Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38834194

ABSTRACT

Vinculin is a cytoskeletal linker strengthening cell adhesion. The Shigella IpaA invasion effector binds to vinculin to promote vinculin supra-activation associated with head-domain-mediated oligomerization. Our study investigates the impact of mutations of vinculin D1D2 subdomains' residues predicted to interact with IpaA VBS3. These mutations affected the rate of D1D2 trimer formation with distinct effects on monomer disappearance, consistent with structural modeling of a closed and open D1D2 conformer induced by IpaA. Notably, mutations targeting the closed D1D2 conformer significantly reduced Shigella invasion of host cells as opposed to mutations targeting the open D1D2 conformer and later stages of vinculin head-domain oligomerization. In contrast, all mutations affected the formation of focal adhesions (FAs), supporting the involvement of vinculin supra-activation in this process. Our findings suggest that IpaA-induced vinculin supra-activation primarily reinforces matrix adhesion in infected cells, rather than promoting bacterial invasion. Consistently, shear stress studies pointed to a key role for IpaA-induced vinculin supra-activation in accelerating and strengthening cell-matrix adhesion.


Subject(s)
Cell Adhesion , Focal Adhesions , Vinculin , Vinculin/metabolism , Vinculin/genetics , Humans , Focal Adhesions/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Mutation , Host-Pathogen Interactions , HeLa Cells , Protein Binding , Shigella/metabolism , Shigella/genetics , Antigens, Bacterial/metabolism , Antigens, Bacterial/genetics , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/metabolism
2.
Nat Commun ; 15(1): 3890, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719850

ABSTRACT

Shigella flexneri is a Gram-negative bacterium causing severe bloody dysentery. Its pathogenesis is largely dictated by a plasmid-encoded type III secretion system (T3SS) and its associated effectors. Among these, the effector OspG has been shown to bind to the ubiquitin conjugation machinery (E2~Ub) to activate its kinase activity. However, the cellular targets of OspG remain elusive despite years of extensive efforts. Here we show by unbiased phosphoproteomics that a major target of OspG is CAND1, a regulatory protein controlling the assembly of cullin-RING ubiquitin ligases (CRLs). CAND1 phosphorylation weakens its interaction with cullins, which is expected to impact a large panel of CRL E3s. Indeed, global ubiquitome profiling reveals marked changes in the ubiquitination landscape when OspG is introduced. Notably, OspG promotes ubiquitination of a class of cytoskeletal proteins called septins, thereby inhibiting formation of cage-like structures encircling cytosolic bacteria. Overall, we demonstrate that pathogens have evolved an elaborate strategy to modulate host ubiquitin signaling to evade septin-cage entrapment.


Subject(s)
Bacterial Proteins , Septins , Shigella flexneri , Signal Transduction , Ubiquitin , Ubiquitination , Shigella flexneri/metabolism , Shigella flexneri/pathogenicity , Septins/metabolism , Septins/genetics , Humans , Ubiquitin/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Phosphorylation , Host-Pathogen Interactions , HeLa Cells , Cullin Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , HEK293 Cells , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/metabolism
3.
Front Cell Infect Microbiol ; 12: 1012533, 2022.
Article in English | MEDLINE | ID: mdl-36389142

ABSTRACT

Shigella, the causative agent of bacillary dysentery, subvert cytoskeletal and trafficking processes to invade and replicate in epithelial cells using an arsenal of bacterial effectors translocated through a type III secretion system. Here, we review the various roles of the type III effector IpgD, initially characterized as phosphatidylinositol 4,5 bisphosphate (PI4,5P2) 4-phosphatase. By decreasing PI4,5P2 levels, IpgD triggers the disassembly of cortical actin filaments required for bacterial invasion and cell migration. PI5P produced by IpgD further stimulates signaling pathways regulating cell survival, macropinosome formation, endosomal trafficking and dampening of immune responses. Recently, IpgD was also found to exhibit phosphotransferase activity leading to PI3,4P2 synthesis adding a new flavor to this multipotent bacterial enzyme. The substrate of IpgD, PI4,5P2 is also the main substrate hydrolyzed by endogenous phospholipases C to produce inositoltriphosphate (InsP3), a major Ca2+ second messenger. Hence, beyond the repertoire of effects associated with the direct diversion of phoshoinositides, IpgD indirectly down-regulates InsP3-mediated Ca2+ release by limiting InsP3 production. Furthermore, IpgD controls the intracellular lifestyle of Shigella promoting Rab8/11 -dependent recruitment of the exocyst at macropinosomes to remove damaged vacuolar membrane remnants and promote bacterial cytosolic escape. IpgD thus emerges as a key bacterial effector for the remodeling of host cell membranes.


