1
|
Vassallo N. Poration of mitochondrial membranes by amyloidogenic peptides and other biological toxins. J Neurochem 2025; 169:e16213. [PMID: 39213385 DOI: 10.1111/jnc.16213] [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: 06/26/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria are essential organelles known to serve broad functions, including in cellular metabolism, calcium buffering, signaling pathways and the regulation of apoptotic cell death. Maintaining the integrity of the outer (OMM) and inner mitochondrial membranes (IMM) is vital for mitochondrial health. Cardiolipin (CL), a unique dimeric glycerophospholipid, is the signature lipid of energy-converting membranes. It plays a significant role in maintaining mitochondrial architecture and function, stabilizing protein complexes and facilitating efficient oxidative phosphorylation (OXPHOS) whilst regulating cytochrome c release from mitochondria. CL is especially enriched in the IMM and at sites of contact between the OMM and IMM. Disorders of protein misfolding, such as Alzheimer's and Parkinson's diseases, involve amyloidogenic peptides like amyloid-β, tau and α-synuclein, which form metastable toxic oligomeric species that interact with biological membranes. Electrophysiological studies have shown that these oligomers form ion-conducting nanopores in membranes mimicking the IMM's phospholipid composition. Poration of mitochondrial membranes disrupts the ionic balance, causing osmotic swelling, loss of the voltage potential across the IMM, release of pro-apoptogenic factors, and leads to cell death. The interaction between CL and amyloid oligomers appears to favour their membrane insertion and pore formation, directly implicating CL in amyloid toxicity. Additionally, pore formation in mitochondrial membranes is not limited to amyloid proteins and peptides; other biological peptides, as diverse as the pro-apoptotic Bcl-2 family members, gasdermin proteins, cobra venom cardiotoxins and bacterial pathogenic toxins, have all been described to punch holes in mitochondria, contributing to cell death processes. Collectively, these findings underscore the vulnerability of mitochondria and the involvement of CL in various pathogenic mechanisms, emphasizing the need for further research on targeting CL-amyloid interactions to mitigate mitochondrial dysfunction.
Collapse
Affiliation(s)
- Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Tal-Qroqq, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Tal-Qroqq, Malta
| |
Collapse
|
2
|
Jarzab M, Skorko-Glonek J. There Are No Insurmountable Barriers: Passage of the Helicobacter pylori VacA Toxin from Bacterial Cytoplasm to Eukaryotic Cell Organelle. MEMBRANES 2023; 14:11. [PMID: 38248700 PMCID: PMC10821523 DOI: 10.3390/membranes14010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 01/23/2024]
Abstract
The Gram-negative bacterium Helicobacter pylori is a very successful pathogen, one of the most commonly identified causes of bacterial infections in humans worldwide. H. pylori produces several virulence factors that contribute to its persistence in the hostile host habitat and to its pathogenicity. The most extensively studied are cytotoxin-associated gene A (CagA) and vacuolating cytotoxin A (VacA). VacA is present in almost all H. pylori strains. As a secreted multifunctional toxin, it assists bacterial colonization, survival, and proliferation during long-lasting infections. To exert its effect on gastric epithelium and other cell types, VacA undergoes several modifications and crosses multiple membrane barriers. Once inside the gastric epithelial cell, VacA disrupts many cellular-signaling pathways and processes, leading mainly to changes in the efflux of various ions, the depolarization of membrane potential, and perturbations in endocytic trafficking and mitochondrial function. The most notable effect of VacA is the formation of vacuole-like structures, which may lead to apoptosis. This review focuses on the processes involved in VacA secretion, processing, and entry into host cells, with a particular emphasis on the interaction of the mature toxin with host membranes and the formation of transmembrane pores.
