrated that S. sonnei dampened the HBD (human ß-defensin)-2 responses whereas there was augmentation by S. dysenteriae and there was a modest but non-significant increase by S. flexneri. A modest increase in HBD-3 by S. sonnei and S. flexneri was observed but was not found to be significant. However, western blotting data showed upregulation of all AMPs by all serotypes. Western blotting is more sensitive than ELISA. Interpretation & conclusions: In the present study, differences in invasiveness and AMP production induced by different serotypes of Shigella were found. Human intestinal IVOC represents a model system to investigate early interaction between pathogenic bacteria and the human gut.


Subject(s)
Dysentery, Bacillary , Shigella , Animals , Child , Humans , Serogroup , Antimicrobial Peptides , Shigella/genetics , Dysentery, Bacillary/genetics , Dysentery, Bacillary/microbiology , Shigella flexneri/genetics
2.
PLoS Pathog ; 16(8): e1008822, 2020 08.
Article in English | MEDLINE | ID: mdl-32866204

ABSTRACT

Shigella flexneri invades host cells by entering within a bacteria-containing vacuole (BCV). In order to establish its niche in the host cytosol, the bacterium ruptures its BCV. Contacts between S. flexneri BCV and infection-associated macropinosomes (IAMs) formed in situ have been reported to enhance BCV disintegration. The mechanism underlying S. flexneri vacuolar escape remains however obscure. To decipher the molecular mechanism priming the communication between the IAMs and S. flexneri BCV, we performed mass spectrometry-based analysis of the magnetically purified IAMs from S. flexneri-infected cells. While proteins involved in host recycling and exocytic pathways were significantly enriched at the IAMs, we demonstrate more precisely that the S. flexneri type III effector protein IpgD mediates the recruitment of the exocyst to the IAMs through the Rab8/Rab11 pathway. This recruitment results in IAM clustering around S. flexneri BCV. More importantly, we reveal that IAM clustering subsequently facilitates an IAM-mediated unwrapping of the ruptured vacuole membranes from S. flexneri, enabling the naked bacterium to be ready for intercellular spread via actin-based motility. Taken together, our work untangles the molecular cascade of S. flexneri-driven host trafficking subversion at IAMs to develop its cytosolic lifestyle, a crucial step en route for infection progression at cellular and tissue level.


Subject(s)
Dysentery, Bacillary , Shigella flexneri , Signal Transduction , Vacuoles , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Dysentery, Bacillary/genetics , Dysentery, Bacillary/metabolism , HeLa Cells , Humans , Shigella flexneri/genetics , Shigella flexneri/metabolism , Shigella flexneri/pathogenicity , Vacuoles/genetics , Vacuoles/metabolism , Vacuoles/microbiology , Virulence Factors/genetics , Virulence Factors/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
3.
Molecules ; 27(6)2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35335300

ABSTRACT

Shigella species account for the second-leading cause of deaths due to diarrheal diseases among children of less than 5 years of age. The emergence of multi-drug-resistant Shigella isolates and the lack of availability of Shigella vaccines have led to the pertinence in the efforts made for the development of new therapeutic strategies against shigellosis. Consequently, designing small-interfering RNA (siRNA) candidates against such infectious agents represents a novel approach to propose new therapeutic candidates to curb the rampant rise of anti-microbial resistance in such pathogens. In this study, we analyzed 264 conserved sequences from 15 different conserved virulence genes of Shigella sp., through extensive rational validation using a plethora of first-generation and second-generation computational algorithms for siRNA designing. Fifty-eight siRNA candidates were obtained by using the first-generation algorithms, out of which only 38 siRNA candidates complied with the second-generation rules of siRNA designing. Further computational validation showed that 16 siRNA candidates were found to have a substantial functional efficiency, out of which 11 siRNA candidates were found to be non-immunogenic. Finally, three siRNA candidates exhibited a sterically feasible three-dimensional structure as exhibited by parameters of nucleic acid geometry such as: the probability of wrong sugar puckers, bad backbone confirmations, bad bonds, and bad angles being within the accepted threshold for stable tertiary structure. Although the findings of our study require further wet-lab validation and optimization for therapeutic use in the treatment of shigellosis, the computationally validated siRNA candidates are expected to suppress the expression of the virulence genes, namely: IpgD (siRNA 9) and OspB (siRNA 15 and siRNA 17) and thus act as a prospective tool in the RNA interference (RNAi) pathway. However, the findings of our study require further wet-lab validation and optimization for regular therapeutic use for treatment of shigellosis.


Subject(s)
Dysentery, Bacillary , Shigella , Child , Diarrhea/drug therapy , Dysentery, Bacillary/drug therapy , Dysentery, Bacillary/genetics , Humans , RNA Interference , RNA, Small Interfering/metabolism , Shigella/genetics
4.
Genomics ; 112(1): 774-781, 2020 01.
Article in English | MEDLINE | ID: mdl-31125598

ABSTRACT

In recent years, researchers have begun to use Caenorhabditis elegans as a potential animal model to study Shigella pathogenesis. This study aims to further develop this model using RNA-sequencing to understand which pathways/cellular characteristics are affected and potentially cause death in Shigella-exposed worms. We identified 1631 differentially expressed genes in Shigella-exposed worms (6 h exposure). A number of these genes encode proteins involved in fatty-acid ß-oxidation (FAO), antioxidant defense and autophagy. The down-regulation of acyl-CoA dehydrogenases would impede FAO, reducing the overall energy to combat Shigella in the worm's intestinal tract. This is potentially coupled with the production of reactive oxygen species (ROS) that may not be fully quenched by antioxidant defense proteins, leading to damaged cellular organelles in the worm's intestinal cells. These cells may undergo autophagy to remove the mounting damage, but may eventually undergo cell death.


Subject(s)
Caenorhabditis elegans/genetics , Dysentery, Bacillary/genetics , Shigella flexneri , Animals , Antioxidants/metabolism , Autophagy/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Disease Models, Animal , Dysentery, Bacillary/metabolism , Fatty Acids/metabolism , RNA-Seq , Transcriptome
5.
Infect Immun ; 87(4)2019 04.
Article in English | MEDLINE | ID: mdl-30642906

ABSTRACT

The enteric bacterium and intracellular human pathogen Shigella causes hundreds of millions of cases of the diarrheal disease shigellosis per year worldwide. Shigella is acquired by ingestion of contaminated food or water; upon reaching the colon, the bacteria invade colonic epithelial cells, replicate intracellularly, spread to adjacent cells, and provoke an intense inflammatory response. There is no animal model that faithfully recapitulates human disease; thus, cultured cells have been used to model Shigella pathogenesis. However, the use of transformed cells in culture does not provide the same environment to the bacteria as the normal human intestinal epithelium. Recent advances in tissue culture now enable the cultivation of human intestinal enteroids (HIEs), which are derived from human intestinal stem cells, grown ex vivo, and then differentiated into "mini-intestines." Here, we demonstrate that HIEs can be used to model Shigella pathogenesis. We show that Shigella flexneri invades polarized HIE monolayers preferentially via the basolateral surface. After S. flexneri invades HIE monolayers, S. flexneri replicates within HIE cells and forms actin tails. S. flexneri also increases the expression of HIE proinflammatory signals and the amino acid transporter SLC7A5. Finally, we demonstrate that disruption of HIE tight junctions enables S. flexneri invasion via the apical surface.


Subject(s)
Dysentery, Bacillary/microbiology , Intestinal Mucosa/microbiology , Models, Biological , Organoids/microbiology , Shigella flexneri/physiology , Cell Culture Techniques , Dysentery, Bacillary/genetics , Dysentery, Bacillary/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Organoids/growth & development , Organoids/metabolism , Shigella flexneri/pathogenicity , Stem Cells/cytology , Stem Cells/microbiology , Virulence
6.
Indian J Med Res ; 149(2): 247-256, 2019 02.
Article in English | MEDLINE | ID: mdl-31219090

ABSTRACT

Background & objectives: Bacillary dysentery caused by Shigella spp. remains an important cause of the crisis in low-income countries. It has been observed that Shigella species have become increasingly resistant to most widely used antimicrobials. In this study, the antimicrobial resistance, virulence and plasmid profile of clinical isolates of Shigella species were determined. Methods: Sixty clinical Shigella isolates were subjected to whole-genome sequencing using Ion Torrent platform and the genome sequences were analyzed for the presence of acquired resistance genes, virulence genes and plasmids using web-based software tools. Results: Genome analysis revealed more resistance genes in Shigella flexneri than in other serogroups. Among ß-lactamases, blaOXA-1was predominantly seen followed by the blaTEM-1B and blaEC genes. For quinolone resistance, the qnr S gene was widely seen. Novel mutations in gyr B, par C and par E genes were observed. Cephalosporins resistance gene, blaCTX-M-15 was identified and plasmid-mediated AmpC ß-lactamases genes were found among the isolates. Further, a co-trimoxazole resistance gene was identified in most of the isolates studied. Virulence genes such as ipaD, ipaH, virF, senB, iha, capU, lpfA, sigA, pic, sepA, celb and gad were identified. Plasmid analysis revealed that the IncFII was the most commonly seen plasmid type in the isolates. Interpretation & conclusions: The presence of quinolone and cephalosporin resistance genes in Shigella serogroups has serious implications for the further spread of this resistance to other enteric pathogens or commensal organisms. This suggests the need for continuous surveillance to understand the epidemiology of the resistance.


Subject(s)
Bacterial Proteins/genetics , Drug Resistance, Bacterial/genetics , Dysentery, Bacillary/genetics , Shigella/genetics , beta-Lactamases/genetics , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Cephalosporins/adverse effects , Cephalosporins/therapeutic use , Dysentery, Bacillary/drug therapy , Dysentery, Bacillary/epidemiology , Dysentery, Bacillary/microbiology , Feces/microbiology , Humans , Microbial Sensitivity Tests , Plasmids/drug effects , Plasmids/genetics , Serogroup , Shigella/pathogenicity , Whole Genome Sequencing
7.
Indian J Med Res ; 149(3): 412-417, 2019 03.
Article in English | MEDLINE | ID: mdl-31249208

ABSTRACT

Background & objectives: : Shiga toxin (Stx) is produced by Shigella dysenteriae, a Gram-negative, facultative anaerobic bacillus that causes shigellosis, haemolytic uraemic syndrome (HUS) and Reiter's syndrome. The detection methods for shiga toxin needs to be rapid, accurate, reliable and must be extensively evaluated under field conditions. The aim of this study was to develop rapid, sensitive and specific detection method for Stx. Methods: : Mice and rabbits were immunized with purified recombinant Shiga toxin B (rStxB). Using these antibodies dot ELISA, sandwich ELISA and flow through assay were developed. Results: : The high-titre antibodies specifically reacted with purified rStxB. Dot-ELISA, sandwich ELISA and flow-through assay were developed and standardized that could detect StxB with limit of detection (LOD) of 9.75, 9.7 ng/ml and 0.46 µg/cassette, respectively. Interpretation & conclusions: : The rStxB was used to produce antibodies to avoid handling of pathogen. The Flow through assay 'developed was specific, rapid and field amenable.


Subject(s)
Dysentery, Bacillary/diagnosis , Hemolytic-Uremic Syndrome/diagnosis , Shiga Toxin/isolation & purification , Shigella dysenteriae/genetics , Animals , Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Arthritis, Reactive/diagnosis , Arthritis, Reactive/genetics , Arthritis, Reactive/microbiology , Dysentery, Bacillary/genetics , Dysentery, Bacillary/microbiology , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/microbiology , Humans , Mice , Shiga Toxin/genetics , Shigella dysenteriae/pathogenicity
8.
EMBO J ; 33(22): 2606-22, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25216677

ABSTRACT

HP1 proteins are transcriptional regulators that, like histones, are targets for post-translational modifications defining an HP1-mediated subcode. HP1γ has multiple phosphorylation sites, including serine 83 (S83) that marks it to sites of active transcription. In a guinea pig model for Shigella enterocolitis, we observed that the defective type III secretion mxiD Shigella flexneri strain caused more HP1γ phosphorylation in the colon than the wild-type strain. Shigella interferes with HP1 phosphorylation by injecting the phospholyase OspF. This effector interacts with HP1γ and alters its phosphorylation at S83 by inactivating ERK and consequently MSK1, a downstream kinase. MSK1 that here arises as a novel HP1γ kinase, phosphorylates HP1γ at S83 in the context of an MSK1-HP1γ complex, and thereby favors its accumulation on its target genes. Genome-wide transcriptome analysis reveals that this mechanism is linked to up-regulation of proliferative gene and fine-tuning of immune gene expression. Thus, in addition to histones, bacteria control host transcription by modulating the activity of HP1 proteins, with potential implications in transcriptional reprogramming at the mucosal barrier.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Carbon-Oxygen Lyases/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Dysentery, Bacillary/metabolism , Enterocolitis/metabolism , Shigella flexneri/metabolism , Transcriptome , Animals , Bacterial Outer Membrane Proteins/genetics , Carbon-Oxygen Lyases/genetics , Cells, Cultured , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Disease Models, Animal , Dysentery, Bacillary/genetics , Dysentery, Bacillary/pathology , Enterocolitis/genetics , Enterocolitis/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Genome-Wide Association Study , Guinea Pigs , Mice , Mice, Mutant Strains , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Shigella flexneri/genetics
9.
J Biol Chem ; 291(38): 20149-62, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27445336

ABSTRACT

The type III secretion system effector protein NleE from enteropathogenic Escherichia coli plays a key role in the inhibition of NF-κB activation during infection. NleE inactivates the ubiquitin chain binding activity of host proteins TAK1-binding proteins 2 and 3 (TAB2 and TAB3) by modifying the Npl4 zinc finger domain through S-adenosyl methionine-dependent cysteine methylation. Using yeast two-hybrid protein interaction studies, we found that a conserved region between amino acids 34 and 52 of NleE, in particular the motif (49)GITR(52), was critical for TAB2 and TAB3 binding. NleE mutants lacking (49)GITR(52) were unable to methylate TAB3, and wild type NleE but not NleE(49AAAA52) where each of GITR was replaced with alanine restored the ability of an nleE mutant to inhibit IL-8 production during infection. Another NleE target, ZRANB3, also associated with NleE through the (49)GITR(52) motif. Ectopic expression of an N-terminal fragment of NleE (NleE(34-52)) in HeLa cells showed competitive inhibition of wild type NleE in the suppression of IL-8 secretion during enteropathogenic E. coli infection. Similar results were observed for the NleE homologue OspZ from Shigella flexneri 6 that also bound TAB3 through the (49)GITR(52) motif and decreased IL-8 transcription through modification of TAB3. In summary, we have identified a unique substrate-binding motif in NleE and OspZ that is required for the ability to inhibit the host inflammatory response.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA Helicases/metabolism , Dysentery, Bacillary/metabolism , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Shigella flexneri/metabolism , Virulence Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Motifs , DNA Helicases/genetics , Dysentery, Bacillary/genetics , Enteropathogenic Escherichia coli/genetics , Escherichia coli Infections/genetics , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Binding , Shigella flexneri/genetics
10.
Curr Top Microbiol Immunol ; 397: 91-115, 2016.
Article in English | MEDLINE | ID: mdl-27460806

ABSTRACT

Shigella spp. are the causative agents of bacillary dysentery, leading to extensive mortality and morbidity worldwide. These facultative intracellular bacteria invade the epithelium of the colon and the rectum, inducing a severe inflammatory response from which the symptoms of the disease originate. Shigella are human pathogens able to manipulate and subvert the innate immune system surveillance. Shigella dampens inflammasome activation in epithelial cells. In infected macrophages, inflammasome activation and IL-1ß and IL-18 release lead to massive neutrophil recruitment and greatly contribute to inflammation. Here, we describe how Shigella hijacks and finely tunes inflammasome activation in the different cell populations involved in pathogenesis: epithelial cells, macrophages, neutrophils, DCs, and B and T lymphocytes. Shigella emerges as a "sly" pathogen that switches on/off the inflammasome mechanisms in order to optimize the interaction with the host and establish a successful infection.


Subject(s)
Dysentery, Bacillary/immunology , Inflammasomes/immunology , Shigella/immunology , Animals , Dysentery, Bacillary/genetics , Dysentery, Bacillary/microbiology , Host-Pathogen Interactions , Humans , Inflammasomes/genetics , Macrophages/immunology , Neutrophils/immunology , Shigella/genetics , Shigella/physiology
11.
EMBO Rep ; 15(9): 965-72, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25097252

ABSTRACT

Shigella flexneri, the etiological agent of bacillary dysentery, invades the human colonic epithelium and causes its massive inflammatory destruction. Little is known about the post-translational modifications implicated in regulating the host defense pathway against Shigella. Here, we show that SUMO-2 impairs Shigella invasion of epithelial cells in vitro. Using mice haploinsufficient for the SUMO E2 enzyme, we found that sumoylation regulates intestinal permeability and is required to restrict epithelial invasion and control mucosal inflammation. Quantitative proteomics reveals that Shigella infection alters the sumoylation status of a restricted set of transcriptional regulators involved in intestinal functions and inflammation. Consistent with this, sumoylation restricts the pro-inflammatory transcriptional response of Shigella-infected guts. Altogether, our results show that the SUMO pathway is an essential component of host innate protection, as it reduces the efficiency of two key steps of shigellosis: invasion and inflammatory destruction of the intestinal epithelium.


Subject(s)
Dysentery, Bacillary/metabolism , Intestines/microbiology , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation/genetics , Animals , Dysentery, Bacillary/genetics , Dysentery, Bacillary/microbiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Haploinsufficiency/genetics , Host-Pathogen Interactions/genetics , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Intestines/pathology , Mice , Protein Processing, Post-Translational/genetics , Shigella flexneri/metabolism , Shigella flexneri/pathogenicity , Small Ubiquitin-Related Modifier Proteins/genetics
12.
Proc Natl Acad Sci U S A ; 110(12): 4458-63, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23417297

ABSTRACT

The Gram-negative enteroinvasive bacterium Shigella flexneri is responsible for the endemic form of bacillary dysentery, an acute rectocolitis in humans. S. flexneri uses a type III secretion system to inject effector proteins into host cells, thus diverting cellular functions to its own benefit. Protective immunity to reinfection requires several rounds of infection to be elicited and is short-lasting, suggesting that S. flexneri interferes with the priming of specific immunity. Considering the key role played by T-lymphocyte trafficking in priming of adaptive immunity, we investigated the impact of S. flexneri on T-cell dynamics in vivo. By using two-photon microscopy to visualize bacterium-T-cell cross-talks in the lymph nodes, where the adaptive immunity is initiated, we provide evidence that S. flexneri, via its type III secretion system, impairs the migration pattern of CD4(+) T cells independently of cognate recognition of bacterial antigens. We show that bacterial invasion of CD4(+) T lymphocytes occurs in vivo, and results in cell migration arrest. In the absence of invasion, CD4(+) T-cell migration parameters are also dramatically altered. Signals resulting from S. flexneri interactions with subcapsular sinus macrophages and dendritic cells, and recruitment of polymorphonuclear cells are likely to contribute to this phenomenon. These findings indicate that S. flexneri targets T lymphocytes in vivo and highlight the role of type III effector secretion in modulating host adaptive immune responses.


Subject(s)
Adaptive Immunity , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Dysentery, Bacillary/immunology , Host-Pathogen Interactions/immunology , Shigella flexneri/physiology , Animals , Dysentery, Bacillary/genetics , Female , Mice , Mice, Knockout , Signal Transduction/immunology
13.
PLoS Pathog ; 9(9): e1003588, 2013.
Article in English | MEDLINE | ID: mdl-24039575

ABSTRACT

Autophagy, an ancient and highly conserved intracellular degradation process, is viewed as a critical component of innate immunity because of its ability to deliver cytosolic bacteria to the lysosome. However, the role of bacterial autophagy in vivo remains poorly understood. The zebrafish (Danio rerio) has emerged as a vertebrate model for the study of infections because it is optically accessible at the larval stages when the innate immune system is already functional. Here, we have characterized the susceptibility of zebrafish larvae to Shigella flexneri, a paradigm for bacterial autophagy, and have used this model to study Shigella-phagocyte interactions in vivo. Depending on the dose, S. flexneri injected in zebrafish larvae were either cleared in a few days or resulted in a progressive and ultimately fatal infection. Using high resolution live imaging, we found that S. flexneri were rapidly engulfed by macrophages and neutrophils; moreover we discovered a scavenger role for neutrophils in eliminating infected dead macrophages and non-immune cell types that failed to control Shigella infection. We observed that intracellular S. flexneri could escape to the cytosol, induce septin caging and be targeted to autophagy in vivo. Depletion of p62 (sequestosome 1 or SQSTM1), an adaptor protein critical for bacterial autophagy in vitro, significantly increased bacterial burden and host susceptibility to infection. These results show the zebrafish larva as a new model for the study of S. flexneri interaction with phagocytes, and the manipulation of autophagy for anti-bacterial therapy in vivo.


Subject(s)
Autophagy , Dysentery, Bacillary/metabolism , Macrophages/metabolism , Neutrophils/metabolism , Shigella flexneri/metabolism , Zebrafish/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Disease Models, Animal , Dysentery, Bacillary/genetics , Dysentery, Bacillary/pathology , Humans , Macrophages/microbiology , Macrophages/pathology , Neutrophils/microbiology , Neutrophils/pathology , Zebrafish/genetics , Zebrafish/microbiology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
PLoS Pathog ; 9(6): e1003409, 2013.
Article in English | MEDLINE | ID: mdl-23754945

ABSTRACT

NF-κB plays a central role in modulating innate immune responses to bacterial infections. Therefore, many bacterial pathogens deploy multiple mechanisms to counteract NF-κB activation. The invasion of and subsequent replication of Shigella within epithelial cells is recognized by various pathogen recognition receptors as pathogen-associated molecular patterns. These receptors trigger innate defense mechanisms via the activation of the NF-κB signaling pathway. Here, we show the inhibition of the NF-κB activation by the delivery of the IpaH E3 ubiquitin ligase family member IpaH0722 using Shigella's type III secretion system. IpaH0722 dampens the acute inflammatory response by preferentially inhibiting the PKC-mediated activation of NF-κB by ubiquitinating TRAF2, a molecule downstream of PKC, and by promoting its proteasome-dependent degradation.


Subject(s)
Bacterial Proteins/metabolism , Dysentery, Bacillary/enzymology , Epithelial Cells/metabolism , NF-kappa B/metabolism , Protein Kinase C/metabolism , Proteolysis , Shigella/enzymology , TNF Receptor-Associated Factor 2/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Secretion Systems/genetics , COS Cells , Chlorocebus aethiops , Dysentery, Bacillary/genetics , Dysentery, Bacillary/pathology , Epithelial Cells/microbiology , Epithelial Cells/pathology , HeLa Cells , Humans , Mice , Mice, Knockout , NF-kappa B/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Kinase C/genetics , Shigella/genetics , Signal Transduction/genetics , TNF Receptor-Associated Factor 2/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
15.
Curr Opin Gastroenterol ; 31(1): 30-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25394237

ABSTRACT

PURPOSE OF REVIEW: Shigella spp. are important etiologic agents of diarrhea worldwide. This review summarizes the recent findings on the epidemiology, diagnosis, virulence genes, and pathobiology of Shigella infection. RECENT FINDINGS: Shigella flexneri and Shigella sonnei have been identified as the main serogroups circulating in developing and developed countries, respectively. However, a shift in the dominant species from S. flexneri to S. sonnei has been observed in countries that have experienced recent improvements in socioeconomic conditions. Despite the increasing usage of molecular methods in the diagnosis and virulence characterization of Shigella strains, researchers have been unsuccessful in finding a specific target gene for this bacillus. New research has demonstrated the role of proteins whose expressions are temperature-regulated, as well as genes involved in the processes of adhesion, invasion, dissemination, and inflammation, aiding in the clarification of the complex pathobiology of shigellosis. SUMMARY: Knowledge about the epidemiologic profile of circulating serogroups of Shigella and an understanding of its pathobiology as well as of the virulence genes is important for the development of preventive measures and interventions to reduce the worldwide spread of shigellosis.


Subject(s)
Dysentery, Bacillary/genetics , Dysentery, Bacillary/immunology , Dysentery, Bacillary/diagnosis , Dysentery, Bacillary/epidemiology , Genes, Bacterial , Humans , Shigella/classification , Shigella/genetics , Shigella/pathogenicity , Virulence/genetics
16.
Infect Immun ; 82(9): 3612-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24935977

ABSTRACT

Elongation factor P (EF-P) is a universally conserved bacterial translation factor. In many bacteria, EF-P is posttranslationally modified by PoxA, which covalently attaches a ß-lysine to a conserved lysine residue of EF-P. Here we show that both EF-P and PoxA are necessary for virulence of the human diarrheal pathogen Shigella flexneri. Loss of either EF-P or PoxA leads to an impaired ability of S. flexneri to invade epithelial cells and form plaques in an epithelial cell monolayer. Proteomic analysis of efp and poxA deletion mutants revealed decreased levels of several virulence effector proteins, including IpaA, -B, and -C and IcsA. Additionally, mRNA levels of virB and virF, which encode master virulence regulators, were decreased in the efp mutant. The reduction in virF transcription was at least partially due to decreased levels of CpxA, which activates virF through the response regulator CpxR. The role of CpxAR in reduced synthesis of VirF and its downstream effectors was indicated by restoration of invasion when a mutation resulting in constitutively activated CpxR was introduced into the efp mutant. Thus, modified EF-P is required for appropriate synthesis of proteins involved in the virulence of this bacterial pathogen.


Subject(s)
Peptide Elongation Factors/genetics , Shigella flexneri/genetics , Virulence Factors/genetics , Virulence/genetics , Bacterial Proteins/genetics , Cells, Cultured , Dysentery, Bacillary/genetics , Dysentery, Bacillary/microbiology , Epithelial Cells/microbiology , Gene Expression Regulation, Bacterial/genetics , Humans , Proteomics , Sequence Deletion/genetics , Transcription, Genetic/genetics
17.
J Cell Sci ; 125(Pt 3): 703-13, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22389405

ABSTRACT

An understanding of the signaling mechanism(s) that regulate the differential expression of gastric mucin MUC5AC in colonic epithelial cells would contribute significantly to investigations of its role in colonic mucosa infected with the bacterial pathogen Shigella dysenteriae. Here we show that S. dysenteriae-Sinduced expression of interleukin-1ß upregulates MUC2 expression and the differential expression of MUC5AC. Differential expression of MUC5AC involves crosstalk between interleukin-1ß and Akt, whereby the trefoil factor family peptide TFF3 activates Akt by phosphorylation of EGFR. TFF3 also downregulates E-cadherin expression, causing accumulation of ß-catenin in the cytosol. Phosphorylation of GSK-3ß (inactivated) by activated Akt inhibits ubiquitylation of ß-catenin, leading to its nuclear translocation, which then induces the expression of MUC5AC and cyclin D1. Accumulation of cyclin D1 alters the cell cycle, promoting cell survival and proliferation. Human colon HT29MTX cells, which overexpress MUC5AC, were resistant to adherence and invasion of S. dysenteriae when compared with other mucin-secreting HT29 cell types. Thus, during infection with S. dysenteriae, crosstalk between interleukin-1ß and Akt wired by TFF3 induces expression of MUC5AC in colonic epithelial cells. Differentially expressed gastric MUC5AC aids in mucosal clearance of S. dysenteriae, inhibiting adherence and invasion of the pathogen to colonic epithelial cells, which protects the host.


Subject(s)
Dysentery, Bacillary/immunology , Dysentery, Bacillary/metabolism , Interleukin-1beta/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mucin 5AC/metabolism , Peptides/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Shigella dysenteriae , Apoproteins , Bacterial Adhesion , Cadherins/genetics , Cadherins/metabolism , Cell Proliferation , Chromones/pharmacology , Dysentery, Bacillary/genetics , Dysentery, Bacillary/pathology , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HT29 Cells , Humans , Immunity, Mucosal , Interleukin-1beta/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Models, Biological , Morpholines/pharmacology , Mucin 5AC/genetics , Mucin-2/genetics , Mucin-2/metabolism , Peptides/antagonists & inhibitors , Peptides/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Shigella dysenteriae/pathogenicity , Trefoil Factor-3 , Up-Regulation , beta Catenin/metabolism
18.
PLoS Pathog ; 8(8): e1002809, 2012.
Article in English | MEDLINE | ID: mdl-22912573

ABSTRACT

The activation of host cells by interferon gamma (IFNγ) is essential for inhibiting the intracellular replication of most microbial pathogens. Although significant advances have been made in identifying IFNγ-dependent host factors that suppress intracellular bacteria, little is known about how IFNγ enables cells to recognize, or restrict, the growth of pathogens that replicate in the host cytoplasm. The replication of the cytosolic bacterial pathogen Shigella flexneri is significantly inhibited in IFNγ-stimulated cells, however the specific mechanisms that mediate this inhibition have remained elusive. We found that S. flexneri efficiently invades IFNγ-activated mouse embryonic fibroblasts (MEFs) and escapes from the vacuole, suggesting that IFNγ acts by blocking S. flexneri replication in the cytosol. This restriction on cytosolic growth was dependent on interferon regulatory factor 1 (IRF1), an IFNγ-inducible transcription factor capable of inducing IFNγ-mediated cell-autonomous immunity. To identify host factors that restrict S. flexneri growth, we used whole genome microarrays to identify mammalian genes whose expression in S. flexneri-infected cells is controlled by IFNγ and IRF1. Among the genes we identified was the pattern recognition receptor (PRR) retanoic acid-inducible gene I (RIG-I), a cytoplasmic sensor of foreign RNA that had not been previously known to play a role in S. flexneri infection. We found that RIG-I and its downstream signaling adaptor mitochondrial antiviral signaling protein (MAVS)--but not cytosolic Nod-like receptors (NLRs)--are critically important for IFNγ-mediated S. flexneri growth restriction. The recently described RNA polymerase III pathway, which transcribes foreign cytosolic DNA into the RIG-I ligand 5'-triphosphate RNA, appeared to be involved in this restriction. The finding that RIG-I responds to S. flexneri infection during the IFNγ response extends the range of PRRs that are capable of recognizing this bacterium. Additionally, these findings expand our understanding of how IFNγ recognizes, and ultimately restricts, bacterial pathogens within host cells.


Subject(s)
Cytoplasm/immunology , DEAD-box RNA Helicases/immunology , Dysentery, Bacillary/immunology , Immunity, Innate , Interferon-gamma/immunology , Shigella flexneri/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Cells, Cultured , Cytoplasm/genetics , Cytoplasm/microbiology , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Dysentery, Bacillary/genetics , Embryo, Mammalian/immunology , Embryo, Mammalian/microbiology , Fibroblasts/immunology , Fibroblasts/microbiology , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/immunology , Interferon-gamma/genetics , Mice , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology
19.
Dis Model Mech ; 17(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38131137

ABSTRACT

Shigella flexneri is a human-adapted pathovar of Escherichia coli that can invade the intestinal epithelium, causing inflammation and bacillary dysentery. Although an important human pathogen, the host response to S. flexneri has not been fully described. Zebrafish larvae represent a valuable model for studying human infections in vivo. Here, we use a Shigella-zebrafish infection model to generate mRNA expression profiles of host response to Shigella infection at the whole-animal level. Immune response-related processes dominate the signature of early Shigella infection (6 h post-infection). Consistent with its clearance from the host, the signature of late Shigella infection (24 h post-infection) is significantly changed, and only a small set of immune-related genes remain differentially expressed, including acod1 and gpr84. Using mutant lines generated by ENU, CRISPR mutagenesis and F0 crispants, we show that acod1- and gpr84-deficient larvae are more susceptible to Shigella infection. Together, these results highlight the power of zebrafish to model infection by bacterial pathogens and reveal the mRNA expression of the early (acutely infected) and late (clearing) host response to Shigella infection.


Subject(s)
Dysentery, Bacillary , Animals , Humans , Dysentery, Bacillary/genetics , Shigella flexneri/genetics , Shigella flexneri/metabolism , Zebrafish/genetics , Zebrafish/microbiology , Inflammation/microbiology , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Biomed Res Int ; 2024: 5554208, 2024.
Article in English | MEDLINE | ID: mdl-38595330

ABSTRACT

Shigella stands as a major contributor to bacterial dysentery worldwide scale, particularly in developing countries with inadequate sanitation and hygiene. The emergence of multidrug-resistant strains exacerbates the challenge of treating Shigella infections, particularly in regions where access to healthcare and alternative antibiotics is limited. Therefore, investigations on how bacteria evade antibiotics and eventually develop resistance could open new avenues for research to develop novel therapeutics. The aim of this study was to analyze whole genome sequence (WGS) of human pathogenic Shigella spp. to elucidate the antibiotic resistance genes (ARGs) and their mechanism of resistance, gene-drug interactions, protein-protein interactions, and functional pathways to screen potential therapeutic candidate(s). We comprehensively analyzed 45 WGS of Shigella, including S. flexneri (n = 17), S. dysenteriae (n = 14), S. boydii (n = 11), and S. sonnei (n = 13), through different bioinformatics tools. Evolutionary phylogenetic analysis showed three distinct clades among the circulating strains of Shigella worldwide, with less genomic diversity. In this study, 2,146 ARGs were predicted in 45 genomes (average 47.69 ARGs/genome), of which only 91 ARGs were found to be shared across the genomes. Majority of these ARGs conferred their resistance through antibiotic efflux pump (51.0%) followed by antibiotic target alteration (23%) and antibiotic target replacement (18%). We identified 13 hub proteins, of which four proteins (e.g., tolC, acrR, mdtA, and gyrA) were detected as potential hub proteins to be associated with antibiotic efflux pump and target alteration mechanisms. These hub proteins were significantly (p < 0.05) enriched in biological process, molecular function, and cellular components. Therefore, the finding of this study suggests that human pathogenic Shigella strains harbored a wide range of ARGs that confer resistance through antibiotic efflux pumps and antibiotic target modification mechanisms, which must be taken into account to devise and formulate treatment strategy against this pathogen. Moreover, the identified hub proteins could be exploited to design and develop novel therapeutics against MDR pathogens like Shigella.


Subject(s)
Dysentery, Bacillary , Shigella , Humans , Phylogeny , Drug Resistance, Bacterial/genetics , Microbial Sensitivity Tests , Shigella/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Dysentery, Bacillary/drug therapy , Dysentery, Bacillary/genetics , Dysentery, Bacillary/microbiology , Shigella flexneri
SELECTION OF CITATIONS
SEARCH DETAIL