Subject(s)
Dysentery, Bacillary , Shigella , Humans , Phosphatidylinositols/metabolism , Shigella flexneri/metabolism , Dysentery, Bacillary/metabolism , Phosphoric Monoester Hydrolases/metabolism
4.
Nat Commun ; 13(1): 4494, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35918380

ABSTRACT

Enzymes from pathogens often modulate host protein post-translational modifications (PTMs), facilitating survival and proliferation of pathogens. Shigella virulence factors IpaJ and IcsB induce proteolytic cleavage and lysine fatty acylation on host proteins, which cause Golgi stress and suppress innate immunity, respectively. However, it is unknown whether host enzymes could reverse such modifications introduced by pathogens' virulence factors to suppress pathogenesis. Herein, we report that SIRT2, a potent lysine defatty-acylase, is upregulated by the transcription factor CREB3 under Golgi stress induced by Shigella infection. SIRT2 in turn removes the lysine fatty acylation introduced by Shigella virulence factor IcsB to enhance host innate immunity. SIRT2 knockout mice are more susceptible to Shigella infection than wildtype mice, demonstrating the importance of SIRT2 to counteract Shigella infection.


Subject(s)
Dysentery, Bacillary , Shigella , Acylation , Animals , Dysentery, Bacillary/metabolism , Golgi Apparatus/metabolism , Lysine/metabolism , Mice , Sirtuin 2/genetics , Sirtuin 2/metabolism , Virulence Factors/metabolism
5.
mBio ; 13(3): e0094422, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35652591

ABSTRACT

Polymorphonuclear neutrophils (PMN) are recruited to the gastrointestinal mucosa in response to inflammation, injury, and infection. Here, we report the development and the characterization of an ex vivo tissue coculture model consisting of human primary intestinal enteroid monolayers and PMN, and a mechanistic interrogation of PMN-epithelial cell interaction and response to Shigella, a primary cause of childhood dysentery. Cellular adaptation and tissue integration, barrier function, PMN phenotypic and functional attributes, and innate immune responses were examined. PMN within the enteroid monolayers acquired a distinct activated/migratory phenotype that was influenced by direct epithelial cell contact as well as by molecular signals. Seeded on the basal side of the intestinal monolayer, PMN were intercalated within the epithelial cells and moved paracellularly toward the apical side. Cocultured PMN also increased basal secretion of interleukin 8 (IL-8). Shigella added to the apical surface of the monolayers evoked additional PMN phenotypic adaptations, including increased expression of cell surface markers associated with chemotaxis and cell degranulation (CD47, CD66b, and CD88). Apical Shigella infection triggered rapid transmigration of PMN to the luminal side, neutrophil extracellular trap (NET) formation, and bacterial phagocytosis and killing. Shigella infection modulated cytokine production in the coculture; apical monocyte chemoattractant protein (MCP-1), tumor necrosis factor alpha (TNF-α), and basolateral IL-8 production were downregulated, while basolateral IL-6 secretion was increased. We demonstrated, for the first time, PMN phenotypic adaptation and mobilization and coordinated epithelial cell-PMN innate response upon Shigella infection in the human intestinal environment. The enteroid monolayer-PMN coculture represents a technical innovation for mechanistic interrogation of gastrointestinal physiology, host-microbe interaction, innate immunity, and evaluation of preventive/therapeutic tools. IMPORTANCE Studies of mucosal immunity and microbial host cell interaction have traditionally relied on animal models and in vitro tissue culture using immortalized cancer cell lines, which yield nonphysiological and often unreliable results. Herein, we report the development and characterization of an ex vivo enteroid-PMN coculture consisting of normal human intestinal epithelium and a mechanistic interrogation of PMN and epithelial cell interaction and function in the context of Shigella infection. We demonstrated tissue-driven phenotypic and functional adaptation of PMN and a coordinated epithelial cell and PMN response to Shigella, a primary cause of dysentery in young children in the developing world.


Subject(s)
Dysentery, Bacillary , Shigella , Animals , Cells, Cultured , Child, Preschool , Coculture Techniques , Dysentery, Bacillary/metabolism , Epithelial Cells/metabolism , Humans , Interleukin-8 , Intestinal Mucosa/metabolism , Neutrophils , Shigella/metabolism
6.
Methods Mol Biol ; 2523: 113-131, 2022.
Article in English | MEDLINE | ID: mdl-35759194

ABSTRACT

Intracellular bacterial pathogens have evolved a plethora of strategies to invade eukaryotic cells. By manipulating host signaling pathways, in particular vesicular trafficking, these microbes subvert host functions to promote their internalization and to establish an intracellular niche. During these events, host endomembrane compartments are dynamically reorganized. Shigella flexneri, the causative agent of bacillary dysentery, recruits components of the host recycling pathway and the exocyst of non-phagocytic enterocytes in the vicinity of its entry site to facilitate its access to the host cytosol. These factors are either dynamically tethered to in situ formed macropinosomes or to the bacteria-containing vacuole itself. The underlying interactions cannot readily be monitored as individual bacterial infection events take place without synchronicity using cellular infection models. Therefore, time-resolved screens by fluorescence microscopy represent a powerful tool for the study of host subversion. Such screens can be performed with libraries of fluorescently tagged host factors. Using the cytosolic pathogenic agent Shigella flexneri as a model, we provide detailed protocols for such medium-to-high throughput multidimensional imaging screening of the dynamic host-pathogen cross talk. Our workflow is designed to be easily adapted for the study of different host factor libraries and different pathogen models.


Subject(s)
Dysentery, Bacillary , Vacuoles , Bacterial Proteins/metabolism , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/pathology , Endosomes/metabolism , Host-Pathogen Interactions , Humans , Microscopy, Fluorescence , Shigella flexneri , Vacuoles/metabolism
7.
Sci Rep ; 12(1): 251, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997046

ABSTRACT

Shigellosis is characterized as diarrheal disease that causes a high mortality rate especially in children, elderly and immunocompromised patients. More recently, the World Health Organization advised safe vaccine designing against shigellosis due to the emergence of Shigella dysenteriae resistant strains. Therefore, the aim of this study is to identify novel drug targets as well as the design of the potential vaccine candidates and chimeric vaccine models against Shigella dysenteriae. A computational based Reverse Vaccinology along with subtractive genomics analysis is one of the robust approaches used for the prioritization of drug targets and vaccine candidates through direct screening of genome sequence assemblies. Herein, a successfully designed peptide-based novel highly antigenic chimeric vaccine candidate against Shigella dysenteriae sd197 strain is proposed. The study resulted in six epitopes from outer membrane WP_000188255.1 (Fe (3+) dicitrate transport protein FecA) that ultimately leads to the construction of twelve vaccine models. Moreover, V9 construct was found to be highly immunogenic, non-toxic, non-allergenic, highly antigenic, and most stable in terms of molecular docking and simulation studies against six HLAs and TLRS/MD complex. So far, this protein and multiepitope have never been characterized as vaccine targets against Shigella dysenteriae. The current study proposed that V9 could be a significant vaccine candidate against shigellosis and to ascertain that further experiments may be applied by the scientific community focused on shigellosis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Vaccines/pharmacology , Drug Design , Dysentery, Bacillary/prevention & control , Shigella dysenteriae/drug effects , Vaccine Development/methods , Vaccinology/methods , Animals , Antigens, Bacterial/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/microbiology , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Computer-Aided Design , Dysentery, Bacillary/immunology , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/microbiology , Epitopes , Host-Pathogen Interactions , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Network Pharmacology , Shigella dysenteriae/immunology , Shigella dysenteriae/pathogenicity , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/microbiology
8.
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
9.
Cell Host Microbe ; 29(10): 1521-1530.e10, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34492225

ABSTRACT

The pore-forming protein gasdermin D (GSDMD) executes lytic cell death called pyroptosis to eliminate the replicative niche of intracellular pathogens. Evolution favors pathogens that circumvent this host defense mechanism. Here, we show that the Shigella ubiquitin ligase IpaH7.8 functions as an inhibitor of GSDMD. Shigella is an enteroinvasive bacterium that causes hemorrhagic gastroenteritis in primates, but not rodents. IpaH7.8 contributes to species specificity by ubiquitinating human, but not mouse, GSDMD and targeting it for proteasomal degradation. Accordingly, infection of human epithelial cells with IpaH7.8-deficient Shigella flexneri results in increased GSDMD-dependent cell death compared with wild type. Consistent with pyroptosis contributing to murine disease resistance, eliminating GSDMD from NLRC4-deficient mice, which are already sensitized to oral infection with Shigella flexneri, leads to further enhanced bacterial replication and increased disease severity. This work highlights a species-specific pathogen arms race focused on maintenance of host cell viability.


Subject(s)
Bacterial Proteins/metabolism , Dysentery, Bacillary/metabolism , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Shigella flexneri/enzymology , Ubiquitin-Protein Ligases/metabolism , Animals , Bacterial Proteins/genetics , Dysentery, Bacillary/genetics , Dysentery, Bacillary/microbiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Host-Pathogen Interactions , Humans , Mice , Mice, Knockout , Phosphate-Binding Proteins/genetics , Pore Forming Cytotoxic Proteins/genetics , Proteolysis , Shigella flexneri/genetics , Shigella flexneri/physiology , Ubiquitin-Protein Ligases/genetics
10.
PLoS Pathog ; 17(9): e1009932, 2021 09.
Article in English | MEDLINE | ID: mdl-34499700

ABSTRACT

Many bacterial pathogens require a type 3 secretion system (T3SS) to establish a niche. Host contact activates bacterial T3SS assembly of a translocon pore in the host plasma membrane. Following pore formation, the T3SS docks onto the translocon pore. Docking establishes a continuous passage that enables the translocation of virulence proteins, effectors, into the host cytosol. Here we investigate the contribution of actin polymerization to T3SS-mediated translocation. Using the T3SS model organism Shigella flexneri, we show that actin polymerization is required for assembling the translocon pore in an open conformation, thereby enabling effector translocation. Opening of the pore channel is associated with a conformational change to the pore, which is dependent upon actin polymerization and a coiled-coil domain in the pore protein IpaC. Analysis of an IpaC mutant that is defective in ruffle formation shows that actin polymerization-dependent pore opening is distinct from the previously described actin polymerization-dependent ruffles that are required for bacterial internalization. Moreover, actin polymerization is not required for other pore functions, including docking or pore protein insertion into the plasma membrane. Thus, activation of the T3SS is a multilayered process in which host signals are sensed by the translocon pore leading to the activation of effector translocation.


Subject(s)
Actins/metabolism , Host-Pathogen Interactions/physiology , Shigella flexneri/pathogenicity , Type III Secretion Systems/metabolism , Virulence/physiology , Dysentery, Bacillary/metabolism , HeLa Cells , Humans , Polymerization , Shigella flexneri/metabolism
11.
Nat Commun ; 12(1): 4511, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34301939

ABSTRACT

Septins are cytoskeletal proteins that assemble into hetero-oligomeric complexes and sense micron-scale membrane curvature. During infection with Shigella flexneri, an invasive enteropathogen, septins restrict actin tail formation by entrapping bacteria in cage-like structures. Here, we reconstitute septin cages in vitro using purified recombinant septin complexes (SEPT2-SEPT6-SEPT7), and study how these recognize bacterial cells and assemble on their surface. We show that septin complexes recognize the pole of growing Shigella cells. An amphipathic helix domain in human SEPT6 enables septins to sense positively curved membranes and entrap bacterial cells. Shigella strains lacking lipopolysaccharide components are more efficiently entrapped in septin cages. Finally, cryo-electron tomography of in vitro cages reveals how septins assemble as filaments on the bacterial cell surface.


Subject(s)
Actins/metabolism , Dysentery, Bacillary/metabolism , Recombinant Proteins/metabolism , Septins/metabolism , Shigella flexneri/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dysentery, Bacillary/microbiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Lipopolysaccharides/metabolism , Microscopy, Fluorescence , Mutation , Protein Binding , Septins/genetics , Shigella flexneri/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Elife ; 102021 06 04.
Article in English | MEDLINE | ID: mdl-34085925

ABSTRACT

Defective autophagy is strongly associated with chronic inflammation. Loss-of-function of the core autophagy gene Atg16l1 increases risk for Crohn's disease in part by enhancing innate immunity through myeloid cells such as macrophages. However, autophagy is also recognized as a mechanism for clearance of certain intracellular pathogens. These divergent observations prompted a re-evaluation of ATG16L1 in innate antimicrobial immunity. In this study, we found that loss of Atg16l1 in myeloid cells enhanced the killing of virulent Shigella flexneri (S.flexneri), a clinically relevant enteric bacterium that resides within the cytosol by escaping from membrane-bound compartments. Quantitative multiplexed proteomics of murine bone marrow-derived macrophages revealed that ATG16L1 deficiency significantly upregulated proteins involved in the glutathione-mediated antioxidant response to compensate for elevated oxidative stress, which simultaneously promoted S.flexneri killing. Consistent with this, myeloid-specific deletion of Atg16l1 in mice accelerated bacterial clearance in vitro and in vivo. Pharmacological induction of oxidative stress through suppression of cysteine import enhanced microbial clearance by macrophages. Conversely, antioxidant treatment of macrophages permitted S.flexneri proliferation. These findings demonstrate that control of oxidative stress by ATG16L1 and autophagy regulates antimicrobial immunity against intracellular pathogens.


Subject(s)
Autophagy-Related Proteins/deficiency , Autophagy , Dysentery, Bacillary/microbiology , Immunity, Innate , Macrophages/microbiology , Oxidative Stress , Proteome , Proteomics , Shigella flexneri/pathogenicity , Animals , Autophagy-Related Proteins/genetics , Cells, Cultured , Disease Models, Animal , Dysentery, Bacillary/immunology , Dysentery, Bacillary/metabolism , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microbial Viability , Shigella flexneri/immunology , Shigella flexneri/metabolism , Virulence
13.
Cell Mol Immunol ; 18(4): 878-888, 2021 04.
Article in English | MEDLINE | ID: mdl-33731917

ABSTRACT

Protein N-myristoylation is an important fatty acylation catalyzed by N-myristoyltransferases (NMTs), which are ubiquitous enzymes in eukaryotes. Specifically, attachment of a myristoyl group is vital for proteins participating in various biological functions, including signal transduction, cellular localization, and oncogenesis. Recent studies have revealed unexpected mechanisms indicating that protein N-myristoylation is involved in host defense against microbial and viral infections. In this review, we describe the current understanding of protein N-myristoylation (mainly focusing on myristoyl switches) and summarize its crucial roles in regulating innate immune responses, including TLR4-dependent inflammatory responses and demyristoylation-induced innate immunosuppression during Shigella flexneri infection. Furthermore, we examine the role of myristoylation in viral assembly, intracellular host interactions, and viral spread during human immunodeficiency virus-1 (HIV-1) infection. Deeper insight into the relationship between protein N-myristoylation and innate immunity might enable us to clarify the pathogenesis of certain infectious diseases and better harness protein N-myristoylation for new therapeutics.


Subject(s)
Dysentery, Bacillary/immunology , HIV Infections/immunology , Immunity, Innate , Myristic Acid/chemistry , Protein Processing, Post-Translational , Proteins/chemistry , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/microbiology , HIV/immunology , HIV Infections/metabolism , HIV Infections/microbiology , Humans , Shigella flexneri/immunology
14.
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
15.
Toxins (Basel) ; 12(5)2020 05 22.
Article in English | MEDLINE | ID: mdl-32456007

ABSTRACT

Shiga toxin (STx) produced by Shigella and closely related Shiga toxin 1 and 2 (STx1 and STx2) synthesized by Shiga toxin-producing Escherichia coli (STEC) are bacterial AB5 toxins. All three toxins target kidney cells and may cause life-threatening renal disease. While Shigella infections can be treated with antibiotics, resistance is increasing. Moreover, antibiotic therapy is contraindicated for STEC, and there are no definitive treatments for STEC-induced disease. To exert cellular toxicity, STx, STx1, and STx2 must undergo retrograde trafficking to reach their cytosolic target, ribosomes. Direct transport from early endosomes to the Golgi apparatus is an essential step that allows the toxins to bypass degradative late endosomes and lysosomes. The essentiality of this transport step also makes it an ideal target for the development of small-molecule inhibitors of toxin trafficking as potential therapeutics. Here, we review the recent advances in understanding the molecular mechanisms of the early endosome-to-Golgi transport of STx, STx1, and STx2, as well as the development of small-molecule inhibitors of toxin trafficking that act at the endosome/Golgi interface.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Dysentery, Bacillary/drug therapy , Endosomes/metabolism , Escherichia coli Infections/drug therapy , Golgi Apparatus/metabolism , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/drug effects , Shigella/drug effects , Animals , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/microbiology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Humans , Molecular Targeted Therapy , Protein Transport , Shiga-Toxigenic Escherichia coli/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Shigella/metabolism , Shigella/pathogenicity
16.
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
17.
mBio ; 10(6)2019 12 17.
Article in English | MEDLINE | ID: mdl-31848280

ABSTRACT

Two Shigella species, Shigella flexneri and Shigella sonnei, cause approximately 90% of bacterial dysentery worldwide. While S. flexneri is the dominant species in low-income countries, S. sonnei causes the majority of infections in middle- and high-income countries. S. flexneri is a prototypic cytosolic bacterium; once intracellular, it rapidly escapes the phagocytic vacuole and causes pyroptosis of macrophages, which is important for pathogenesis and bacterial spread. In contrast, little is known about the invasion, vacuole escape, and induction of pyroptosis during S. sonnei infection of macrophages. We demonstrate here that S. sonnei causes substantially less pyroptosis in human primary monocyte-derived macrophages and THP1 cells. This is due to reduced bacterial uptake and lower relative vacuole escape, which results in fewer cytosolic S. sonnei and hence reduced activation of caspase-1 inflammasomes. Mechanistically, the O-antigen (O-Ag), which in S. sonnei is contained in both the lipopolysaccharide and the capsule, was responsible for reduced uptake and the type 3 secretion system (T3SS) was required for vacuole escape. Our findings suggest that S. sonnei has adapted to an extracellular lifestyle by incorporating multiple layers of O-Ag onto its surface compared to other Shigella species.IMPORTANCE Diarrheal disease remains the second leading cause of death in children under five. Shigella remains a significant cause of diarrheal disease with two species, S. flexneri and S. sonnei, causing the majority of infections. S. flexneri are well known to cause cell death in macrophages, which contributes to the inflammatory nature of Shigella diarrhea. Here, we demonstrate that S. sonnei causes less cell death than S. flexneri due to a reduced number of bacteria present in the cell cytosol. We identify the O-Ag polysaccharide which, uniquely among Shigella spp., is present in two forms on the bacterial cell surface as the bacterial factor responsible. Our data indicate that S. sonnei differs from S. flexneri in key aspects of infection and that more attention should be given to characterization of S. sonnei infection.


Subject(s)
Dysentery, Bacillary/metabolism , Dysentery, Bacillary/microbiology , Host-Pathogen Interactions/immunology , Inflammasomes/metabolism , O Antigens/immunology , Shigella sonnei/physiology , Vacuoles/metabolism , Endocytosis/immunology , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Models, Biological , Pyroptosis/immunology , Type III Secretion Systems
18.
PLoS Pathog ; 15(11): e1007928, 2019 11.
Article in English | MEDLINE | ID: mdl-31725799

ABSTRACT

Type 3 secretion systems (T3SSs) are conserved bacterial nanomachines that inject virulence proteins (effectors) into eukaryotic cells during infection. Due to their ability to inject heterologous proteins into human cells, these systems are being developed as therapeutic delivery devices. The T3SS assembles a translocon pore in the plasma membrane and then docks onto the pore. Docking activates effector secretion through the pore and into the host cytosol. Here, using Shigella flexneri, a model pathogen for the study of type 3 secretion, we determined the molecular mechanisms by which host intermediate filaments trigger docking and enable effector secretion. We show that the interaction of intermediate filaments with the translocon pore protein IpaC changed the pore's conformation in a manner that was required for docking. Intermediate filaments repositioned residues of the Shigella pore protein IpaC that are located on the surface of the pore and in the pore channel. Restricting these conformational changes blocked docking in an intermediate filament-dependent manner. These data demonstrate that a host-induced conformational change to the pore enables T3SS docking and effector secretion, providing new mechanistic insight into the regulation of type 3 secretion.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Cell Membrane/metabolism , Dysentery, Bacillary/metabolism , Host-Pathogen Interactions , Shigella flexneri/metabolism , Type III Secretion Systems/metabolism , Bacterial Adhesion , Dysentery, Bacillary/microbiology , HeLa Cells , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Transport , Shigella flexneri/pathogenicity , Type III Secretion Systems/chemistry , Virulence
19.
Nano Lett ; 19(10): 7365-7369, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31538793

ABSTRACT

Biomembranes are hard to compress laterally, and membrane area compressibility has not been associated with biological processes. Using X-ray surface scattering, we observed that bacterial Shiga toxin compresses lipid packing in a gel phase monolayer upon binding to its cellular receptor, the glycolipid Gb3. This toxin-induced reorganization of lipid packing reached beyond the immediate membrane patch that the protein was bound to, and linkers separating the Gb3 carbohydrate and ceramide moieties modulated the toxin's capacity to compress the membrane. Within a natural membrane, asymmetric compression of the toxin-bound leaflet could provide a mechanism to initiate narrow membrane bending, as observed upon toxin entry into cells. Such lipid compression and long-range membrane reorganization by glycolipid-binding proteins represent novel concepts in membrane biology that have direct implications for the construction of endocytic pits in clathrin-independent endocytosis.


Subject(s)
Cell Membrane/metabolism , Phosphatidylethanolamines/metabolism , Shiga Toxin/metabolism , Shigella dysenteriae/metabolism , Trihexosylceramides/metabolism , Dysentery, Bacillary/metabolism , Endocytosis , Humans , Models, Molecular
20.
Nat Microbiol ; 4(11): 2001-2009, 2019 11.
Article in English | MEDLINE | ID: mdl-31383999

ABSTRACT

Pathogenic enterobacteria face various oxygen (O2) levels during intestinal colonization from the O2-deprived lumen to oxygenated tissues. Using Shigella flexneri as a model, we have previously demonstrated that epithelium invasion is promoted by O2 in a type III secretion system-dependent manner. However, subsequent pathogen adaptation to tissue oxygenation modulation remained unknown. Assessing single-cell distribution, together with tissue oxygenation, we demonstrate here that the colonic mucosa O2 is actively depleted by S. flexneri aerobic respiration-and not host neutrophils-during infection, leading to the formation of hypoxic foci of infection. This process is promoted by type III secretion system inactivation in infected tissues, favouring colonizers over explorers. We identify the molecular mechanisms supporting infectious hypoxia induction, and demonstrate here how enteropathogens optimize their colonization capacity in relation to their ability to manipulate tissue oxygenation during infection.


Subject(s)
Dysentery, Bacillary/metabolism , Intestinal Mucosa/microbiology , Oxygen/metabolism , Shigella flexneri/pathogenicity , Animals , Cell Hypoxia , Disease Models, Animal , Dysentery, Bacillary/microbiology , Female , Guinea Pigs , Hep G2 Cells , Humans , Intestinal Mucosa/metabolism , Rabbits , Shigella flexneri/metabolism , Type III Secretion Systems/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...