1
|
Little JI, Singh PK, Zhao J, Dunn S, Matz H, Donnenberg MS. Type IV pili of Enterobacteriaceae species. EcoSal Plus 2024; 12:eesp00032023. [PMID: 38294234 PMCID: PMC11636386 DOI: 10.1128/ecosalplus.esp-0003-2023] [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: 04/03/2023] [Accepted: 12/01/2023] [Indexed: 02/01/2024]
Abstract
Type IV pili (T4Ps) are surface filaments widely distributed among bacteria and archaea. T4Ps are involved in many cellular functions and contribute to virulence in some species of bacteria. Due to the diversity of T4Ps, different properties have been observed for homologous proteins that make up T4Ps in various organisms. In this review, we highlight the essential components of T4Ps, their functions, and similarities to related systems. We emphasize the unique T4Ps of enteric pathogens within the Enterobacteriaceae family, which includes pathogenic strains of Escherichia coli and Salmonella. These include the bundle-forming pilus (BFP) of enteropathogenic E. coli (EPEC), longus (Lng) and colonization factor III (CFA/III) of enterotoxigenic E. coli (ETEC), T4P of Salmonella enterica serovar Typhi, Colonization Factor Citrobacter (CFC) of Citrobacter rodentium, T4P of Yersinia pseudotuberculosis, a ubiquitous T4P that was characterized in enterohemorrhagic E. coli (EHEC), and the R64 plasmid thin pilus. Finally, we highlight areas for further study.
Collapse
Affiliation(s)
- Janay I. Little
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Pradip K. Singh
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jinlei Zhao
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Shakeera Dunn
- Internal Medicine Residency, Bayhealth Medical Center, Dover, Delaware, USA
| | - Hanover Matz
- Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
2
|
Li Y, Hu MX, Yan M, Guo YX, Ma XK, Han JZ, Qin YM. Intestinal models based on biomimetic scaffolds with an ECM micro-architecture and intestinal macro-elasticity: close to intestinal tissue and immune response analysis. Biomater Sci 2023; 11:567-582. [PMID: 36484321 DOI: 10.1039/d2bm01051h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The synergetic biological effect of scaffolds with biomimetic properties including the ECM micro-architecture and intestinal macro-mechanical properties on intestinal models in vitro remains unclear. Here, we investigate the profitable role of biomimetic scaffolds on 3D intestinal epithelium models. Gelatin/bacterial cellulose nanofiber composite scaffolds crosslinked by the Maillard reaction are tuned to mimic the chemical component, nanofibrous network, and crypt architecture of intestinal ECM collagen and the stability and mechanical properties of intestinal tissue. In particular, scaffolds with comparable elasticity and viscoelasticity of intestinal tissue possess the highest biocompatibility and best cell proliferation and differentiation ability, which makes the intestinal epithelium models closest to their counterpart intestinal tissues. The constructed duodenal epithelium models and colon epithelium models are utilized to assess the immunobiotics-host interactions, and both of them can sensitively respond to foreign microorganisms, but the secretion levels of cytokines are intestinal cell specific. The results demonstrate that probiotics alleviate the inflammation and cell apoptosis induced by Escherichia coli, indicating that probiotics can protect the intestinal epithelium from damage by inhibiting the adhesion and invasion of E. coli to intestinal cells. The designed biomimetic scaffolds can serve as powerful tools to construct in vitro intestinal epithelium models, providing a convenient platform to screen intestinal anti-inflammatory components and even to assess other physiological functions of the intestine.
Collapse
Affiliation(s)
- Yue Li
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China.
| | - Meng-Xin Hu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China.
| | - Ming Yan
- School of Automation, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Ya-Xin Guo
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China.
| | - Xue-Ke Ma
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China.
| | - Jian-Zhong Han
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China.
| | - Yu-Mei Qin
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China.
| |
Collapse
|
3
|
Das S, Roychoudhury S, Roychoudhury S, Agarwal A, Henkel R. Role of Infection and Leukocytes in Male Infertility. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1358:115-140. [DOI: 10.1007/978-3-030-89340-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
4
|
Fu M, Liang S, Wu J, Hua Y, Chen H, Zhang Z, Liu J, Li X, Zhang B, Zhao W, Wan C. An Escherichia coli Effector Protein EspF May Induce Host DNA Damage via Interaction With SMC1. Front Microbiol 2021; 12:682064. [PMID: 34122393 PMCID: PMC8188558 DOI: 10.3389/fmicb.2021.682064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/30/2021] [Indexed: 12/22/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157: H7 is an important foodborne pathogen that causes human diarrhea, hemorrhagic colitis, and hemolytic uremic syndrome. EspF is one of the most important effector proteins injected by the Type III Secretion System. It can target mitochondria and nucleoli, stimulate host cells to produce ROS, and promote host cell apoptosis. However, the mechanism of the host-pathogen interaction leading to host oxidative stress and cell cytotoxic effects such as DNA damage remains to be elucidated. Here, we used Cell Counting Kit-8 (CCK-8) assays and 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-OHdG) ELISA to study cell viability and DNA oxidative damage level after exposure to EspF. Western blot and immunofluorescence were also used to determine the level of the DNA damage target protein p-H2AX and cell morphology changes after EspF infection. Moreover, we verified the toxicity in intestinal epithelial cells mediated by EspF infection in vivo. In addition, we screened the host proteins that interact with EspF using CoIP-MS. We found that EspF may more depend on its C-terminus to interact with SMC1, and EspF could activate SMC1 phosphorylation and migrate it to the cytoplasm. In summary, this study revealed that EspF might mediate host cell DNA damage and found a new interaction between EspF and the DNA damage repair protein SMC1. Thus, EspF may mediate DNA damage by regulating the subcellular localization and phosphorylation of SMC1.
Collapse
Affiliation(s)
- Muqing Fu
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Song Liang
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiali Wu
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Ying Hua
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hanzong Chen
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhikai Zhang
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jinyue Liu
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaoxia Li
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Bao Zhang
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wei Zhao
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Chengsong Wan
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
- Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
- *Correspondence: Chengsong Wan,
| |
Collapse
|
5
|
Hua Y, Yan K, Wan C. Clever Cooperation: Interactions Between EspF and Host Proteins. Front Microbiol 2018; 9:2831. [PMID: 30524410 PMCID: PMC6262023 DOI: 10.3389/fmicb.2018.02831] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022] Open
Abstract
EspF is a central effector protein of enterohemorrhagic Escherichia coli (EHEC), enteropathogenic E. coli (EPEC), and Citrobacter rodentium (CR) that is secreted through the type III secretion system to host cells. The interaction between EspF and host proteins plays an important role in bacterial pathogenesis. EspF protein binds to host SNX9 and N-WASP proteins to promote the colonization of pathogenic bacteria in intestinal epithelial cells; combines with cytokeratin 18, actin, 14-3-3ζ, Arp2/3, profilin, and ZO-1 proteins to intervene in the redistribution of intermediate filaments, the rearrangement of actin, and the disruption of tight junctions; acts together with Abcf2 to boost host cell intrinsic apoptosis; and collaborates with Anxa6 protein to inhibit phagocytosis. The interaction between EspF and host proteins is key to the pathogenic mechanism of EHEC and EPEC. Here, we review how EspF protein functions through interactions with these 10 host proteins and contributes to the pathogenicity of EHEC/EPEC.
Collapse
Affiliation(s)
- Ying Hua
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| | - Kaina Yan
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| | - Chengsong Wan
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| |
Collapse
|
6
|
Cavalcante PA, Prata MMG, Medeiros PHQS, Alves da Silva AV, Quetz JS, Reyes MAV, Rodrigues TS, Santos AKS, Ribeiro SA, Veras HN, Bona MD, Amaral MSMG, Rodrigues FAP, Lima IFN, Havt A, Lima AAM. Intestinal cell migration damage induced by enteropathogenic Escherichia coli strains. ACTA ACUST UNITED AC 2018; 51:e7423. [PMID: 30066727 PMCID: PMC6065879 DOI: 10.1590/1414-431x20187423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/24/2018] [Indexed: 01/27/2023]
Abstract
Epithelial cell migration is an essential response to enteric pathogens such as enteropathogenic Escherichia coli (EPEC). This study aimed to investigate the effects of EPEC infection on intestinal epithelial cell migration in vitro, as well as the involvement of type III secretion system (T3SS) and Rho GTPases. Crypt intestinal epithelial cells (IEC-6) were infected with EPEC strains (E2348/69, ΔescF, and the LDI001 strain isolated from a malnourished Brazilian child) and commensal E. coli HS. Wound migration and cell death assays were performed at different time-points. Transcription and expression of Rho GTPases were evaluated using real-time PCR and western blotting. Overall, EPEC E2348/69 reduced migration and increased apoptosis and necrosis levels compared to EPEC LDI001 and E. coli HS strains. Moreover, EPEC LDI001 impaired cell migration at a higher level than E. coli HS and increased necrosis after 24 hours compared to the control group. The different profiles of virulence genes between the two wild-type EPEC strains, characterized by the absence of espL and nleE genes in the LDI001, might explain the phenotypic results, playing significant roles on cell migration impairment and cell death-related events. Moreover, the type III secretion system is determinant for the inhibition of intestinal epithelial cell migration by EPEC 2348/69, as its deletion prevented the effect. Active Rac1 concentrations were increased in E2348/69 and LDI001-infected cells, while the T3SS-deficient strain did not demonstrate this activation. This study contributes with valuable insight to characterize the mechanisms involved in the impairment of intestinal cell migration induced by EPEC.