Collapse
Affiliation(s)
| | - Joanna Skorko-Glonek
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland;
| |
Collapse
|
3
|
Wroblewski LE, Peek RM. Clinical Pathogenesis, Molecular Mechanisms of Gastric Cancer Development. Curr Top Microbiol Immunol 2023; 444:25-52. [PMID: 38231214 PMCID: PMC10924282 DOI: 10.1007/978-3-031-47331-9_2] [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] [Indexed: 01/18/2024]
Abstract
The human pathogen Helicobacter pylori is the strongest known risk factor for gastric disease and cancer, and gastric cancer remains a leading cause of cancer-related death across the globe. Carcinogenic mechanisms associated with H. pylori are multifactorial and are driven by bacterial virulence constituents, host immune responses, environmental factors such as iron and salt, and the microbiota. Infection with strains that harbor the cytotoxin-associated genes (cag) pathogenicity island, which encodes a type IV secretion system (T4SS) confer increased risk for developing more severe gastric diseases. Other important H. pylori virulence factors that augment disease progression include vacuolating cytotoxin A (VacA), specifically type s1m1 vacA alleles, serine protease HtrA, and the outer-membrane adhesins HopQ, BabA, SabA and OipA. Additional risk factors for gastric cancer include dietary factors such as diets that are high in salt or low in iron, H. pylori-induced perturbations of the gastric microbiome, host genetic polymorphisms, and infection with Epstein-Barr virus. This chapter discusses in detail host factors and how H. pylori virulence factors augment the risk of developing gastric cancer in human patients as well as how the Mongolian gerbil model has been used to define mechanisms of H. pylori-induced inflammation and cancer.
Collapse
Affiliation(s)
- Lydia E Wroblewski
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Richard M Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
4
|
Luong P, Li Q, Chen PF, Wrighton PJ, Chang D, Dwyer S, Bayer MT, Snapper SB, Hansen SH, Thiagarajah JR, Goessling W, Lencer WI. A quantitative single-cell assay for retrograde membrane traffic enables rapid detection of defects in cellular organization. Mol Biol Cell 2019; 31:511-519. [PMID: 31774722 PMCID: PMC7202069 DOI: 10.1091/mbc.e19-07-0375] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Retrograde membrane trafficking from plasma membrane to Golgi and endoplasmic reticulum typifies one of the key sorting steps emerging from the early endosome that affects cell surface and intracellular protein dynamics underlying cell function. While some cell surface proteins and lipids are known to sort retrograde, there are few effective methods to quantitatively measure the extent or kinetics of these events. Here we took advantage of the well-known retrograde trafficking of cholera toxin and newly defined split fluorescent protein technology to develop a quantitative, sensitive, and effectively real-time single-cell flow cytometry assay for retrograde membrane transport. The approach can be applied in high throughput to elucidate the underlying biology of membrane traffic and how endosomes adapt to the physiologic needs of different cell types and cell states.
Collapse
Affiliation(s)
- Phi Luong
- Department of Pediatrics, Harvard Medical School, and.,Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115
| | - Qian Li
- Department of Pediatrics, Harvard Medical School, and.,Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115.,Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200000, China
| | - Pin-Fang Chen
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Paul J Wrighton
- Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Denis Chang
- Department of Pediatrics, Harvard Medical School, and.,Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115
| | - Sean Dwyer
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Marie-Theres Bayer
- Department of Pediatrics, Harvard Medical School, and.,Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115
| | - Scott B Snapper
- Department of Pediatrics, Harvard Medical School, and.,Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115.,Harvard Digestive Disease Center, Harvard Medical School, Boston, MA 02115
| | - Steen H Hansen
- Department of Pediatrics, Harvard Medical School, and.,Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115
| | - Jay R Thiagarajah
- Department of Pediatrics, Harvard Medical School, and.,Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115.,Harvard Digestive Disease Center, Harvard Medical School, Boston, MA 02115
| | - Wolfram Goessling
- Harvard Stem Cell Institute, Cambridge, MA 02138.,Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115.,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Wayne I Lencer
- Department of Pediatrics, Harvard Medical School, and.,Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115.,Harvard Digestive Disease Center, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
5
|
Su M, Erwin AL, Campbell AM, Pyburn TM, Salay LE, Hanks JL, Lacy DB, Akey DL, Cover TL, Ohi MD. Cryo-EM Analysis Reveals Structural Basis of Helicobacter pylori VacA Toxin Oligomerization. J Mol Biol 2019; 431:1956-1965. [PMID: 30954575 PMCID: PMC6625667 DOI: 10.1016/j.jmb.2019.03.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori colonizes the human stomach and contributes to the development of gastric cancer and peptic ulcer disease. H. pylori secretes a pore-forming toxin called vacuolating cytotoxin A (VacA), which contains two domains (p33 and p55) and assembles into oligomeric structures. Using single-particle cryo-electron microscopy, we have determined low-resolution structures of a VacA dodecamer and heptamer, as well as a 3.8-Å structure of the VacA hexamer. These analyses show that VacA p88 consists predominantly of a right-handed beta-helix that extends from the p55 domain into the p33 domain. We map the regions of p33 and p55 involved in hexamer assembly, model how interactions between protomers support heptamer formation, and identify surfaces of VacA that likely contact membrane. This work provides structural insights into the process of VacA oligomerization and identifies regions of VacA protomers that are predicted to contact the host cell surface during channel formation.
Collapse
Affiliation(s)
- Min Su
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amanda L Erwin
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anne M Campbell
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37212, USA
| | - Tasia M Pyburn
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Lauren E Salay
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37212, USA
| | - Jessica L Hanks
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - D Borden Lacy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37212, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - David L Akey
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Timothy L Cover
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37212, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37212, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA.
| | - Melanie D Ohi
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
6
|
Linn AK, Samainukul N, Sakdee S, Angsuthanasombat C, Katzenmeier G. A Helicobacter pylori Vacuolating Cytotoxin A: Mouse DHFR Fusion Protein Triggers Dye Release from Liposomes. Curr Microbiol 2017; 75:223-230. [DOI: 10.1007/s00284-017-1369-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 10/09/2017] [Indexed: 01/08/2023]
|
7
|
McClain MS, Beckett AC, Cover TL. Helicobacter pylori Vacuolating Toxin and Gastric Cancer. Toxins (Basel) 2017; 9:toxins9100316. [PMID: 29023421 PMCID: PMC5666363 DOI: 10.3390/toxins9100316] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/03/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori VacA is a channel-forming toxin unrelated to other known bacterial toxins. Most H. pylori strains contain a vacA gene, but there is marked variation among strains in VacA toxin activity. This variation is attributable to strain-specific variations in VacA amino acid sequences, as well as variations in the levels of VacA transcription and secretion. In this review, we discuss epidemiologic studies showing an association between specific vacA allelic types and gastric cancer, as well as studies that have used animal models to investigate VacA activities relevant to gastric cancer. We also discuss the mechanisms by which VacA-induced cellular alterations may contribute to the pathogenesis of gastric cancer.
Collapse
Affiliation(s)
- Mark S McClain
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Amber C Beckett
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Timothy L Cover
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA.
| |
Collapse
|
8
|
Fahimi F, Tohidkia MR, Fouladi M, Aghabeygi R, Samadi N, Omidi Y. Pleiotropic cytotoxicity of VacA toxin in host cells and its impact on immunotherapy. ACTA ACUST UNITED AC 2017; 7:59-71. [PMID: 28546954 PMCID: PMC5439391 DOI: 10.15171/bi.2017.08] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 12/17/2022]
Abstract
![]()
Introduction: In the recent decades, a number of studies have highlighted the importance of Helicobacter pylori in the initiation and development of peptic ulcer and gastric cancer. Some potential virulence factors (e.g., urease, CagA, VacA, BabA) are exploited by this microorganism, facilitating its persistence through evading human defense mechanisms. Among these toxins and enzymes, vacuolating toxin A (VacA) is of a great importance in the pathogenesis of H. pylori. VacA toxin shows different pattern of cytotoxicity through binding to different cell surface receptors in various cells.
Methods: To highlight attempts in treatment for H. pylori infection, here, we discussed the VacA potential as a candidate for development of vaccine and targeted immunotherapy. Furthermore, we reviewed the related literature to provide key insights on association of the genetic variants of VacA with the toxicity of the toxin in cells.
Results: A number of investigations on the receptor(s) binding of VacA toxin confirmed the pleiotropic nature of VacA that uses a unique mechanism for internalization through some membrane components such as lipid rafts and glycophosphatidylinositol (GPI)-anchored proteins (GPI-AP). Considering the high potency of VacA toxin in the clinical presentations in infection and assisting persistence and colonization of H. pylori, it is considered as one of the pivotal components in production vaccines and monoclonal antibodies (mAbs).
Conclusion: It is possible to generate mAbs with a considerable potential to convert into secretory immunoglobulins that could penetrate into the niche of H. pylori and inhibit its normal functionalities. Further, conjugation of H. pylori targeting Ab fragments with the toxic agents or drug delivery systems (DDSs) offers new generation of H. pylori treatments.
Collapse
Affiliation(s)
- Farnaz Fahimi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Fouladi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Aghabeygi
- School of Advanced Biomedical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naser Samadi
- School of Advanced Biomedical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,School of Advanced Biomedical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
Pyburn TM, Foegeding NJ, González-Rivera C, McDonald NA, Gould KL, Cover TL, Ohi MD. Structural organization of membrane-inserted hexamers formed by Helicobacter pylori VacA toxin. Mol Microbiol 2016; 102:22-36. [PMID: 27309820 PMCID: PMC5035229 DOI: 10.1111/mmi.13443] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2016] [Indexed: 01/08/2023]
Abstract
Helicobacter pylori colonizes the human stomach and is a potential cause of peptic ulceration or gastric adenocarcinoma. H. pylori secretes a pore-forming toxin known as vacuolating cytotoxin A (VacA). The 88 kDa secreted VacA protein, composed of an N-terminal p33 domain and a C-terminal p55 domain, assembles into water-soluble oligomers. The structural organization of membrane-bound VacA has not been characterized in any detail and the role(s) of specific VacA domains in membrane binding and insertion are unclear. We show that membrane-bound VacA organizes into hexameric oligomers. Comparison of the two-dimensional averages of membrane-bound and soluble VacA hexamers generated using single particle electron microscopy reveals a structural difference in the central region of the oligomers (corresponding to the p33 domain), suggesting that membrane association triggers a structural change in the p33 domain. Analyses of the isolated p55 domain and VacA variants demonstrate that while the p55 domain can bind membranes, the p33 domain is required for membrane insertion. Surprisingly, neither VacA oligomerization nor the presence of putative transmembrane GXXXG repeats in the p33 domain is required for membrane insertion. These findings provide new insights into the process by which VacA binds and inserts into the lipid bilayer to form membrane channels.
Collapse
Affiliation(s)
- Tasia M Pyburn
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232
| | - Nora J Foegeding
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232
| | - Christian González-Rivera
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232
| | - Nathan A McDonald
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232
| | - Kathleen L Gould
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232
| | - Timothy L Cover
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, 37212
| | - Melanie D Ohi
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232.
- Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232.
| |
Collapse
|
10
|
Mixed Infections of Helicobacter pylori Isolated from Patients with Gastrointestinal Diseases in Taiwan. Gastroenterol Res Pract 2016; 2016:7521913. [PMID: 27738429 PMCID: PMC5055960 DOI: 10.1155/2016/7521913] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/17/2016] [Indexed: 01/26/2023] Open
Abstract
Background. