1
|
Ding D, Guo J, Sun H, Yang J. Modulation of host Rab GTPases by Salmonella: mechanisms of immune evasion and intracellular replication. Mol Biol Rep 2025; 52:440. [PMID: 40304792 DOI: 10.1007/s11033-025-10547-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025]
Abstract
Salmonella is one of the major pathogens responsible for foodborne illnesses worldwide, characterized by diverse serotypes and a broad host range. As an intracellular bacterium, Salmonella invades host cells and establishes a protected niche known as the Salmonella-containing vacuoles (SCVs), which provide a suitable environment for intracellular survival. Rab GTPases, key regulators of intracellular membrane trafficking, play a crucial role in the biogenesis and dynamics of SCVs. Through its type III secretion systems (T3SSs), Salmonella delivers a repertoire of effector proteins into host cells, which modulate the activity of Rab GTPases and alter membrane trafficking to facilitate SCVs formation and maintenance. This review summarizes recent advances in understanding how Salmonella effectors manipulate Rab GTPases to promote intracellular survival and evade host innate immune responses.
Collapse
Affiliation(s)
- Dandan Ding
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, 730030, China
| | - Jiayin Guo
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, 730030, China
| | - Hui Sun
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, 730030, China
| | - Jing Yang
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, 730030, China.
| |
Collapse
|
2
|
Zhao H, Zhang X, Zhang N, Zhu L, Lian H. The interplay between Salmonella and host: Mechanisms and strategies for bacterial survival. CELL INSIGHT 2025; 4:100237. [PMID: 40177681 PMCID: PMC11964643 DOI: 10.1016/j.cellin.2025.100237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 04/05/2025]
Abstract
Salmonella, an intracellular pathogen, infects both humans and animals, causing diverse diseases such as gastroenteritis and enteric fever. The Salmonella type III secretion system (T3SS), encoded within its pathogenicity islands (SPIs), is critical for bacterial virulence by directly delivering multiple effectors into eukaryotic host cells. Salmonella utilizes these effectors to facilitate its survival and replication within the host through modulating cytoskeletal dynamics, inflammatory responses, the biogenesis of Salmonella-containing vacuole (SCV), and host cell survival. Moreover, these effectors also interfere with immune responses via inhibiting innate immunity or antigen presentation. In this review, we summarize the current progress in the survival strategies employed by Salmonella and the molecular mechanisms underlying its interactions with the host. Understanding the interplay between Salmonella and host can enhance our knowledge of the bacterium's pathogenic processes and provide new insights into how it manipulates host cellular physiological activities to ensure its survival.
Collapse
Affiliation(s)
- Hongyu Zhao
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, Hubei, China
| | - Xinyue Zhang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, Hubei, China
| | - Ningning Zhang
- Yale Stem Cell Center, New Haven, CT, 06520, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06510, USA
- Yale Cooperative Center of Excellence in Hematology, New Haven, CT, 12208, USA
| | - Li Zhu
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Huan Lian
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, Hubei, China
| |
Collapse
|
3
|
Zhang Y, Chen S, Chen G, Zhou L, Zhou G, Yu X, Yuan L, Deng W, Wang Z, Li J, Tu Y, Zhang D, li Y, Sammad A, Zhu X, Yin K. The Type III Secretion System (T3SS) of Escherichia Coli Promotes Atherosclerosis in Type 2 Diabetes Mellitus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413296. [PMID: 39807021 PMCID: PMC12005784 DOI: 10.1002/advs.202413296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Large-scale studies indicate a strong relationship between the gut microbiome, type 2 diabetes mellitus (T2DM), and atherosclerotic cardiovascular disease (ASCVD). Here, a higher abundance of the type III secretion system (T3SS) virulence factors of Enterobacteriaceae/Escherichia-Shigella in patients with T2DM-related-ASCVD, which correlates with their atherosclerotic stenosis is reported. Overexpression of T3SS via Citrobacter rodentium (CR) infection in Apoe-/- T2DM mice exacerbated atherosclerotic lesion formation and increased gut permeability. Non-targeted metabolomic and proteomic analysis of mouse serum showed that T3SS caused abnormal glycerophospholipid metabolism in mice. Proteomics, RNA sequencing, and functional analyses showed that T3SS induced ferroptosis in intestinal epithelial cells, partly due to increased expression of ferritin heavy chains (FTH1). This findings first demonstrated that T3SS increases ferroptosis in intestinal epithelial cells, via disrupting the intestinal barrier and upregulation of phosphatidylcholine, thereby exacerbating T2DM-related ASCVD.
Collapse
Affiliation(s)
- Yao‐Yuan Zhang
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangdong Provincial Key Laboratory of Drug Non‐Clinical Evaluation and ResearchGuangzhou510515China
| | - Song‐Tao Chen
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Gang Chen
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Le Zhou
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541199China
| | - Guo‐Liang Zhou
- Department of CardiologyThe Second Affiliated Hospital of Guilin Medical UniversityGuilin541199China
| | - Xin‐Yuan Yu
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Long Yuan
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Wei‐Qian Deng
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Zhen‐Bo Wang
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Jing Li
- Department of Imaging DiagnosisZhujiang Hospital of Southern Medical UniversityGuangzhou510515China
| | - Yi‐Fu Tu
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Da‐Wei Zhang
- Group on the Molecular and Cell Biology of Lipids and Department of PediatricsFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaT6G 2R3Canada
| | - Yuan li
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541199China
| | - Abdul Sammad
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Xiao Zhu
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangdong Provincial Key Laboratory of Drug Non‐Clinical Evaluation and ResearchGuangzhou510515China
- Guangzhou Key Laboratory of Metabolic remodeling and Precise Prevention and Control of DiabetesGuangzhou510515China
| | - Kai Yin
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangdong Provincial Key Laboratory of Drug Non‐Clinical Evaluation and ResearchGuangzhou510515China
- Guangzhou Key Laboratory of Metabolic remodeling and Precise Prevention and Control of DiabetesGuangzhou510515China
- Guangxi Clinical Research Center for Diabetes and Metabolic DiseasesThe Second Affiliated Hospital of Guilin Medical UniversityGuilin541199China
| |
Collapse
|
4
|
Meng Y, Zhang Q, Xu M, Ding K, Yu Z, Li J. Pyroptosis regulation by Salmonella effectors. Front Immunol 2024; 15:1464858. [PMID: 39507539 PMCID: PMC11538000 DOI: 10.3389/fimmu.2024.1464858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The genus Salmonella contains the most common foodborne pathogens frequently isolated from food-producing animals and is responsible for zoonotic infections in humans and animals. Salmonella infection in humans and animals can cause intestinal damage, resulting in intestinal inflammation and disruption of intestinal homeostasis more severe cases can lead to bacteremia. Pyroptosis, a proinflammatory form of programmed cell death, is involved in many disease processes. Inflammasomes, pyroptosis, along with their respective signaling cascades, are instrumental in the preservation of intestinal homeostasis. In recent years, with the in-depth study of pyroptosis, our comprehension of the virulence factors and effector proteins in Salmonella has reached an extensive level, a deficit persists in our knowledge regarding the intrinsic pathogenic mechanisms about pyroptosis, necessitating a continued pursuit of understanding and investigation. In this review, we discuss the occurrence of pyroptosis induced by Salmonella effectors to provide new ideas for elucidating the regulatory mechanisms through which Salmonella virulence factors and effector proteins trigger pyroptosis could pave the way for novel concepts and strategies in the clinical prevention of Salmonella infections and the treatment of associated diseases.
Collapse
Affiliation(s)
- Yuan Meng
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang, Henan, China
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, Henan, China
| | - Qianjin Zhang
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang, Henan, China
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, Henan, China
| | - Mengen Xu
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang, Henan, China
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, Henan, China
| | - Ke Ding
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang, Henan, China
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, Henan, China
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Zuhua Yu
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang, Henan, China
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, Henan, China
| | - Jing Li
- College of Animal Science and Technology/Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang, Henan, China
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, Henan, China
| |
Collapse
|
5
|
Ray J, Sapp DG, Fairn GD. Phosphatidylinositol 3,4-bisphosphate: Out of the shadows and into the spotlight. Curr Opin Cell Biol 2024; 88:102372. [PMID: 38776601 DOI: 10.1016/j.ceb.2024.102372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/15/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Phosphoinositide 3-kinases regulate many cellular functions, including migration, growth, proliferation, and cell survival. Early studies equated the inhibition of Class I PI3Ks with loss of; phosphatidylinositol 3,4,5-trisphosphate (PIP3), but over time, it was realised that these; treatments also depleted phosphatidylinositol 3,4-bisphosphate (PI(3,4)P2). In recent years, the; use of better tools and an improved understanding of its metabolism have allowed for the; identification of specific roles of PI(3,4)P2. This includes the production of PI(3,4)P2 and the; activation of its effector Akt2 in response to growth factor signalling. In contrast, a lysosomal pool of PI(3,4)P2 is a negative regulator of mTORC1 during growth factor deprivation. A growing body of literature also demonstrates that PI(3,4)P2 controls many dynamic plasmalemmal processes. The significance of PI(3,4)P2 in cell biology is increasingly evident.
Collapse
Affiliation(s)
- Jayatee Ray
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - David G Sapp
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gregory D Fairn
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
6
|
Oke MT, D’Costa VM. Functional Divergence of the Paralog Salmonella Effector Proteins SopD and SopD2 and Their Contributions to Infection. Int J Mol Sci 2024; 25:4191. [PMID: 38673776 PMCID: PMC11050076 DOI: 10.3390/ijms25084191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Salmonella enterica is a leading cause of bacterial food-borne illness in humans and is responsible for millions of cases annually. A critical strategy for the survival of this pathogen is the translocation of bacterial virulence factors termed effectors into host cells, which primarily function via protein-protein interactions with host proteins. The Salmonella genome encodes several paralogous effectors believed to have arisen from duplication events throughout the course of evolution. These paralogs can share structural similarities and enzymatic activities but have also demonstrated divergence in host cell targets or interaction partners and contributions to the intracellular lifecycle of Salmonella. The paralog effectors SopD and SopD2 share 63% amino acid sequence similarity and extensive structural homology yet have demonstrated divergence in secretion kinetics, intracellular localization, host targets, and roles in infection. SopD and SopD2 target host Rab GTPases, which represent critical regulators of intracellular trafficking that mediate diverse cellular functions. While SopD and SopD2 both manipulate Rab function, these paralogs display differences in Rab specificity, and the effectors have also evolved multiple mechanisms of action for GTPase manipulation. Here, we highlight this intriguing pair of paralog effectors in the context of host-pathogen interactions and discuss how this research has presented valuable insights into effector evolution.
Collapse
Affiliation(s)
- Mosopefoluwa T. Oke
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Vanessa M. D’Costa
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
7
|
Kirchenwitz M, Halfen J, von Peinen K, Prettin S, Kollasser J, Zur Lage S, Blankenfeldt W, Brakebusch C, Rottner K, Steffen A, Stradal TEB. RhoB promotes Salmonella survival by regulating autophagy. Eur J Cell Biol 2023; 102:151358. [PMID: 37703749 DOI: 10.1016/j.ejcb.2023.151358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/05/2023] [Accepted: 08/29/2023] [Indexed: 09/15/2023] Open
Abstract
Salmonella enterica serovar Typhimurium manipulates cellular Rho GTPases for host cell invasion by effector protein translocation via the Type III Secretion System (T3SS). The two Guanine nucleotide exchange (GEF) mimicking factors SopE and -E2 and the inositol phosphate phosphatase (PiPase) SopB activate the Rho GTPases Rac1, Cdc42 and RhoA, thereby mediating bacterial invasion. S. Typhimurium lacking these three effector proteins are largely invasion-defective. Type III secretion is crucial for both early and later phases of the intracellular life of S. Typhimurium. Here we investigated whether and how the small GTPase RhoB, known to localize on endomembrane vesicles and at the invasion site of S. Typhimurium, contributes to bacterial invasion and to subsequent steps relevant for S. Typhimurium lifestyle. We show that RhoB is significantly upregulated within hours of Salmonella infection. This effect depends on the presence of the bacterial effector SopB, but does not require its phosphatase activity. Our data reveal that SopB and RhoB bind to each other, and that RhoB localizes on early phagosomes of intracellular S. Typhimurium. Whereas both SopB and RhoB promote intracellular survival of Salmonella, RhoB is specifically required for Salmonella-induced upregulation of autophagy. Finally, in the absence of RhoB, vacuolar escape and cytosolic hyper-replication of S. Typhimurium is diminished. Our findings thus uncover a role for RhoB in Salmonella-induced autophagy, which supports intracellular survival of the bacterium and is promoted through a positive feedback loop by the Salmonella effector SopB.
Collapse
Affiliation(s)
- Marco Kirchenwitz
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Jessica Halfen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Kristin von Peinen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Silvia Prettin
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Jana Kollasser
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Susanne Zur Lage
- Department Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Wulf Blankenfeldt
- Department Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Cord Brakebusch
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Klemens Rottner
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany; Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
| | - Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany.
| |
Collapse
|
8
|
Chaussé AM, Roche SM, Moroldo M, Hennequet-Antier C, Holbert S, Kempf F, Barilleau E, Trotereau J, Velge P. Epithelial cell invasion by salmonella typhimurium induces modulation of genes controlled by aryl hydrocarbon receptor signaling and involved in extracellular matrix biogenesis. Virulence 2023; 14:2158663. [PMID: 36600181 PMCID: PMC9828750 DOI: 10.1080/21505594.2022.2158663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Salmonella is the only bacterium able to enter a host cell by the two known mechanisms: trigger and zipper. The trigger mechanism relies on the injection of bacterial effectors into the host cell through the Salmonella type III secretion system 1. In the zipper mechanism, mediated by the invasins Rck and PagN, the bacterium takes advantage of a cellular receptor for invasion. This study describes the transcriptomic reprogramming of the IEC-6 intestinal epithelial cell line to Salmonella Typhimurium strains that invaded cells by a trigger, a zipper, or both mechanisms. Using S. Typhimurium strains invalidated for one or other entry mechanism, we have shown that IEC-6 cells could support both entries. Comparison of the gene expression profiles of exposed cells showed that irrespective of the mechanism used for entry, the transcriptomic reprogramming of the cell was nearly the same. On the other hand, when gene expression was compared between cells unexposed or exposed to the bacterium, the transcriptomic reprogramming of exposed cells was significantly different. It is particularly interesting to note the modulation of expression of numerous target genes of the aryl hydrocarbon receptor showing that this transcription factor was activated by S. Typhimurium infection. Numerous genes associated with the extracellular matrix were also modified. This was confirmed at the protein level by western-blotting showing a dramatic modification in some extracellular matrix proteins. Analysis of a selected set of modulated genes showed that the expression of the majority of these genes was modulated during the intracellular life of S. Typhimurium.
Collapse
Affiliation(s)
| | | | - Marco Moroldo
- INRAE, AgroParisTech, Université Paris Saclay, Jouy-en-Josas, France
| | | | | | | | | | | | - Philippe Velge
- INRAE, ISP, Université de Tours, Nouzilly, France,CONTACT Philippe Velge
| |
Collapse
|
9
|
Xie LY, Xu YB, Ding XQ, Liang S, Li DL, Fu AK, Zhan XA. Itaconic acid and dimethyl itaconate exert antibacterial activity in carbon-enriched environments through the TCA cycle. Biomed Pharmacother 2023; 167:115487. [PMID: 37713987 DOI: 10.1016/j.biopha.2023.115487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
Itaconic acid (IA), a metabolite generated by the tricarboxylic acid (TCA) cycle in eukaryotic immune cells, and its derivative dimethyl itaconate (DI) exert antibacterial functions in intracellular environments. Previous studies suggested that IA and DI only inhibit bacterial growth in carbon-limited environments; however, whether IA and DI maintain antibacterial activity in carbon-enriched environments remains unknown. Here, IA and DI inhibited the bacteria with minimum inhibitory concentrations of 24.02 mM and 39.52 mM, respectively, in a carbon-enriched environment. The reduced bacterial pathogenicity was reflected in cell membrane integrity, motility, biofilm formation, AI-2/luxS, and virulence. Mechanistically, succinate dehydrogenase (SDH) activity and fumaric acid levels decreased in the IA and DI treatments, while isocitrate lyase (ICL) activity was upregulated. Inhibited TCA circulation was also observed through untargeted metabolomics. In addition, energy-related aspartate metabolism and lysine degradation were suppressed. In summary, these results indicated that IA and DI reduced bacterial pathogenicity while exerting antibacterial functions by inhibiting TCA circulation. This study enriches knowledge on the inhibition of bacteria by IA and DI in a carbon-mixed environment, suggesting an alternative method for treating bacterial infections by immune metabolites.
Collapse
Affiliation(s)
- L Y Xie
- Key Laboratory of Animal Nutrition and Feed in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Y B Xu
- Key Laboratory of Animal Nutrition and Feed in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - X Q Ding
- Key Laboratory of Animal Nutrition and Feed in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - S Liang
- Key Laboratory of Animal Nutrition and Feed in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - D L Li
- Key Laboratory of Animal Nutrition and Feed in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - A K Fu
- Key Laboratory of Animal Nutrition and Feed in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - X A Zhan
- Key Laboratory of Animal Nutrition and Feed in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
10
|
Meng K, Zhu P, Shi L, Li S. Determination of the Salmonella intracellular lifestyle by the diversified interaction of Type III secretion system effectors and host GTPases. WIREs Mech Dis 2023; 15:e1587. [PMID: 36250298 DOI: 10.1002/wsbm.1587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 11/06/2022]
Abstract
Intracellular bacteria have developed sophisticated strategies to subvert the host endomembrane system to establish a stable replication niche. Small GTPases are critical players in regulating each step of membrane trafficking events, such as vesicle biogenesis, cargo transport, tethering, and fusion events. Salmonella is a widely studied facultative intracellular bacteria. Salmonella delivers several virulence proteins, termed effectors, to regulate GTPase dynamics and subvert host trafficking for their benefit. In this review, we summarize an updated and systematic understanding of the interactions between bacterial effectors and host GTPases in determining the intracellular lifestyle of Salmonella. This article is categorized under: Infectious Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Kun Meng
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ping Zhu
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Liuliu Shi
- School of Basic Medical Science, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shan Li
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
11
|
Lange Y, Tabei SMA, Steck TL. A basic model for the association of ligands with membrane cholesterol: application to cytolysin binding. J Lipid Res 2023; 64:100344. [PMID: 36791915 PMCID: PMC10119614 DOI: 10.1016/j.jlr.2023.100344] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Almost all the cholesterol in cellular membranes is associated with phospholipids in simple stoichiometric complexes. This limits the binding of sterol ligands such as filipin and Perfringolysin O (PFO) to a small fraction of the total. We offer a simple mathematical model that characterizes this complexity. It posits that the cholesterol accessible to ligands has two forms: active cholesterol, which is that not complexed with phospholipids; and extractable cholesterol, that which ligands can capture competitively from the phospholipid complexes. Simulations based on the model match published data for the association of PFO oligomers with liposomes, plasma membranes and the isolated endoplasmic reticulum. The model shows how the binding of a probe greatly underestimates cholesterol abundance when its affinity for the sterol is so weak that it competes poorly with the membrane phospholipids. Two examples are the under-staining of plasma membranes by filipin and the failure of domain D4 of PFO to label their cytoplasmic leaflets. Conversely, the exaggerated staining of endolysosomes suggests that their cholesterol, being uncomplexed, is readily available. The model is also applicable to the association of cholesterol with intrinsic membrane proteins. For example, it supports the hypothesis that the sharp threshold in the regulation of homeostatic ER proteins by cholesterol derives from the cooperativity of their binding to the sterol weakly held by the phospholipid. § Thus, the model explicates the complexity inherent in the binding of ligands like PFO and filipin to the small accessible fraction of membrane cholesterol.
Collapse
Affiliation(s)
- Yvonne Lange
- 1Department of Pathology, Rush University Medical Center, Chicago, Il 60612, USA.
| | - S M Ali Tabei
- Department of Physics, University of Northern Iowa, Cedar Falls, Iowa 50614, USA
| | - Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Il 60637, USA
| |
Collapse
|
12
|
Salmonella effector SopB reorganizes cytoskeletal vimentin to maintain replication vacuoles for efficient infection. Nat Commun 2023; 14:478. [PMID: 36717589 PMCID: PMC9885066 DOI: 10.1038/s41467-023-36123-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
A variety of intracellular bacteria modulate the host cytoskeleton to establish subcellular niches for replication. However, the role of intermediate filaments, which are crucial for mechanical strength and resilience of the cell, and in bacterial vacuole preservation remains unclear. Here, we show that Salmonella effector SopB reorganizes the vimentin network to form cage-like structures that surround Salmonella-containing vacuoles (SCVs). Genetic removal of vimentin markedly disrupts SCV organization, significantly reduces bacterial replication and cell death. Mechanistically, SopB uses its N-terminal Cdc42-binding domain to interact with and activate Cdc42 GTPase, which in turn recruits vimentin around SCVs. A high-content imaging-based screening identified that MEK1/2 inhibition led to vimentin dispersion. Our work therefore elucidates the signaling axis SopB-Cdc42-MEK1/2 as mobilizing host vimentin to maintain concrete SCVs and identifies a mechanism contributing to Salmonella replication. Importantly, Trametinib, a clinically-approved MEK1/2 inhibitor identified in the screen, displayed significant anti-infection efficacy against Salmonella both in vitro and in vivo, and may provide a therapeutic option for treating drug-tolerant salmonellosis.
Collapse
|
13
|
Bezada-Quintana SG, Carcelén-Cáceres FD, López-Guerra S, Guevara-Vásquez JE. Respuesta histomorfométrica de la mucosa del intestino delgado en cuyes (Cavia porcellus) de engorde desafiados con Salmonella enterica var. Typhimurium. REVISTA CIENTÍFICA DE LA FACULTAD DE CIENCIAS VETERINARIAS 2023. [DOI: 10.52973/rcfcv-e33203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Con el objetivo de evaluar la respuesta histomorfométrica de la mucosa del intestino delgado en cuyes (Cavia porcellus) desafiados con Salmonella enterica var. Typhimurium (SeVT) (2×106 UFC·0,5mL-1 por cuy) se tomaron 40 cuyes machos de 15 días (d) de edad, distribuidos aleatoriamente en cuatro tratamientos SeVT; 1.- cuyes que recibieron dieta base (DB) y se desafiaron con SeVT (T1); 2.- cuyes que recibieron DB con 50 ppm de zinc bacitracina y se desafiaron con SeVT (T2); 3.- cuyes que recibieron DB con 50 ppm de zinc bacitracina, sin desafío (T3) y 4.- cuyes que recibieron únicamente DB, sin desafío (T4, Grupo Control). Cada T estuvo conformado por 10 repeticiones. Al finalizar la fase experimental en la 8va semana (sem) se colectaron muestras de las secciones duodeno, yeyuno e íleon y se procesaron con hematoxilina y eosina (H-E) para el análisis histomorfométrico. Los parámetros evaluados fueron longitud de la vellosidad (LV), ancho de la vellosidad (AV), profundidad de la cripta de Lieberkühn (PC) y la relación longitud/cripta (LV/PC). Los datos se analizaron mediante análisis de varianza y la prueba de Tukey. Los resultados (promedio ± DE) fueron significativamente diferentes a P<0,05 en todos los T indicando vellosidades reducidas en longitud, ancho y con una relación LV/PC, menor de 2:1 en el T1, observando vellosidades hasta 40 % más largas en duodeno, 39 % en yeyuno y 55 % en íleon en los cuyes que no fueron desafiados con SeVT, concluyendo que existe un efecto negativo de este enteropatógeno sobre la histomorfometría de las vellosidades del intestino delgado en esta especie.
Collapse
Affiliation(s)
- Sandra Gracia Bezada-Quintana
- Universidad Nacional Mayor de San Marcos, Facultad de Medicina Veterinaria, Laboratorio de Bioquímica, Nutrición y Alimentación Animal. Lima, Perú
| | - Fernando Demetrio Carcelén-Cáceres
- Universidad Nacional Mayor de San Marcos, Facultad de Medicina Veterinaria, Laboratorio de Bioquímica, Nutrición y Alimentación Animal. Lima, Perú
| | - Sofía López-Guerra
- Universidad Nacional Mayor de San Marcos, Facultad de Medicina Veterinaria, Laboratorio de Bioquímica, Nutrición y Alimentación Animal. Lima, Perú
| | - Jorge Ernesto Guevara-Vásquez
- Universidad Nacional Mayor de San Marcos, Facultad de Química e Ingeniería Química, Escuela Profesional de Ingeniería Agroindustrial. Lima, Perú
| |
Collapse
|
14
|
Martins Morasi R, Zimbardi da Silva A, Thais Alves Dantas S, Faganello C, Cristina Bastos Juliano L, Lúcia Mores Rall V, Ribeiro Tiba-Casas M, Pantoja JC, Ferreira Amarante A, Cristina Cirone Silva N. Overview of antimicrobial resistance and virulence factors in Salmonella spp. isolated in the last two decades from chicken in Brazil. Food Res Int 2022; 162:111955. [DOI: 10.1016/j.foodres.2022.111955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/29/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022]
|
15
|
Capitani N, Baldari CT. The Immunological Synapse: An Emerging Target for Immune Evasion by Bacterial Pathogens. Front Immunol 2022; 13:943344. [PMID: 35911720 PMCID: PMC9325968 DOI: 10.3389/fimmu.2022.943344] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Similar to other pathogens, bacteria have developed during their evolution a variety of mechanisms to overcome both innate and acquired immunity, accounting for their ability to cause disease or chronic infections. The mechanisms exploited for this critical function act by targeting conserved structures or pathways that regulate the host immune response. A strategic potential target is the immunological synapse (IS), a highly specialized structure that forms at the interface between antigen presenting cells (APC) and T lymphocytes and is required for the establishment of an effective T cell response to the infectious agent and for the development of long-lasting T cell memory. While a variety of bacterial pathogens are known to impair or subvert cellular processes essential for antigen processing and presentation, on which IS assembly depends, it is only recently that the possibility that IS may be a direct target of bacterial virulence factors has been considered. Emerging evidence strongly supports this notion, highlighting IS targeting as a powerful, novel means of immune evasion by bacterial pathogens. In this review we will present a brief overview of the mechanisms used by bacteria to affect IS assembly by targeting APCs. We will then summarize what has emerged from the current handful of studies that have addressed the direct impact of bacterial virulence factors on IS assembly in T cells and, based on the strategic cellular processes targeted by these factors in other cell types, highlight potential IS-related vulnerabilities that could be exploited by these pathogens to evade T cell mediated immunity.
Collapse
Affiliation(s)
- Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | | |
Collapse
|
16
|
Cohen H, Adani B, Cohen E, Piscon B, Azriel S, Desai P, Bähre H, McClelland M, Rahav G, Gal-Mor O. The ancestral stringent response potentiator, DksA has been adapted throughout Salmonella evolution to orchestrate the expression of metabolic, motility, and virulence pathways. Gut Microbes 2022; 14:1997294. [PMID: 34923900 PMCID: PMC8726615 DOI: 10.1080/19490976.2021.1997294] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
DksA is a conserved RNA polymerase-binding protein known to play a key role in the stringent response of proteobacteria species, including many gastrointestinal pathogens. Here, we used RNA-sequencing of Escherichia coli, Salmonella bongori and Salmonella enterica serovar Typhimurium, together with phenotypic comparison to study changes in the DksA regulon, during Salmonella evolution. Comparative RNA-sequencing showed that under non-starved conditions, DksA controls the expression of 25%, 15%, and 20% of the E. coli, S. bongori, and S. enterica genes, respectively, indicating that DksA is a pleiotropic regulator, expanding its role beyond the canonical stringent response. We demonstrate that DksA is required for the growth of these three enteric bacteria species in minimal medium and controls the expression of the TCA cycle, glycolysis, pyrimidine biosynthesis, and quorum sensing. Interestingly, at multiple steps during Salmonella evolution, the type I fimbriae and various virulence genes encoded within SPIs 1, 2, 4, 5, and 11 have been transcriptionally integrated under the ancestral DksA regulon. Consequently, we show that DksA is necessary for host cells invasion by S. Typhimurium and S. bongori and for intracellular survival of S. Typhimurium in bone marrow-derived macrophages (BMDM). Moreover, we demonstrate regulatory inversion of the conserved motility-chemotaxis regulon by DksA, which acts as a negative regulator in E. coli, but activates this pathway in S. bongori and S. enterica. Overall, this study demonstrates the regulatory assimilation of multiple horizontally acquired virulence genes under the DksA regulon and provides new insights into the evolution of virulence genes regulation in Salmonella spp.
Collapse
Affiliation(s)
- Helit Cohen
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel
| | - Boaz Adani
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel
| | - Emiliano Cohen
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel
| | - Bar Piscon
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Shalhevet Azriel
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel
| | - Prerak Desai
- Janssen Research & Development, LLC, Raritan, New Jersey, USA,Department of Microbiology and Molecular Genetics, University of California, Irvine, California, USA
| | - Heike Bähre
- Hannover Medical School, Research Core Unit Metabolomics, Hannover, Germany
| | - Michael McClelland
- Department of Microbiology and Molecular Genetics, University of California, Irvine, California, USA
| | - Galia Rahav
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ohad Gal-Mor
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel,Contact Ohad Gal-Mor The Infectious Diseases Research Laboratory Sheba Medical Cente, Tel-Hashomer, Israel
| |
Collapse
|
17
|
Boddy KC, Zhu H, D'Costa VM, Xu C, Beyrakhova K, Cygler M, Grinstein S, Coyaud E, Laurent EMN, St-Germain J, Raught B, Brumell JH. Salmonella effector SopD promotes plasma membrane scission by inhibiting Rab10. Nat Commun 2021; 12:4707. [PMID: 34349110 PMCID: PMC8339009 DOI: 10.1038/s41467-021-24983-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/16/2021] [Indexed: 12/17/2022] Open
Abstract
Salmonella utilizes translocated virulence proteins (termed effectors) to promote host cell invasion. The effector SopD contributes to invasion by promoting scission of the plasma membrane, generating Salmonella-containing vacuoles. SopD is expressed in all Salmonella lineages and plays important roles in animal models of infection, but its host cell targets are unknown. Here we show that SopD can bind to and inhibit the small GTPase Rab10, through a C-terminal GTPase activating protein (GAP) domain. During infection, Rab10 and its effectors MICAL-L1 and EHBP1 are recruited to invasion sites. By inhibiting Rab10, SopD promotes removal of Rab10 and recruitment of Dynamin-2 to drive scission of the plasma membrane. Together, our study uncovers an important role for Rab10 in regulating plasma membrane scission and identifies the mechanism used by a bacterial pathogen to manipulate this function during infection.
Collapse
Affiliation(s)
- Kirsten C Boddy
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Hongxian Zhu
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Vanessa M D'Costa
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.,Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada
| | - Caishuang Xu
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ksenia Beyrakhova
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Miroslaw Cygler
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Sergio Grinstein
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Etienne Coyaud
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Estelle M N Laurent
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jonathan St-Germain
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - John H Brumell
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada. .,SickKids IBD Centre, Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
18
|
Molecular determinants of peaceful coexistence versus invasiveness of non-Typhoidal Salmonella: Implications in long-term side-effects. Mol Aspects Med 2021; 81:100997. [PMID: 34311996 DOI: 10.1016/j.mam.2021.100997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/02/2021] [Accepted: 07/16/2021] [Indexed: 01/28/2023]
Abstract
The genus Salmonella represents a wide range of strains including Typhoidal and Non-Typhoidal Salmonella (NTS) isolates that exhibit illnesses of varied pathophysiologies. The more frequent NTS ensues a self-limiting enterocolitis with rare occasions of bacteremia or systemic infections. These self-limiting Salmonella strains are capable of subverting and dampening the host immune system to achieve a more prolonged survival inside the host system thus leading to chronic manifestations. Notably, emergence of new invasive NTS isolates known as invasive Non-Typhoidal Salmonella (iNTS) have worsened the disease burden significantly in some parts of the world. NTS strains adapt to attain persister phenotype intracellularly and cause relapsing infections. These chronic infections, in susceptible hosts, are also capable of causing diseases like IBS, IBD, reactive arthritis, gallbladder cancer and colorectal cancer. The present understanding of molecular mechanism of how these chronic infections are manifested is quite limited. The current work is an effort to review the prevailing knowledge emanating from a large volume of research focusing on various forms of NTS infections including those that cause localized, systemic and persistent disease. The review will further dwell into the understanding of how this pathogen contributes to the associated long term sequelae.
Collapse
|
19
|
ARHGEF26 enhances Salmonella invasion and inflammation in cells and mice. PLoS Pathog 2021; 17:e1009713. [PMID: 34242364 PMCID: PMC8294491 DOI: 10.1371/journal.ppat.1009713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 07/21/2021] [Accepted: 06/14/2021] [Indexed: 11/25/2022] Open
Abstract
Salmonella hijack host machinery in order to invade cells and establish infection. While considerable work has described the role of host proteins in invasion, much less is known regarding how natural variation in these invasion-associated host proteins affects Salmonella pathogenesis. Here we leveraged a candidate cellular GWAS screen to identify natural genetic variation in the ARHGEF26 (Rho Guanine Nucleotide Exchange Factor 26) gene that renders lymphoblastoid cells susceptible to Salmonella Typhi and Typhimurium invasion. Experimental follow-up redefined ARHGEF26’s role in Salmonella epithelial cell infection. Specifically, we identified complex serovar-by-host interactions whereby ARHGEF26 stimulation of S. Typhi and S. Typhimurium invasion into host cells varied in magnitude and effector-dependence based on host cell type. While ARHGEF26 regulated SopB- and SopE-mediated S. Typhi (but not S. Typhimurium) infection of HeLa cells, the largest effect of ARHGEF26 was observed with S. Typhimurium in polarized MDCK cells through a SopB- and SopE2-independent mechanism. In both cell types, knockdown of the ARHGEF26-associated protein DLG1 resulted in a similar phenotype and serovar specificity. Importantly, we show that ARHGEF26 plays a critical role in S. Typhimurium pathogenesis by contributing to bacterial burden in the enteric fever murine model, as well as inflammation in the colitis infection model. In the enteric fever model, SopB and SopE2 are required for the effects of Arhgef26 deletion on bacterial burden, and the impact of sopB and sopE2 deletion in turn required ARHGEF26. In contrast, SopB and SopE2 were not required for the impacts of Arhgef26 deletion on colitis. A role for ARHGEF26 on inflammation was also seen in cells, as knockdown reduced IL-8 production in HeLa cells. Together, these data reveal pleiotropic roles for ARHGEF26 during infection and highlight that many of the interactions that occur during infection that are thought to be well understood likely have underappreciated complexity. During infection, Salmonella manipulates host cells into engulfing the bacteria and establishing an intracellular niche. While many studies have identified genes involved in different stages of this Salmonella invasion process, few studies have examined how differences between human hosts contribute to infection susceptibility. Here we leveraged a candidate genetic screen to identify natural genetic variation in the human ARHGEF26 gene that correlates with Salmonella invasion. Springboarding from this result, we experimentally tested and redefined ARHGEF26’s role in Salmonella invasion, discovered a new role for ARHGEF26 in regulating inflammation during Salmonella disease, and demonstrated the relevance of these findings in mouse models. Building on how ARHGEF26 functions in other contexts, we implicated two ARHGEF26-interacting host proteins as contributors to Salmonella pathobiology. Collectively, these results identify a potential source of inter-person diversity in susceptibility to Salmonella disease and expand our molecular understanding of Salmonella infection to include a multifaceted role for ARHGEF26. They further identify important future directions in understanding how Salmonella recruit and manipulate ARHGEF26 as well as how ARHGEF26 is able to drive Salmonella-beneficial processes.
Collapse
|
20
|
Engelhardt S, Trutzenberg A, Hückelhoven R. Regulation and Functions of ROP GTPases in Plant-Microbe Interactions. Cells 2020; 9:E2016. [PMID: 32887298 PMCID: PMC7565977 DOI: 10.3390/cells9092016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023] Open
Abstract
Rho proteins of plants (ROPs) form a specific clade of Rho GTPases, which are involved in either plant immunity or susceptibility to diseases. They are intensively studied in grass host plants, in which ROPs are signaling hubs downstream of both cell surface immune receptor kinases and intracellular nucleotide-binding leucine-rich repeat receptors, which activate major branches of plant immune signaling. Additionally, invasive fungal pathogens may co-opt the function of ROPs for manipulation of the cytoskeleton, cell invasion and host cell developmental reprogramming, which promote pathogenic colonization. Strikingly, mammalian bacterial pathogens also initiate both effector-triggered susceptibility for cell invasion and effector-triggered immunity via Rho GTPases. In this review, we summarize central concepts of Rho signaling in disease and immunity of plants and briefly compare them to important findings in the mammalian research field. We focus on Rho activation, downstream signaling and cellular reorganization under control of Rho proteins involved in disease progression and pathogen resistance.
Collapse
Affiliation(s)
| | | | - Ralph Hückelhoven
- Phytopathology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Emil-Ramann-Straße 2, 85354 Freising, Germany; (S.E.); (A.T.)
| |
Collapse
|
21
|
Fattinger SA, Böck D, Di Martino ML, Deuring S, Samperio Ventayol P, Ek V, Furter M, Kreibich S, Bosia F, Müller-Hauser AA, Nguyen BD, Rohde M, Pilhofer M, Hardt WD, Sellin ME. Salmonella Typhimurium discreet-invasion of the murine gut absorptive epithelium. PLoS Pathog 2020; 16:e1008503. [PMID: 32365138 PMCID: PMC7224572 DOI: 10.1371/journal.ppat.1008503] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/14/2020] [Accepted: 03/26/2020] [Indexed: 01/15/2023] Open
Abstract
Salmonella enterica serovar Typhimurium (S.Tm) infections of cultured cell lines have given rise to the ruffle model for epithelial cell invasion. According to this model, the Type-Three-Secretion-System-1 (TTSS-1) effectors SopB, SopE and SopE2 drive an explosive actin nucleation cascade, resulting in large lamellipodia- and filopodia-containing ruffles and cooperative S.Tm uptake. However, cell line experiments poorly recapitulate many of the cell and tissue features encountered in the host’s gut mucosa. Here, we employed bacterial genetics and multiple imaging modalities to compare S.Tm invasion of cultured epithelial cell lines and the gut absorptive epithelium in vivo in mice. In contrast to the prevailing ruffle-model, we find that absorptive epithelial cell entry in the mouse gut occurs through “discreet-invasion”. This distinct entry mode requires the conserved TTSS-1 effector SipA, involves modest elongation of local microvilli in the absence of expansive ruffles, and does not favor cooperative invasion. Discreet-invasion preferentially targets apicolateral hot spots at cell–cell junctions and shows strong dependence on local cell neighborhood. This proof-of-principle evidence challenges the current model for how S.Tm can enter gut absorptive epithelial cells in their intact in vivo context. Bacterial pathogens can use secreted effector molecules to drive entry into host cells. Studies of the intestinal pathogen S.Tm have been central to uncover the mechanistic basis for the entry process. More than two decades of research have resulted in a detailed model for how S.Tm invades gut epithelial cells through effector triggering of large Rho-GTPase-dependent actin ruffles. However, the evidence for this model comes predominantly from studies in cultured cell lines. These experimental systems lack many of the architectural and signaling features of the intact gut epithelium. Our study surprisingly reveals that in the intact mouse gut, S.Tm invades absorptive epithelial cells through a process that does not require the Rho-GTPase-activating effectors and can proceed in the absence of the prototypical ruffling response. Instead, S.Tm exploits another effector, SipA, to sneak in through discreet entry structures close to cell–cell junctions. Our results challenge the current model for S.Tm epithelial cell entry and emphasizes the need of taking a physiological host cell context into account when studying bacterium–host cell interactions.
Collapse
Affiliation(s)
- Stefan A. Fattinger
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Desirée Böck
- Institute of Molecular Biology & Biophysics, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Maria Letizia Di Martino
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sabrina Deuring
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Pilar Samperio Ventayol
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Viktor Ek
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Markus Furter
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Saskia Kreibich
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Francesco Bosia
- Institute of Molecular Biology & Biophysics, Department of Biology, ETH Zürich, Zürich, Switzerland
| | | | - Bidong D. Nguyen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Martin Pilhofer
- Institute of Molecular Biology & Biophysics, Department of Biology, ETH Zürich, Zürich, Switzerland
- * E-mail: (MP); (WDH); (MES)
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
- * E-mail: (MP); (WDH); (MES)
| | - Mikael E. Sellin
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- * E-mail: (MP); (WDH); (MES)
| |
Collapse
|
22
|
Azimi T, Zamirnasta M, Sani MA, Soltan Dallal MM, Nasser A. Molecular Mechanisms of Salmonella Effector Proteins: A Comprehensive Review. Infect Drug Resist 2020; 13:11-26. [PMID: 32021316 PMCID: PMC6954085 DOI: 10.2147/idr.s230604] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/20/2019] [Indexed: 12/27/2022] Open
Abstract
Salmonella can be categorized into many serotypes, which are specific to known hosts or broadhosts. It makes no difference which one of the serotypes would penetrate the gastrointestinal tract because they all face similar obstacles such as mucus and microbiome. However, following their penetration, some species remain in the gastrointestinal tract; yet, others spread to another organ like gallbladder. Salmonella is required to alter the immune response to sustain its intracellular life. Changing the host response requires particular effector proteins and vehicles to translocate them. To this end, a categorized gene called Salmonella pathogenicity island (SPI) was developed; genes like Salmonella pathogenicity island encode aggressive or modulating proteins. Initially, Salmonella needs to be attached and stabilized via adhesin factor, without which no further steps can be taken. In this review, an attempt has been made to elaborate on each factor attached to the host cell or to modulating and aggressive proteins that evade immune systems. This review includes four sections: (A) attachment factors or T3SS- independent entrance, (B) effector proteins or T3SS-dependent entrance, (c) regulation of invasive genes, and (D) regulation of immune responses.
Collapse
Affiliation(s)
- Taher Azimi
- Pediatric Infections Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Students Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Zamirnasta
- Clinical Microbiology Research Center, Ilam University of Medical Science, Ilam, Iran
| | - Mahmood Alizadeh Sani
- Food Safety and Hygiene Division, Environmental health Department, School of Public Health, Tehran University of medical sciences, Tehran, Iran
- Students Research Committee, Department of Food Sciences and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahmad Nasser
- Clinical Microbiology Research Center, Ilam University of Medical Science, Ilam, Iran
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Microbiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran
| |
Collapse
|
23
|
Wang C, Nie T, Lin F, Connerton IF, Lu Z, Zhou S, Hang H. Resistance mechanisms adopted by a Salmonella Typhimurium mutant against bacteriophage. Virus Res 2019; 273:197759. [PMID: 31539557 DOI: 10.1016/j.virusres.2019.197759] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/13/2019] [Accepted: 09/14/2019] [Indexed: 01/21/2023]
Abstract
Bacteriophages have key roles in regulating bacterial populations in most habitats. A Salmonella Typhimurium mutant (N18) with impaired sensitivity to phage fmb-p1 was obtained and examined, the adsorption efficiency of fmb-p1 to N18 was reduced to 6%, compared to more than 97% for wild type S. Typhimurium CMCC50115. Reduced adsorption was accompanied by a reduction of 90% in the LPS content compared to wild type. Electron microscopy showed phage scattered around N18 with minimal engagement, while the phage were efficiently adsorbed to the wild type with tails oriented towards the bacterial surface. Evidence suggests fmb-p1 can slightly infect N18 and this does not give rise to an increase of phage titer. RT-qPCR data show that several Salmonella genes involved in lipopolysaccharide synthesis and five virulence related genes were down-regulated upon exposure of N18 to phage fmb-p1. In contrast, phage resistance related genes such as the SOS response, restriction-modification (RM), and Cas1 gene were up-regulated in N18. These data suggest that although inefficient adsorption and entry is the primary mechanism of resistance, transcriptional responses to phage exposure indicate that alternative resistance mechanisms against phage infection are also brought to bear, including digestion of phage nucleic acids and activation of the SOS. These findings may help develop strategies for biocontrol of Salmonella where multi-resistant bacteria are encountered or emerge in applications for food production, bioremediation or wastewater treatment.
Collapse
Affiliation(s)
- Changbao Wang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China; College of Environmental Science and Engineering, Anhui Normal University, Wuhu 241002, PR China
| | - Ting Nie
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Fuxing Lin
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ian F Connerton
- Division of Food Sciences, School of Biosciences, Sutton Bonington Campus, University of Nottingham, Loughborough, Leicestershire LE12 5RD, United Kingdom
| | - Zhaoxin Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - Shoubiao Zhou
- College of Environmental Science and Engineering, Anhui Normal University, Wuhu 241002, PR China
| | - Hua Hang
- College of Environmental Science and Engineering, Anhui Normal University, Wuhu 241002, PR China
| |
Collapse
|
24
|
Wemyss MA, Pearson JS. Host Cell Death Responses to Non-typhoidal Salmonella Infection. Front Immunol 2019; 10:1758. [PMID: 31402916 PMCID: PMC6676415 DOI: 10.3389/fimmu.2019.01758] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022] Open
Abstract
Salmonella enterica subsp. enterica serovar Typhimurium (S. Typhimurium) is a Gram-negative bacterium with a broad host range that causes non-typhoidal salmonellosis in humans. S. Typhimurium infects epithelial cells and macrophages in the small intestine where it replicates in a specialized intracellular niche called the Salmonella-containing vacuole (SCV) and promotes inflammation of the mucosa to induce typically self-limiting gastroenteritis. Virulence and spread of the bacterium is determined in part by the host individual's ability to limit the infection through innate immune responses at the gastrointestinal mucosa, including programmed cell death. S. Typhimurium however, has evolved a myriad of mechanisms to counteract or exploit host responses through the use of Type III Secretion Systems (T3SS), which allow the translocation of virulence (effector) proteins into the host cell for the benefit of optimal bacterial replication and dissemination. T3SS effectors have been found to interact with apoptotic, necroptotic, and pyroptotic cell death cascades, interfering with both efficient clearance of the bacteria and the recruitment of neutrophils or dendritic cells to the area of infection. The interplay of host inflammation, programmed cell death responses, and bacterial defenses in the context of non-typhoidal Salmonella (NTS) infection is a continuing area of interest within the field, and as such has been reviewed here.
Collapse
Affiliation(s)
- Madeleine A Wemyss
- Department of Molecular and Translational Research, Monash University, Clayton, VIC, Australia.,Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Jaclyn S Pearson
- Department of Molecular and Translational Research, Monash University, Clayton, VIC, Australia.,Department of Microbiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
25
|
Barcoded Consortium Infections Resolve Cell Type-Dependent Salmonella enterica Serovar Typhimurium Entry Mechanisms. mBio 2019; 10:mBio.00603-19. [PMID: 31113898 PMCID: PMC6529635 DOI: 10.1128/mbio.00603-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Salmonella enterica serovar Typhimurium (S.Tm) is a widespread and broad-host-spectrum enteropathogen with the capacity to invade diverse cell types. Still, the molecular basis for the host cell invasion process has largely been inferred from studies of a few selected cell lines. Our work resolves the mechanisms that Salmonellae employ to invade prototypical host cell types, i.e., human epithelial, monocyte, and macrophage cells, at a previously unattainable level of temporal and quantitative precision. This highlights efficient bacterium-driven entry into innate immune cells and uncovers a type III secretion system effector module that dominates active bacterial invasion of not only epithelial cells but also monocytes and macrophages. The results are derived from a generalizable method, where we combine barcoding of the bacterial chromosome with mixed consortium infections of cultured host cells. The application of this methodology across bacterial species and infection models will provide a scalable means to address host-pathogen interactions in diverse contexts. Bacterial host cell invasion mechanisms depend on the bacterium’s virulence factors and the properties of the target cell. The enteropathogen Salmonella enterica serovar Typhimurium (S.Tm) invades epithelial cell types in the gut mucosa and a variety of immune cell types at later infection stages. The molecular mechanism(s) of host cell entry has, however, been studied predominantly in epithelial cell lines. S.Tm uses a type three secretion system (TTSS-1) to translocate effectors into the host cell cytosol, thereby sparking actin ruffle-dependent entry. The ruffles also fuel cooperative invasion by bystander bacteria. In addition, several TTSS-1-independent entry mechanisms exist, involving alternative S.Tm virulence factors, or the passive uptake of bacteria by phagocytosis. However, it remains ill-defined how S.Tm invasion mechanisms vary between host cells. Here, we developed an internally controlled and scalable method to map S.Tm invasion mechanisms across host cell types and conditions. The method relies on host cell infections with consortia of chromosomally tagged wild-type and mutant S.Tm strains, where the abundance of each strain can be quantified by qPCR or amplicon sequencing. Using this methodology, we quantified cooccurring TTSS-1-dependent, cooperative, and TTSS-1-independent invasion events in epithelial, monocyte, and macrophage cells. We found S.Tm invasion of epithelial cells and monocytes to proceed by a similar MOI-dependent mix of TTSS-1-dependent and cooperative mechanisms. TTSS-1-independent entry was more frequent in macrophages. Still, TTSS-1-dependent invasion dominated during the first minutes of interaction also with this cell type. Finally, the combined action of the SopB/SopE/SopE2 effectors was sufficient to explain TTSS-1-dependent invasion across both epithelial and phagocytic cells.
Collapse
|