Collapse
Affiliation(s)
- P A Cavalcante
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M M G Prata
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - P H Q S Medeiros
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A V Alves da Silva
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - J S Quetz
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M A V Reyes
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - T S Rodrigues
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A K S Santos
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - S A Ribeiro
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - H N Veras
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M D Bona
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M S M G Amaral
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - F A P Rodrigues
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - I F N Lima
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A Havt
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A A M Lima
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
7
|
Priya A, Kaur K, Bhattacharyya S, Chakraborti A, Ghosh S. Cell cycle arrest and apoptosis induced by enteroaggregative Escherichia coli in cultured human intestinal epithelial cells. J Med Microbiol 2017; 66:217-225. [DOI: 10.1099/jmm.0.000405] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Anshu Priya
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education & Research, Chandigarh 160012, India
| | - Kiranjeet Kaur
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education & Research, Chandigarh 160012, India
| | - Shalmoli Bhattacharyya
- Department of Biophysics, Post Graduate Institute of Medical Education & Research, Chandigarh 160012, India
| | - Anuradha Chakraborti
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education & Research, Chandigarh 160012, India
| | - Sujata Ghosh
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education & Research, Chandigarh 160012, India
| |
Collapse
|
8
|
Chatterjee A, Caballero-Franco C, Bakker D, Totten S, Jardim A. Pore-forming Activity of the Escherichia coli Type III Secretion System Protein EspD. J Biol Chem 2015; 290:25579-94. [PMID: 26324713 DOI: 10.1074/jbc.m115.648204] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Indexed: 12/20/2022] Open
Abstract
Enterohemorrhagic Escherichia coli is a causative agent of gastrointestinal and diarrheal diseases. Pathogenesis associated with enterohemorrhagic E. coli involves direct delivery of virulence factors from the bacteria into epithelial cell cytosol via a syringe-like organelle known as the type III secretion system. The type III secretion system protein EspD is a critical factor required for formation of a translocation pore on the host cell membrane. Here, we show that recombinant EspD spontaneously integrates into large unilamellar vesicle (LUV) lipid bilayers; however, pore formation required incorporation of anionic phospholipids such as phosphatidylserine and an acidic pH. Leakage assays performed with fluorescent dextrans confirmed that EspD formed a structure with an inner diameter of ∼2.5 nm. Protease mapping indicated that the two transmembrane helical hairpin of EspD penetrated the lipid layer positioning the N- and C-terminal domains on the extralumenal surface of LUVs. Finally, a combination of glutaraldehyde cross-linking and rate zonal centrifugation suggested that EspD in LUV membranes forms an ∼280-320-kDa oligomeric structure consisting of ∼6-7 subunits.
Collapse
Affiliation(s)
- Abhishek Chatterjee
- From the Institute of Parasitology and Centre for Host-Parasite Interactions, McGill University, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Celia Caballero-Franco
- From the Institute of Parasitology and Centre for Host-Parasite Interactions, McGill University, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Dannika Bakker
- From the Institute of Parasitology and Centre for Host-Parasite Interactions, McGill University, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Stephanie Totten
- From the Institute of Parasitology and Centre for Host-Parasite Interactions, McGill University, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Armando Jardim
- From the Institute of Parasitology and Centre for Host-Parasite Interactions, McGill University, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| |
Collapse
|
9
|
Wong Fok Lung T, Pearson JS, Schuelein R, Hartland EL. The cell death response to enteropathogenic Escherichia coli infection. Cell Microbiol 2014; 16:1736-45. [PMID: 25266336 DOI: 10.1111/cmi.12371] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/24/2014] [Accepted: 09/26/2014] [Indexed: 12/13/2022]
Abstract
Given the critical roles of inflammation and programmed cell death in fighting infection, it is not surprising that many bacterial pathogens have evolved strategies to inactivate these defences. The causative agent of infant diarrhoea, enteropathogenic Escherichia coli (EPEC), is an extracellular, intestinal pathogen that blocks both inflammation and programmed cell death. EPEC attaches to enterocytes, remains in the gut lumen and utilizes a type III secretion system (T3SS) to inject multiple virulence effector proteins directly into the infected cell, many of which subvert host antimicrobial processes through the disruption of signalling pathways. Recently, T3SS effector proteins from EPEC have been identified that inhibit death receptor-induced apoptosis. Here we review the mechanisms used by EPEC T3SS effectors to manipulate apoptosis and promote host cell survival and discuss the role of these activities during infection.
Collapse
Affiliation(s)
- Tania Wong Fok Lung
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, 3000, Australia
| | | | | | | |
Collapse
|
10
|
Navarro-Garcia F, Serapio-Palacios A, Vidal JE, Salazar MI, Tapia-Pastrana G. EspC promotes epithelial cell detachment by enteropathogenic Escherichia coli via sequential cleavages of a cytoskeletal protein and then focal adhesion proteins. Infect Immun 2014; 82:2255-65. [PMID: 24643541 PMCID: PMC4019189 DOI: 10.1128/iai.01386-13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 03/06/2014] [Indexed: 12/19/2022] Open
Abstract
EspC is a non-locus of enterocyte effacement (LEE)-encoded autotransporter produced by enteropathogenic Escherichia coli (EPEC) that is secreted to the extracellular milieu by a type V secretion system and then translocated into epithelial cells by the type III secretion system. Here, we show that this efficient EspC delivery into the cell leads to a cytopathic effect characterized by cell rounding and cell detachment. Thus, EspC is the main protein involved in epithelial cell cytotoxicity detected during EPEC adhesion and pedestal formation assays. The cell detachment phenotype is triggered by cytoskeletal and focal adhesion disruption. EspC-producing EPEC is able to cleave fodrin, paxillin, and focal adhesion kinase (FAK), but these effects are not observed when cells are infected with an espC isogenic mutant. Recovery of these phenotypes by complementing the mutant with the espC gene but not with the espC gene mutated in the serine protease motif highlights the role of the protease activity of EspC in the cell detachment phenotype. In vitro assays using purified proteins showed that EspC, but not EspC with an S256I substitution [EspCS256I], directly cleaves these cytoskeletal and focal adhesion proteins. Kinetics of protein degradation indicated that EspC-producing EPEC first cleaves fodrin (within the 11th and 9th repetitive units at the Q1219 and D938 residues, respectively), and this event sequentially triggers paxillin degradation, FAK dephosphorylation, and FAK degradation. Thus, cytoskeletal and focal adhesion protein cleavage leads to the cell rounding and cell detachment promoted by EspC-producing EPEC.
Collapse
Affiliation(s)
- Fernando Navarro-Garcia
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | | | | | | | | |
Collapse
|
11
|
Barnett Foster D. Modulation of the enterohemorrhagic E. coli virulence program through the human gastrointestinal tract. Virulence 2013; 4:315-23. [PMID: 23552827 PMCID: PMC3710334 DOI: 10.4161/viru.24318] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Enteric pathogens must not only survive passage through the gastrointestinal tract but must also coordinate expression of virulence determinants in response to localized microenvironments with the host. Enterohemorrhagic Escherichia coli (EHEC), a serious food and waterborne human pathogen, is well equipped with an arsenal of molecular factors that allows it to survive passage through the gastrointestinal tract and successfully colonize the large intestine. This review will explore how EHEC responds to various environmental cues associated with particular microenvironments within the host and how it employs these cues to modulate virulence factor expression, with a view to developing a conceptual framework for understanding modulation of EHEC’s virulence program in response to the host. In vitro studies offer significant insights into the role of individual environmental cues but in vivo studies using animal models as well as data from natural infections will ultimately provide a more comprehensive picture of the highly regulated virulence program of this pathogen.
Collapse
Affiliation(s)
- Debora Barnett Foster
- Department of Chemistry and Biology, Faculty of Science, Ryerson University, Toronto, ONT, Canada.
| |
Collapse
|
12
|
Abstract
Enterohemorrhagic and enteropathogenic E. coli (EHEC and EPEC) can cause severe and potentially life-threatening infections. Their pathogenicity is mediated by at least 40 effector proteins which they inject into their host cells by a type-III secretion system leading to the subversion of several cellular pathways. However, the molecular function of several effectors remains unknown, even though they contribute to virulence. Here we show that one of them, NleF, binds to caspase-4, -8, and -9 in yeast two-hybrid, LUMIER, and direct interaction assays. NleF inhibits the catalytic activity of the caspases in vitro and in cell lysate and prevents apoptosis in HeLa and Caco-2 cells. We have solved the crystal structure of the caspase-9/NleF complex which shows that NleF uses a novel mode of caspase inhibition, involving the insertion of the carboxy-terminus of NleF into the active site of the protease. In conformance with our structural model, mutagenized NleF with truncated or elongated carboxy-termini revealed a complete loss in caspase binding and apoptosis inhibition. Evasion of apoptosis helps pathogenic E. coli and other pathogens to take over the host cell by counteracting the cell’s ability to self-destruct upon infection. Recently, two other effector proteins, namely NleD and NleH, were shown to interfere with apoptosis. Even though NleF is not the only effector protein capable of apoptosis inhibition, direct inhibition of caspases by bacterial effectors has not been reported to date. Also unique so far is its mode of inhibition that resembles the one obtained for synthetic peptide-type inhibitors and as such deviates substantially from previously reported caspase-9 inhibitors such as the BIR3 domain of XIAP.
Collapse
|
13
|
Buret AG, Bhargava A. Modulatory mechanisms of enterocyte apoptosis by viral, bacterial and parasitic pathogens. Crit Rev Microbiol 2013; 40:1-17. [PMID: 23297858 DOI: 10.3109/1040841x.2012.746952] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14
|
Buryakina TY, Su PT, Syu W, Chang CA, Fan HF, Kao FJ. Metabolism of HeLa cells revealed through autofluorescence lifetime upon infection with enterohemorrhagic Escherichia coli. JOURNAL OF BIOMEDICAL OPTICS 2012; 17:101503. [PMID: 23223979 DOI: 10.1117/1.jbo.17.10.101503] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Fluorescence lifetime imaging microscopy (FLIM) is a sensitive technique in monitoring functional and conformational states of nicotinamide adenine dinucleotide reduced (NADH) and flavin adenine dinucleotide (FAD),main compounds participating in oxidative phosphorylation in cells. In this study, we have applied FLIM to characterize the metabolic changes in HeLa cells upon bacterial infection and made comparison with the results from the cells treated with staurosporine (STS), a well-known apoptosis inducer. The evolving of NADH's average autofluorescence lifetime during the 3 h after infection with enterohemorragic Escherichia coli (EHEC) or STS treatment has been observed. The ratio of the short and the long lifetime components' relative contributions of NADH increases with time, a fact indicating cellular metabolic activity, such as a decrease of oxidative phosphorylation over the course of infection, while opposite dynamics is observed in FAD. Being associated with mitochondria, FAD lifetimes and redox ratio could indicate heterogeneous mitochondrial function, microenvironment with bacterial infection, and further pathway to cell death. The redox ratios for both EHEC-infected and STS-treated HeLa cells have been observed and these observations also indicate possible apoptosis induced by bacterial infection.
Collapse
Affiliation(s)
- Tatyana Yu Buryakina
- Institute of Biophotonics, National Yang-Ming University, 155 Linong Street, Section 2, Taipei 11221, Taiwan
| | | | | | | | | | | |
Collapse
|
15
|
The enteropathogenic Escherichia coli-secreted protein EspZ inhibits host cell apoptosis. Infect Immun 2012; 80:3850-7. [PMID: 22907816 DOI: 10.1128/iai.00335-12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The diarrheagenic pathogen enteropathogenic Escherichia coli (EPEC) limits the death of infected enterocytes early in infection. A number of bacterial molecules and host signaling pathways contribute to the enhanced survival of EPEC-infected host cells. EspZ, a type III secreted effector protein that is unique to EPEC and related "attaching and effacing" (A/E) pathogens, plays a role in limiting host cell death, but the precise host signaling pathways responsible for this phenotype are not known. We hypothesized that EspZ contributes to the survival of infected intestinal epithelial cells by interfering with apoptosis. Consistent with previous studies, scanning electron microscopy analysis of intestinal epithelial cells infected with an EPEC espZ mutant (ΔespZ) showed increased levels of apoptotic and necrotic cells compared to cells infected with the isogenic parent strain. Correspondingly, higher levels of cytosolic cytochrome c and increased activation of caspases 9, 7, and 3 were observed for ΔespZ strain-infected cells compared to wild-type (WT) EPEC-infected cells. Finally, espZ-transfected epithelial cells were significantly protected from staurosporine-induced, but not tumor necrosis factor alpha (TNF-α)/cycloheximide-induced, apoptosis. Thus, EspZ contributes to epithelial cell survival by mechanisms that include the inhibition of the intrinsic apoptotic pathway. The enhanced survival of infected enterocytes by molecules such as EspZ likely plays a key role in optimal colonization by A/E pathogens.
Collapse
|
16
|
Meningococcal outer membrane protein NhhA triggers apoptosis in macrophages. PLoS One 2012; 7:e29586. [PMID: 22238624 PMCID: PMC3251587 DOI: 10.1371/journal.pone.0029586] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 12/01/2011] [Indexed: 11/19/2022] Open
Abstract
Phagocytotic cells play a fundamental role in the defense against bacterial pathogens. One mechanism whereby bacteria evade phagocytosis is to produce factors that trigger apoptosis. Here we identify for the first time a meningococcal protein capable of inducing macrophage apoptosis. The conserved meningococcal outer membrane protein NhhA (Neisseria hia/hsf homologue A, also known as Hsf) mediates bacterial adhesion and interacts with extracellular matrix components heparan sulphate and laminin. Meningococci lacking NhhA fail to colonise nasal mucosa in a mouse model of meningococcal disease. We found that exposure of macrophages to NhhA resulted in a highly increased rate of apoptosis that proceeded through caspase activation. Exposure of macrophages to NhhA also led to iNOS induction and nitric oxide production. However, neither nitric oxide production nor TNF-α signaling was found to be a prerequisite for NhhA-induced apoptosis. Macrophages exposed to wildtype NhhA-expressing meningococci were also found to undergo apoptosis whereas NhhA-deficient meningococci had a markedly decreased capacity to induce macrophage apoptosis. These data provide new insights on the role of NhhA in meningococcal disease. NhhA-induced macrophage apoptosis could be a mechanism whereby meningococci evade immunoregulatory and phagocytotic actions of macrophages.
Collapse
|
17
|
Tsumagari K, Chang SC, Lacey M, Baribault C, Chittur SV, Sowden J, Tawil R, Crawford GE, Ehrlich M. Gene expression during normal and FSHD myogenesis. BMC Med Genomics 2011; 4:67. [PMID: 21951698 PMCID: PMC3204225 DOI: 10.1186/1755-8794-4-67] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 09/27/2011] [Indexed: 01/31/2023] Open
Abstract
Background Facioscapulohumeral muscular dystrophy (FSHD) is a dominant disease linked to contraction of an array of tandem 3.3-kb repeats (D4Z4) at 4q35. Within each repeat unit is a gene, DUX4, that can encode a protein containing two homeodomains. A DUX4 transcript derived from the last repeat unit in a contracted array is associated with pathogenesis but it is unclear how. Methods Using exon-based microarrays, the expression profiles of myogenic precursor cells were determined. Both undifferentiated myoblasts and myoblasts differentiated to myotubes derived from FSHD patients and controls were studied after immunocytochemical verification of the quality of the cultures. To further our understanding of FSHD and normal myogenesis, the expression profiles obtained were compared to those of 19 non-muscle cell types analyzed by identical methods. Results Many of the ~17,000 examined genes were differentially expressed (> 2-fold, p < 0.01) in control myoblasts or myotubes vs. non-muscle cells (2185 and 3006, respectively) or in FSHD vs. control myoblasts or myotubes (295 and 797, respectively). Surprisingly, despite the morphologically normal differentiation of FSHD myoblasts to myotubes, most of the disease-related dysregulation was seen as dampening of normal myogenesis-specific expression changes, including in genes for muscle structure, mitochondrial function, stress responses, and signal transduction. Other classes of genes, including those encoding extracellular matrix or pro-inflammatory proteins, were upregulated in FSHD myogenic cells independent of an inverse myogenesis association. Importantly, the disease-linked DUX4 RNA isoform was detected by RT-PCR in FSHD myoblast and myotube preparations only at extremely low levels. Unique insights into myogenesis-specific gene expression were also obtained. For example, all four Argonaute genes involved in RNA-silencing were significantly upregulated during normal (but not FSHD) myogenesis relative to non-muscle cell types. Conclusions DUX4's pathogenic effect in FSHD may occur transiently at or before the stage of myoblast formation to establish a cascade of gene dysregulation. This contrasts with the current emphasis on toxic effects of experimentally upregulated DUX4 expression at the myoblast or myotube stages. Our model could explain why DUX4's inappropriate expression was barely detectable in myoblasts and myotubes but nonetheless linked to FSHD.
Collapse
Affiliation(s)
- Koji Tsumagari
- Human Genetics Program, Tulane Medical School, New Orleans, LA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wong ARC, Pearson JS, Bright MD, Munera D, Robinson KS, Lee SF, Frankel G, Hartland EL. Enteropathogenic and enterohaemorrhagic Escherichia coli: even more subversive elements. Mol Microbiol 2011; 80:1420-38. [PMID: 21488979 DOI: 10.1111/j.1365-2958.2011.07661.x] [Citation(s) in RCA: 256] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Alexander R C Wong
- Centre for Molecular Microbiology and Infection, Division of Cell and Molecular Biology, Imperial College London, London SW7 2AZ, UK
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Brereton CF, Blander JM. The unexpected link between infection-induced apoptosis and a TH17 immune response. J Leukoc Biol 2011; 89:565-76. [PMID: 21248151 DOI: 10.1189/jlb.0710421] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Microbial pathogens can initiate MOMP in host cells and as such, initiate the mitochondrial pathway of apoptosis. Innate immune recognition of cells dying in this way by infection-induced apoptosis would involve recognition of ligands derived from the apoptotic host cell simultaneously with those derived from the infecting pathogen. The resultant signal transduction pathways engaged direct DCs to concomitantly synthesize TGF-β and IL-6, two cytokines that subsequently favor the differentiation of naïve CD4 T cells into T(h)17 cells. Citrobacter rodentium is one rodent pathogen that targets mitochondria and induces apoptosis, and blockade of apoptosis during enteric Citrobacter infection impairs the characteristic T(h)17 response in the intestinal LP. Here, we review these original findings. We discuss microbial infections other than Citrobacter that have been shown to induce T(h)17 responses, and we examine what is known about the ability of those pathogens to induce apoptosis. We also consider types of cell death other than apoptosis that can be triggered by microbial infection, and we highlight how little we know about the impact of various forms of cell death on the ensuing adaptive immune response.
Collapse
Affiliation(s)
- Corinna F Brereton
- Mount Sinai School of Medicine, Immunology Institute, Department of Medicine, 1425 Madison Ave., 12-20D, New York, NY 10029, USA
| | | |
Collapse
|
20
|
Abstract
Two of the critical cytokines required for the differentiation of T helper 17 (T(H)17) cells from naive CD4 T cells are transforming growth factor-beta (TGF-β) and interleukin-6 (IL-6). Innate recognition of apoptotic cells in the presence of Toll-like receptor engagement directs the simultaneous synthesis of these cytokines by antigen-presenting cells (APCs), and as such provides a cytokine milieu that favors T(H)17 cell induction. In this situation, APCs are activated in response to ligands derived from apoptotic cells, but also to those from the infecting pathogen. Induction of a T(H)17 response against Citrobacter rodentium infection was dependent on the ability of Citrobacter to induce apoptosis of intestinal epithelial cells. In this review, we will discuss how simultaneous activation of inflammatory and noninflammatory pattern recognition receptors on APCs impacts T helper cell differentiation, and what relevance this effect has on the immune response generated against bacterial infections that cause host cell apoptosis.
Collapse
Affiliation(s)
- Corinna F Brereton
- Department of Medicine, Immunology Institute, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | |
Collapse
|
21
|
Breaking the stereotype: virulence factor-mediated protection of host cells in bacterial pathogenesis. PLoS Pathog 2010; 6:e1001057. [PMID: 20862318 PMCID: PMC2940752 DOI: 10.1371/journal.ppat.1001057] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
22
|
Isolates of the Enterobacter cloacae complex induce apoptosis of human intestinal epithelial cells. Microb Pathog 2010; 49:83-9. [DOI: 10.1016/j.micpath.2010.04.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 04/22/2010] [Accepted: 04/23/2010] [Indexed: 12/15/2022]
|
23
|
Shames SR, Deng W, Guttman JA, de Hoog CL, Li Y, Hardwidge PR, Sham HP, Vallance BA, Foster LJ, Finlay BB. The pathogenic E. coli type III effector EspZ interacts with host CD98 and facilitates host cell prosurvival signalling. Cell Microbiol 2010; 12:1322-39. [PMID: 20374249 DOI: 10.1111/j.1462-5822.2010.01470.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Enterohaemorrhagic and enteropathogenic Escherichia coli (EHEC and EPEC respectively) are diarrhoeal pathogens that cause the formation of attaching and effacing (A/E) lesions on infected host cells. These pathogens encode a type III secretion system (T3SS) used to inject effector proteins directly into host cells, an essential requirement for virulence. In this study, we identified a function for the type III secreted effector EspZ. Infection with EPEC DeltaespZ caused increased cytotoxicity in HeLa and MDCK cells compared with wild-type EPEC, and expressing espZ in cells abrogated this effect. Using yeast two-hybrid, proteomics, immunofluorescence and co-immunoprecipitation, it was demonstrated that EspZ interacts with the host protein CD98, which contributes to protection against EPEC-mediated cytotoxicity. EspZ enhanced phosphorylation of focal adhesion kinase (FAK) and AKT during infection with EPEC, but CD98 only appeared to facilitate FAK phosphorylation. This study provides evidence that EspZ and CD98 promote host cell survival mechanisms involving FAK during A/E pathogen infection.
Collapse
Affiliation(s)
- Stephanie R Shames
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Sukumaran SK, Fu NY, Tin CB, Wan KF, Lee SS, Yu VC. A Soluble Form of the Pilus Protein FimA Targets the VDAC-Hexokinase Complex at Mitochondria to Suppress Host Cell Apoptosis. Mol Cell 2010; 37:768-83. [DOI: 10.1016/j.molcel.2010.02.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 10/28/2009] [Accepted: 12/24/2009] [Indexed: 11/16/2022]
|
25
|
NleH effectors interact with Bax inhibitor-1 to block apoptosis during enteropathogenic Escherichia coli infection. Proc Natl Acad Sci U S A 2010; 107:3129-34. [PMID: 20133763 DOI: 10.1073/pnas.0911609106] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The human pathogens enteropathogenic (EPEC) and enterohemorrhagic Escherichia coli and the related mouse pathogen Citrobacter rodentium subvert a variety of host cell signaling pathways via their plethora of type III secreted effectors, including triggering of an early apoptotic response. EPEC-infected cells do not develop late apoptotic symptoms, however. In this study we demonstrate that the NleH family effectors, homologs of the Shigella effector kinase OspG, blocks apoptosis. During EPEC infection, NleH effectors inhibit elevation of cytosolic Ca(2+) concentrations, nuclear condensation, caspase-3 activation, and membrane blebbing and promote cell survival. NleH1 alone is sufficient to prevent procaspase-3 cleavage induced by the proapoptotic compounds staurosporine, brefeldin A, and tunicamycin. Using C. rodentium, we found that NleH inhibits procaspase-3 cleavage at the bacterial attachment sites in vivo. A yeast two-hybrid screen identified the endoplasmic reticulum six-transmembrane protein Bax inhibitor-1 (BI-1) as an NleH-interacting partner. We mapped the NleH-binding site to the N-terminal 40 amino acids of BI-1. Knockdown of BI-1 resulted in the loss of NleH's antiapoptotic activity. These results indicate that NleH effectors are inhibitors of apoptosis that may act through BI-1 to carry out their cytoprotective function.
Collapse
|
26
|
The enteropathogenic Escherichia coli effector Cif induces delayed apoptosis in epithelial cells. Infect Immun 2009; 77:5471-7. [PMID: 19786559 DOI: 10.1128/iai.00860-09] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The cycle inhibiting factor (Cif) belongs to a family of bacterial toxins, the cyclomodulins, which modulate the host cell cycle. Upon injection into the host cell by the type III secretion system of enteropathogenic Escherichia coli (EPEC), Cif induces both G(2) and G(1) cell cycle arrests. The cell cycle arrests correlate with the accumulation of p21(waf1) and p27(kip1) proteins that inhibit CDK-cyclin complexes, whose activation is required for G(1)/S and G(2)/M transitions. Increases of p21 and p27 levels are independent of p53 transcriptional induction and result from protein stabilization through inhibition of the ubiquitin/proteasome degradation pathway. In this study, we show that Cif not only induces cell cycle arrest but also eventually provokes a delayed cell death. Indeed, 48 h after infection with EPEC expressing Cif, cultured IEC-6 intestinal cells were positive for extracellular binding of annexin V and exhibited high levels of cleaved caspase-3 and lactate dehydrogenase release, indicating evidence of apoptosis. Cif was necessary and sufficient for inducing this late apoptosis, and the cysteine residue of the catalytic site was required for Cif activity. These results highlight a more complex role of Cif than previously thought, as a cyclomodulin but also as an apoptosis inducer.
Collapse
|
27
|
Deghmane AE, Veckerlé C, Giorgini D, Hong E, Ruckly C, Taha MK. Differential modulation of TNF-alpha-induced apoptosis by Neisseria meningitidis. PLoS Pathog 2009; 5:e1000405. [PMID: 19412525 PMCID: PMC2669886 DOI: 10.1371/journal.ppat.1000405] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Accepted: 03/31/2009] [Indexed: 01/01/2023] Open
Abstract
Infections by Neisseria meningitidis show duality between frequent asymptomatic carriage and occasional life-threatening disease. Bacterial and host factors involved in this balance are not fully understood. Cytopathic effects and cell damage may prelude to pathogenesis of isolates belonging to hyper-invasive lineages. We aimed to analyze cell–bacteria interactions using both pathogenic and carriage meningococcal isolates. Several pathogenic isolates of the ST-11 clonal complex and carriage isolates were used to infect human epithelial cells. Cytopathic effect was determined and apoptosis was scored using several methods (FITC-Annexin V staining followed by FACS analysis, caspase assays and DNA fragmentation). Only pathogenic isolates were able to induce apoptosis in human epithelial cells, mainly by lipooligosaccharide (endotoxin). Bioactive TNF-α is only detected when cells were infected by pathogenic isolates. At the opposite, carriage isolates seem to provoke shedding of the TNF-α receptor I (TNF-RI) from the surface that protect cells from apoptosis by chelating TNF-α. Ability to induce apoptosis and inflammation may represent major traits in the pathogenesis of N. meningitidis. However, our data strongly suggest that carriage isolates of meningococci reduce inflammatory response and apoptosis induction, resulting in the protection of their ecological niche at the human nasopharynx. Acquisition of Neisseria meningitidis often leads to asymptomatic colonization (carriage) and rarely results in invasive disease associated with tissue injury. The reasons that make disease-associated isolates (pathogenic isolates) but not asymptomatic carriage isolates able to invade the host to establish disease are not understood. Isolates belonging to the ST-11 clonal complex are most frequently associated with the disease and rarely found in carriers. These hyper-invasive isolates may be able to induce cytopathic effects in target cells. We aimed to investigate the cytopathic effect of meningococcal isolates on epithelial cells using both ST-11 pathogenic isolates and carriage isolates. We showed that cytopathic effects were strongly associated with pathogenic isolates and infected cells exhibited features of apoptosis. This effect is mainly mediated by bacterial endotoxin (lipooligosaccharide) and involved an autocrine signaling mechanism of secreted TNF-α through its receptor TNF-RI. In contrast, carriage isolates down-regulate TNF-RI on the surface of infected cells by increasing TNF-RI shedding into the medium. We suggest that chelating secreted TNF-α protects cells from apoptosis. Our results unravel a differential modulation of TNF-α signaling by meningococcal isolates leading to cell survival or death and would therefore contribute to better understanding of the duality between carriage and invasiveness.
Collapse
|
28
|
Sason H, Milgrom M, Weiss AM, Melamed-Book N, Balla T, Grinstein S, Backert S, Rosenshine I, Aroeti B. Enteropathogenic Escherichia coli subverts phosphatidylinositol 4,5-bisphosphate and phosphatidylinositol 3,4,5-trisphosphate upon epithelial cell infection. Mol Biol Cell 2008; 20:544-55. [PMID: 18987340 DOI: 10.1091/mbc.e08-05-0516] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Phosphatidylinositol 4,5-bisphosphate [PI(4,5)P(2)] and phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P(3)] are phosphoinositides (PIs) present in small amounts in the inner leaflet of the plasma membrane (PM) lipid bilayer of host target cells. They are thought to modulate the activity of proteins involved in enteropathogenic Escherichia coli (EPEC) infection. However, the role of PI(4,5)P(2) and PI(3,4,5)P(3) in EPEC pathogenesis remains obscure. Here we show that EPEC induces a transient PI(4,5)P(2) accumulation at bacterial infection sites. Simultaneous actin accumulation, likely involved in the construction of the actin-rich pedestal, is also observed at these sites. Acute PI(4,5)P(2) depletion partially diminishes EPEC adherence to the cell surface and actin pedestal formation. These findings are consistent with a bimodal role, whereby PI(4,5)P(2) contributes to EPEC association with the cell surface and to the maximal induction of actin pedestals. Finally, we show that EPEC induces PI(3,4,5)P(3) clustering at bacterial infection sites, in a translocated intimin receptor (Tir)-dependent manner. Tir phosphorylated on tyrosine 454, but not on tyrosine 474, forms complexes with an active phosphatidylinositol 3-kinase (PI3K), suggesting that PI3K recruited by Tir prompts the production of PI(3,4,5)P(3) beneath EPEC attachment sites. The functional significance of this event may be related to the ability of EPEC to modulate cell death and innate immunity.
Collapse
Affiliation(s)
- Hagit Sason
- Department of Cell and Animal Biology, Confocal Unit, Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Flynn AN, Buret AG. Caspases-3, -8, and -9 are required for induction of epithelial cell apoptosis by enteropathogenic E. coli but are dispensable for increased paracellular permeability. Microb Pathog 2008; 44:311-9. [DOI: 10.1016/j.micpath.2007.10.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2007] [Revised: 10/10/2007] [Accepted: 10/11/2007] [Indexed: 11/24/2022]
|
30
|
Cheung KJ, Li G, Urban TA, Goldberg JB, Griffith A, Lu F, Burns JL. Pilus-mediated epithelial cell death in response to infection with Burkholderia cenocepacia. Microbes Infect 2007; 9:829-37. [PMID: 17537663 DOI: 10.1016/j.micinf.2007.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2006] [Revised: 01/09/2007] [Accepted: 03/01/2007] [Indexed: 11/20/2022]
Abstract
Burkholderia cenocepacia is an opportunistic pathogen that can cause serious infections in cystic fibrosis (CF) patients. The ET12 lineage appears particularly virulent in CF; however, its pathogenesis is poorly understood and may be associated with host response. To help characterize this response, the ability of B. cenocepacia to induce cytotoxicity and apoptosis in an epithelial cell model was examined. Upon infection with B. cenocepacia strain K56-2, A549 human lung epithelial cells underwent significant cell death; propidium iodine staining and DNA fragmentation assays suggested apoptosis. Initiation of cell death was independent of the type III secretion system, biofilm formation, and secreted bacterial cytotoxins. However, the frequency of cell death was lower in cells infected with a non-piliated mutant, K56-2 cblA::Tp. Furthermore, purified cbl pili were found to directly induce cytotoxicity in A549 cells and activate caspase-9, -8, -7, and -3, the major cysteine proteinases involved in apoptosis. It appears that B. cenocepacia cbl pili, which are a distinctive feature of the ET12 lineage, act as an initiator of cytotoxicity and apoptosis. Understanding the role of cbl pili in the pathogenesis of B. cenocepacia infections offers the potential for decreasing the virulence of these potentially life-threatening organisms in CF patients.
Collapse
Affiliation(s)
- K-John Cheung
- Division of Infectious Diseases, Immunology and Rheumatology, Children's Hospital and Regional Medical Center, 307 Westlake Avenue N., Seattle, WA 98109, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Barry DP, Beaman BL. Nocardia asteroides strain GUH-2 induces proteasome inhibition and apoptotic death of cultured cells. Res Microbiol 2006; 158:86-96. [PMID: 17258894 PMCID: PMC1831872 DOI: 10.1016/j.resmic.2006.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2006] [Revised: 11/22/2006] [Accepted: 11/22/2006] [Indexed: 01/03/2023]
Abstract
Many bacterial pathogens have the ability to induce apoptosis in their hosts. It was previously shown that Nocardia asteroides strain GUH-2, a Gram-positive facultatively intracellular pathogen, is capable of inducing the apoptotic death of dopaminergic cells in the murine brain and in PC12 cells, a rat cell line. In this study, the apoptosis-inducing potential of N. asteroides GUH-2 was further explored using HeLa cells, a human epithelial cell line. HeLa cells were incubated for 5h with live nocardiae, heat-killed bacteria, or unconcentrated nocardial culture filtrate, and changes to the cells were monitored. Consistent with the previous studies, N. asteroides GUH-2 induced DNA fragmentation and apoptosis in HeLa cells. Caspase activation and disruption of the mitochondrial membrane potential were also investigated to determine their roles in the induction of cell death. In all these experiments, significant changes were only induced by live nocardiae. A recent publication demonstrated that systemic administration of proteasome inhibitors can induce a Parkinsonian syndrome in rats that includes intraneuronal inclusions and characteristic behavioral alterations. Similar effects have been observed in mice and monkeys infected with N. asteroides GUH-2. In addition, some reports have shown that proteasome inhibition causes apoptotic death of affected cells. We therefore investigated the ability of N. asteroides GUH-2 to inhibit proteasome activity. Proteasome activity was significantly reduced, suggesting that this mechanism may be involved in the induction of apoptosis by these bacteria.
Collapse
Affiliation(s)
- Daniel P. Barry
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616
| | - Blaine L. Beaman
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616
- * Correspondence and reprints:
| |
Collapse
|
32
|
Fabian D, Dusan F, Sabol M, Marián S, Domaracká K, Katarína D, Bujnáková D, Dobroslava B. Essential oils--their antimicrobial activity against Escherichia coli and effect on intestinal cell viability. Toxicol In Vitro 2006; 20:1435-45. [PMID: 16919909 DOI: 10.1016/j.tiv.2006.06.012] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2006] [Revised: 06/19/2006] [Accepted: 06/26/2006] [Indexed: 11/26/2022]
Abstract
Essential oils are known to possess antimicrobial activity against a wide spectrum of bacteria. The main objective of this study was to evaluate possible harmful effects of four commonly used essential oils and their major components on intestinal cells. Antimicrobial activity of selected plant extracts against enteroinvasive Escherichia coli was dose dependent. However, doses of essential oils with the ability to completely inhibit bacterial growth (0.05%) showed also relatively high cytotoxicity to intestinal-like cells cultured in vitro. Lower doses of essential oils (0.01%) had only partial antimicrobial activity and their damaging effect on Caco-2 cells was only modest. Cell death assessment based on morphological and viability staining followed by fluorescence microscopy showed that essential oils of cinnamon and clove and their major component eugenol had almost no cytotoxic effect at lower doses. Although essential oil of oregano and its component carvacrol slightly increased the incidence of apoptotic cell death, they showed extensive antimicrobial activity even at lower concentrations. Relatively high cytotoxicity was demonstrated by thyme oil, which increased both apoptotic and necrotic cell death incidence. In contrast, its component thymol showed no cytotoxic effect as well as greatly-reduced ability to inhibit visible growth of the chosen pathogen in the doses used. On the other hand, the addition of all essential oils and their components at lower doses, with the exception of thyme oil, to bacterial suspension significantly reduced the cytotoxic effect of E. coli on Caco-2 cells after 1h culture. In conclusion, it is possible to find appropriate doses of essential oils showing both antimicrobial activity and very low detrimental effect on intestinal cells.
Collapse
Affiliation(s)
- Dusan Fabian
- Institute of Animal Physiology, Slovak Academy of Sciences, Soltésovej 4-6, 04001 Kosice, Slovakia.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Edelblum KL, Yan F, Yamaoka T, Polk DB. Regulation of apoptosis during homeostasis and disease in the intestinal epithelium. Inflamm Bowel Dis 2006; 12:413-24. [PMID: 16670531 DOI: 10.1097/01.mib.0000217334.30689.3e] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A single epithelial layer serves as the interface between the organism and the contents of the gastrointestinal tract, underlining the importance of regulating cellular viability despite an onslaught of pathogens, toxins, waste by-products, and cytokines. A balance between cellular proliferation and apoptosis is necessary to maintain this critical barrier. Recent findings have begun to explain the mechanisms by which intestinal epithelial cells are able to survive in such an environment and how loss of normal regulatory processes may lead to inflammatory bowel disease (IBD) and predispose to inflammation-associated neoplasia. This review focuses on the regulation of physiological apoptosis in development and homeostasis and on pathological apoptosis in intestinal disease, inflammation, and neoplasia, identifying remaining questions and areas of needed investigation.
Collapse
Affiliation(s)
- Karen L Edelblum
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0696, USA
| | | | | | | |
Collapse
|
34
|
Tsalie E, Kouzi K, Poutahidis T, Abas Z, Sarris K, Iliadis N, Kaldrymidou E. Effect of Vitamin E Nutritional Supplementation on the Pathological Changes Induced in the Ileum of Rabbits by Experimental Infection with Enteropathogenic Escherichia coli. J Comp Pathol 2006; 134:308-19. [PMID: 16714030 DOI: 10.1016/j.jcpa.2005.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Accepted: 12/20/2005] [Indexed: 11/15/2022]
Abstract
A well-established rabbit model of enteropathogenic E. coli (EPEC) disease was used to examine whether vitamin E (VE) nutritional supplementation had an effect on the pathological changes induced in the bowel by EPEC. Quantitative methods were used to evaluate the influence of VE on bacterial colonization, intestinal mucosal architecture and inflammation, and intestinal epithelial proliferation and apoptosis. VE did not affect EPEC colonization and did not give significant protection against EPEC-induced changes and diarrhoea. Although VE had no effect on the EPEC-related increase of enterocyte apoptosis, it clearly contributed to an acceleration of epithelial cell proliferation in the ileal crypts. This finding may explain why ileal morphometry undertaken in this study showed that VE ameliorated somewhat the effects of EPEC on intestinal mucosal architecture. Quantitative studies on inflammatory cells in the intestinal mucosa revealed that VE nutritional supplementation resulted in an increased neutrophilic and mononuclear inflammatory cell response to EPEC infection, which did not contribute, however, to the clearance of infection.
Collapse
Affiliation(s)
- E Tsalie
- Laboratory of Pathology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | | | | | | | | | | | | |
Collapse
|
35
|
Crane JK, Vezina CM. Externalization of host cell protein kinase C during enteropathogenic Escherichia coli infection. Cell Death Differ 2005; 12:115-27. [PMID: 15578063 DOI: 10.1038/sj.cdd.4401531] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) is a common cause of diarrhea in children in developing countries. Protein kinase C (PKC), a serine- and threonine-directed protein kinase, is rapidly activated following EPEC infection and this is accompanied by its translocation to a membrane-bound location where it is tightly bound to phosphatidylserine (PS). EPEC infection causes host cell death, one of whose features is externalization of PS. We hypothesized that externalization of PS would be accompanied by externalization of PKC as well. We report that EPEC infection triggers the externalization of PKC to the outer surface of the host cell. Ecto-PKC remains firmly tethered to the cell but can be released by incubation with peptide or protein substrates for the enzyme. Ecto-PKC is intact and biologically active and able to phosphorylate protein substrates on the surface of the host cell. Phosphorylation of whole EPEC bacteria or EPEC-secreted proteins could not be detected. Externalization of PKC could be reproduced by the combination of an apoptotic stimulus (ultraviolet (UV) irradiation) and phorbol myristate acetate (PMA), a procedure which resulted in externalization of >25% of the total cellular content of PKC-alpha. In the presence of ATP, ecto-PKC inhibited UV-induced cell shrinkage, membrane blebbing, and propidium iodide uptake but not the activation of caspases 3 and 7. This is the first report that expression of an ecto-protein kinase is altered by a microbial pathogen and the first to note that externalization of PKC can accompany apoptosis.
Collapse
Affiliation(s)
- J K Crane
- Department of Medicine, University at Buffalo, Buffalo, NY, USA.
| | | |
Collapse
|
36
|
Crane JK, Naeher TM, Choudhari SS, Giroux EM. Two pathways for ATP release from host cells in enteropathogenic Escherichia coli infection. Am J Physiol Gastrointest Liver Physiol 2005; 289:G407-17. [PMID: 16093420 DOI: 10.1152/ajpgi.00137.2005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We previously reported that enteropathogenic Escherichia coli (EPEC) infection triggered a large release of ATP from the host cell that was correlated with and dependent on EPEC-induced killing of the host cell. We noted, however, that under some circumstances, EPEC-induced ATP release exceeded that which could be accounted for on the basis of host cell killing. For example, EPEC-induced ATP release was potentiated by noncytotoxic agents that elevate host cell cAMP, such as forskolin and cholera toxin, and by exposure to hypotonic medium. These findings and the performance of the EPEC espF mutant led us to hypothesize that the CFTR plays a role in EPEC-induced ATP release that is independent of cell death. We report the results of experiments using specific, cell-permeable CFTR activators and inhibitors, as well as transfection of the CFTR into non-CFTR-expressing cell lines, which incriminate the CFTR as a second pathway for ATP release from host cells. Increased ATP release via CFTR is not accompanied by an increase in EPEC adherence to transfected cells. The CFTR-dependent ATP release pathway becomes activated endogenously later in EPEC infection, and this activation is mediated, at least in part, by generation of extracellular adenosine from the breakdown of released ATP.
Collapse
Affiliation(s)
- John K Crane
- Department of Medicine, Division of Infectious Diseases, University of Buffalo, Rm. 317, Biomedical Research Bldg., 3435 Main St., Buffalo, NY 14214, USA.
| | | | | | | |
Collapse
|
37
|
Melo AR, Lasunskaia EB, de Almeida CMC, Schriefer A, Kipnis TL, Dias da Silva W. Expression of the virulence factor, BfpA, by enteropathogenic Escherichia coli is essential for apoptosis signalling but not for NF-kappaB activation in host cells. Scand J Immunol 2005; 61:511-9. [PMID: 15963045 DOI: 10.1111/j.1365-3083.2005.01626.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Localized adherence (LA) of enteropathogenic Escherichia coli (EPEC) to epithelial cells results in attaching and effacing of the surface of these cells. LA depends on the gene bfpA, which codes for the BfpA protein. We found that EPEC-E. coli adherence factor (EAF)((+)), expressing BfpA, significantly reduced HeLa cell viability in comparison with EPEC-EAF((-)), as evaluated by the mitochondrial-dependent succinate dehydrogenase conversion of 3'-[4,5,-dimethylthiazol-2yl]2,5-diphenyltetrazolium bromide (MTT) to its formazan. Apoptosis accounts for a substantial loss of the cell viability, because the cells incubated with EPEC-EAF((+)) or with cloned BfpA (data not shown), but not with EPEC-EAF((-)), were positive for annexin-V binding, demonstrated chromatin condensation and nuclei fragmentation and exhibited a high level of caspase-3 activity. Because the blockade of bacterial cell-surface-associated BfpA by anti-BfpA immunoglobulin (Ig)Y antibody suppressed apoptotic death induced by EPEC-EAF((+)), BfpA may be the trigger for apoptosis. Both EPEC-EAF((+)) and EPEC-EAF((-)), as well as recombinant BfpA (data not shown), activated nuclear factor (NF)-kappaB in a similar manner as analysed by the electrophoretic mobility shift assay (EMSA). EMSA supershift analysis demonstrated the presence of p65/RelA in a DNA-binding complex. In contrast to DNA binding, NF-kappaB-dependent reporter gene transactivation was stimulated more strongly by EPEC B171/EAF((+)), suggesting a role for this virulence factor in the regulation of transcriptional activity of NF-kappaB. Because suppression of NF-kappaB activation by BAY11-7085, a NF-kappaB inhibitor, neither induced apoptosis by itself nor blocked apoptosis induction by EPEC-EAF((+)), it may be suggested that apoptosis is not regulated by the NF-kappaB pathway in HeLa cells.
Collapse
Affiliation(s)
- A R Melo
- Laboratório de Biologia do Reconhecer, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, RJ, Brazil
| | | | | | | | | | | |
Collapse
|
38
|
Malladi V, Shankar B, Williams PH, Balakrishnan A. Enteropathogenic Escherichia coli outer membrane proteins induce changes in cadherin junctions of Caco-2 cells through activation of PKCalpha. Microbes Infect 2005; 6:38-50. [PMID: 14738892 DOI: 10.1016/j.micinf.2003.09.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Enteropathogenic Escherichia coli (EPEC) is a Gram-negative bacterial pathogen that adheres to human intestinal epithelial cells, resulting in watery, persistent diarrhoea. Despite the advances made in understanding EPEC-host cell interactions, the molecular mechanisms underlying watery diarrhoea have not been understood fully. Loss of transepithelial resistance and increased monolayer permeability by disruption of tight junctions has been implicated in this process. Apart from disruption of tight junctions, an important factor known to regulate monolayer permeability is E-cadherin and its interaction with beta-catenin, both of which constitute the adherens junctions. Our previous studies using HEp-2 cells demonstrated the morphological and cytoskeletal changes caused by cell-free outer membrane preparations (OMPs) of EPEC. In this study, we have shown that EPEC and its OMP induce significant changes in the adherens junctions of Caco-2 monolayers. We also observed significant phosphorylation of protein kinase Calpha (PKCalpha) in cells treated with either whole EPEC or its OMP. Immunoprecipitation of cell lysates with anti-E-cadherin and probing with phospho-PKCalpha monoclonal antibodies and anti-beta-catenins revealed that in these cells, phosphorylated PKCalpha is associated with cadherins, leading to the dissociation of the cadherin/beta-catenin complex. Immunofluorescence showed beta-catenins dissociated from the membrane-bound cadherins and redistributed into the cytoplasm. Expression of dominant negative PKCalpha reversed these effects caused by either whole EPEC or its OMP and also reduced the associated increase in monolayer permeability. It is possible that this mechanism may complement the earlier known pathways for loss of barrier function involving myosin light chain kinase activation and also may play a role in causing host cell death by apoptosis.
Collapse
Affiliation(s)
- Vasantha Malladi
- Centre for Biotechnology, Anna University, 600 025, Chennai, India
| | | | | | | |
Collapse
|
39
|
Leverton LQ, Kaper JB. Temporal expression of enteropathogenic Escherichia coli virulence genes in an in vitro model of infection. Infect Immun 2005; 73:1034-43. [PMID: 15664947 PMCID: PMC546935 DOI: 10.1128/iai.73.2.1034-1043.2005] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The hallmark of enteropathogenic Escherichia coli (EPEC) infection is the ability of EPEC to cause attaching and effacing (A/E) lesions on intestinal epithelium. This event is reproducible in in vitro tissue culture models of infection. We used real-time PCR to measure transcription from several locus of enterocyte effacement (LEE) operons (LEE1 to LEE5) and from bfp during a 5-h infection of HEp-2 cells with EPEC. We found that after the initial formation of A/E lesions, which occurs as early as 5 min postinfection, EPEC continues to increase transcription from LEE3 to LEE5 as well as from bfp. These levels are maximized by 3 h postinfection and remain constant throughout the course of infection. This increase in transcription from LEE3 to LEE5 occurs when LEE1 (ler) transcription is decreasing. EspA, EspB, intimin, Tir, and bundle-forming pilus expression is detectable during the entire 5-h infection. These results indicate that the EPEC genes involved in localized and intimate adherence are continually expressed after the initial stages of A/E lesion formation on host cells.
Collapse
Affiliation(s)
- Laura Q Leverton
- Center for Vaccine Development, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | | |
Collapse
|
40
|
Villegas J, Schulz M, Soto L, Sanchez R. Bacteria induce expression of apoptosis in human spermatozoa. Apoptosis 2005; 10:105-10. [PMID: 15711926 DOI: 10.1007/s10495-005-6065-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An increased number of sperm undergoing apoptosis has been observed during inflammatory processes in the male genital tract, which might be associated with elevated reactive oxygen species (ROS) levels. However, another factor to stimulate apoptosis could be the direct contact with bacteria or its products, even in the absence of ROS. The aim of this study was to investigate whether bacteria can directly initiate apoptosis in human spermatozoa. Human spermatozoa selected by density gradient centrifugation were incubated with polymorphonuclear granulocytes (PMN) isolated from blood and/or E. faecalis, E. coli or S. aureus. As ROS inductor in PMN, phorbol-12-myristate-13-acetate was used. After incubating the cells for 60 min at 37 degrees C, ROS were determined by chemiluminescence and phosphatidyl serine (PS) externalization was analyzed by flow cytometry with Annexin V-FITC and propidium iodide (PI). The increase in the percentage of spermatozoa Annexin V-FITC-positive/ PI-negative (early event of late apoptosis) was significant after the incubation with PMN plus PMA, PMN plus E. coli and E. coli alone. The percentage of spermatozoa Annexin V-FITC-positive/ PI-positive (apoptosis/necrosis) increased significantly in sperm incubated with E. coli and S. aureus (20.3% +/- 3 and 13.6% +/- 3.2 compared to sperm alone, 6% +/- 0.5). Sperm incubated with PMN-PMA activated showed only a relative increase in apoptosis/necrosis (8.4% +/- 1). Our results show that bacteria directly increase the PS externalisation in ejaculated human sperm. This way of inducing apoptosis does not require external ROS and may result from anyone of the molecular mechanisms that account for changes in motility, vitality and DNA integrity, that are characteristics of spermatozoa in male genital tract infection.
Collapse
Affiliation(s)
- J Villegas
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Box 54-D, Temuco, Chile
| | | | | | | |
Collapse
|
41
|
Wu Y, Lau B, Smith S, Troyan K, Barnett Foster DE. Enteropathogenic Escherichia coli infection triggers host phospholipid metabolism perturbations. Infect Immun 2004; 72:6764-72. [PMID: 15557596 PMCID: PMC529104 DOI: 10.1128/iai.72.12.6764-6772.2004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) specifically recognizes phosphatidylethanolamine (PE) on the outer leaflet of host epithelial cells. EPEC also induces apoptosis in epithelial cells, which results in increased levels of outer leaflet PE and increased bacterial binding. Consequently, it is of interest to investigate whether EPEC infection perturbs host cell phospholipid metabolism and whether the changes play a role in the apoptotic signaling. Our findings indicate that EPEC infection results in a significant increase in the epithelial cell PE level and a corresponding decrease in the phosphatidylcholine (PC) level. PE synthesis via both the de novo pathway and the serine decarboxylation pathway was enhanced, and de novo synthesis of phosphatidylcholine via CDP-choline was reduced. The changes were transitory, and the maximum change was noted after 4 to 5 h of infection. Addition of exogenous PC or CDP-choline to epithelial cells prior to infection abrogated EPEC-induced apoptosis, suggesting that EPEC infection inhibits the CTP-phosphocholine cytidylyltransferase step in PC synthesis, which is reportedly inhibited during nonmicrobially induced apoptosis. On the other hand, incorporation of exogenous PE by the host cells enhanced EPEC-induced apoptosis and necrosis without increasing bacterial adhesion. This is the first report that pathogen-induced apoptosis is associated with significant changes in PE and PC metabolism, and the results suggest that EPEC adhesion to a host membrane phospholipid plays a role in disruption of host phospholipid metabolism.
Collapse
Affiliation(s)
- Y Wu
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, Canada M5B 2K3
| | | | | | | | | |
Collapse
|
42
|
Nougayrède JP, Donnenberg MS. Enteropathogenic Escherichia coli EspF is targeted to mitochondria and is required to initiate the mitochondrial death pathway. Cell Microbiol 2004; 6:1097-111. [PMID: 15469437 DOI: 10.1111/j.1462-5822.2004.00421.x] [Citation(s) in RCA: 163] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Enteropathogenic Escherichia coli (EPEC) is a causative agent of infant diarrhoea in developing countries. The EspF protein is the product of the espF gene found on the locus of enterocyte effacement, the key pathogenicity island carried by EPEC and enterohemorrhagic E. coli. EspF is injected from adherent EPEC into host cells via a type III secretion system and was previously shown to induce apoptotic cell death and to be required for disruption of host intestinal barrier function. In this work, we show by immunofluorescence and fractionation studies that EspF is targeted to host mitochondria. The N-terminal region of EspF serves as a mitochondrial import signal and, when expressed within cells, can target hybrid green fluorescent protein to mitochondria. Assessment of mitochondrial membrane potential in infected epithelial cells indicated that EspF plays a role in the mitochondrial membrane permeabilization induced by EPEC infection. Furthermore, EspF was associated with the release of cytochrome c from mitochondria into the cytoplasm and with caspase-9 and caspase-3 cleavage. These findings indicate a role for EspF in initiating the mitochondrial death pathway.
Collapse
Affiliation(s)
- Jean-Philippe Nougayrède
- University of Maryland, Baltimore School of Medicine, Division of Infectious Diseases, 10 S. Pine Street, MSTF 900, Baltimore, MD 21201, USA
| | | |
Collapse
|
43
|
Nagai T, Abe A, Sasakawa C. Targeting of enteropathogenic Escherichia coli EspF to host mitochondria is essential for bacterial pathogenesis: critical role of the 16th leucine residue in EspF. J Biol Chem 2004; 280:2998-3011. [PMID: 15533930 DOI: 10.1074/jbc.m411550200] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The attachment of enteropathogenic Escherichia coli (EPEC) to host cells and the induction of attaching and effacing (A/E) lesions are prominent pathogenic features. EPEC infection also leads to host cell death and damage to the intestinal mucosa, which is partly dependent upon EspF, one of the effectors. In this study, we demonstrate that EspF is a mitochondrial import protein with a functional mitochondrial targeting signal (MTS), because EspF activity for importing into the mitochondria was abrogated by MTS deletion mutants. Substitution of the 16th leucine with glutamic acid (EspF(L16E)) completely abolished EspF activity. Infection of HeLa cells with wild type but not the espF mutant (DeltaespF) decreased mitochondrial membrane potential (DeltaPsi(m)), leading to cell death. The DeltaPsi(m) decrease and cell death were restored in cells infected with DeltaespF/pEspF but not DeltaespF/pEspF(L16E), suggesting that the 16th leucine in the MTS is a critical amino acid for EspF function. To demonstrate the impact of EspF in vivo, we exploited Citrobacter rodentium by infecting C3H/HeJ mice with DeltaespF(CR), DeltaespF(CR)/pEspF(CR), or DeltaespF(CR)/pEspF(L16E)(CR). These results indicate that EspF activity contributes to bacterial pathogenesis, as judged by murine lethality and intestinal histopathology, and promotion of bacterial colonization of the intestinal mucosa.
Collapse
Affiliation(s)
- Takeshi Nagai
- Department of Microbiology and Immunity, Institute of Medical Science, University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | | | |
Collapse
|
44
|
Craig L, Pique ME, Tainer JA. Type IV pilus structure and bacterial pathogenicity. Nat Rev Microbiol 2004; 2:363-78. [PMID: 15100690 DOI: 10.1038/nrmicro885] [Citation(s) in RCA: 574] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Lisa Craig
- Department of Molecular Biology and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | |
Collapse
|
45
|
Zhou X, Girón JA, Torres AG, Crawford JA, Negrete E, Vogel SN, Kaper JB. Flagellin of enteropathogenic Escherichia coli stimulates interleukin-8 production in T84 cells. Infect Immun 2003; 71:2120-9. [PMID: 12654834 PMCID: PMC152053 DOI: 10.1128/iai.71.4.2120-2129.2003] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The type III secretion system (TTSS) of enteropathogenic Escherichia coli (EPEC) has been associated with the ability of these bacteria to induce secretion of proinflammatory cytokines, including interleukin-8 (IL-8), in cultured epithelial cells. However, the identity of the effector molecule directly involved in this event is unknown. In this study, we determined that the native flagellar filament and its flagellin monomer are activators of IL-8 release in T84 epithelial cells. Supernatants of wild-type EPEC strain E2348/69 and its isogenic mutants deficient in TTSS (escN) and in production of intimin (eae), grown in Luria-Bertani broth, elicited similar amounts of IL-8 secretion by T84 cells. In contrast, supernatants of EPEC fliC mutants and of B171, a nonflagellated EPEC strain, were defective in inducing IL-8 release, a phenotype that was largely restored by complementation of the fliC gene in the mutant lacking flagella. Purified flagella from E. coli K-12, EPEC serotypes H6 and H34, and enterohemorrhagic E. coli serotype H7 all induced IL-8 release in T84 cells. Induction of IL-8 by purified flagella or His-tagged FliC from EPEC strain E2348/69 was dose dependent and was blocked by a polyclonal anti-H6 antibody. Finally, the mitogen-activated protein kinases (Erk1 and -2 and Jnk) were phosphorylated in flagellin-treated T84 cells, and inhibition of the p38 and Erk pathways significantly decreased the IL-8 response induced by EPEC flagellin. Our data clearly indicate that FliC of EPEC is sufficient to induce IL-8 release in T84 cells and that activation of the Erk and p38 pathways is required for IL-8 induction.
Collapse
Affiliation(s)
- Xin Zhou
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
de Souza Campos Fernandes RC, Quintana Flores VM, Medina-Acosta E. Prevalent transfer of human colostral IgA antibody activity for the enteropathogenic Escherichia coli bundle-forming pilus structural repeating subunit A in neonates. Diagn Microbiol Infect Dis 2002; 44:331-6. [PMID: 12543537 DOI: 10.1016/s0732-8893(02)00463-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The frequency of IgA antibody activity to the structural protein subunit BfpA of the enteropathogenic Escherichia coli bundle-forming pilus was determined in 40 mother-infant pairs by immunoblot analysis using affinity purified recombinant BfpA to monitor for IgA in maternal colostrum and in feces of the neonates. Fecal samples were collected from exclusively breastfed term infants < 24-h after the first breastmilk feeding and colostral samples from their mothers. Infants were monitored prospectively with monthly visits to ascertain dietary practices and diarrheal illnesses. The percentage of colostral anti-BfpA IgA positive patients that were also coproantibody positive was 67.5%. The median duration of lactation was 108 days and the incidence of infantile diarrheal disease was 7.5%. Thus, colostral anti-BfpA IgA antibody activity survives passage through the gut of breastfed neonates, persisting in their feces. It is suggested that oral passive immunotherapy may be used to prevent and/or treat typical EPEC infection during infancy.
Collapse
|
47
|
Hodge S, Hodge G, Flower R, Reynolds PN, Scicchitano R, Holmes M. Up-regulation of production of TGF-beta and IL-4 and down-regulation of IL-6 by apoptotic human bronchial epithelial cells. Immunol Cell Biol 2002; 80:537-43. [PMID: 12406387 DOI: 10.1046/j.1440-1711.2002.01120.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Human bronchial epithelial cells secrete cytokines that play a role in immune responses in the lung. However, the roles of these cytokines in regulating epithelial repair following acute lung injury are largely unknown. Responses to injury include hyperplasia of epithelial cells and squamous metaplasia. The resolution stage is characterized by discontinuation of hyperplasia. Apoptosis is considered to be the most efficient mechanism of removal of unwanted cells without causing inflammation. The presence of TGF-beta1 increases apoptosis, induces squamous metaplasia and inhibits proliferation of airway epithelial cells. Interleukin-4 increases the ability of macrophages to phagocytose epithelial cells and produce inflammatory cytokines. The purpose of this study was to investigate the hypothesis that apoptotic lung epithelial cells produce cytokines, which could act in an autocrine manner to control hyperplasia and induce squamous differentiation following acute lung injury. A bronchial epithelial cell line (16 HBE) was used as an in vitro model, to study the production of TGF-beta, IL-4 and IL-6 by lung epithelial cells undergoing apoptosis. Apoptotic and live cells were sorted on the basis of bright and negative staining with FITC-conjugated Annexin V, respectively. Intracellular IL-6, TGF-beta and IL-4 was measured using flow cytometric techniques. Electron microscopy, immunohistochemistry and ELISA were used as supportive techniques. Apoptotic cells produced significantly more TGF-beta and IL-4 (but less IL-6) than viable cells. Increased production of TGF-beta and IL-4 by epithelial cells undergoing apoptosis may contribute to the inhibition of proliferation, squamous metaplasia, and reduction of the inflammatory response in acute lung injury.
Collapse
Affiliation(s)
- Sandra Hodge
- Department of Thoracic Medicine, Royal Adelaide Hospital, North Adelaide, South Australia, Australia.
| | | | | | | | | | | |
Collapse
|
48
|
Kenny B. Enteropathogenic Escherichia coli (EPEC)-- a crafty subversive little bug. MICROBIOLOGY (READING, ENGLAND) 2002; 148:1967-1978. [PMID: 12101285 DOI: 10.1099/00221287-148-7-1967] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Brendan Kenny
- Department of Pathology and Microbiology, School of Medical Sciences, University Walk, Bristol, BS8 1TD, UK1
| |
Collapse
|
49
|
Quintana Flores VM, Campos de Souza Fernandes RC, Sousa de Macedo Z, Medina-Acosta E. Expression and purification of the recombinant enteropathogenic Escherichia coli vaccine candidates BfpA and EspB. Protein Expr Purif 2002; 25:16-22. [PMID: 12071694 DOI: 10.1006/prep.2001.1604] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BfpA, the structural repeating protein subunit A of the bundle-forming pilus and EspB, a type-III-secreted pore-forming protein of enteropathogenic Escherichia coli (EPEC), both virulence factors central for EPEC pathogenesis, were overexpressed in E. coli DH5alpha and M15 laboratory strains, respectively, using the pQE-30 cloning expression system, as chimeric fusions to a NH(2)-terminal histidine hexapeptide (His(6)-tag) sequence. After isopropyl beta-d-thiogalactoside induction, the expression levels achieved were 11 and 40% of total soluble protein for BfpA and EspB, respectively. The His(6)-tagged recombinant proteins were purified (up to 98% homogeneity) by Ni-agarose affinity chromatography and produced yields varying from 0.65 to 3.1 mg of recombinant protein per gram of wet weight cells. The immunogenicity and antigenicity of the final products were tested in rabbits and using fecal specimens obtained from children suffering from acute watery diarrhea, respectively. The recombinant products correspond to antigenically authentic protein standards, useful in future epidemiological and neonatal vaccinology studies.
Collapse
|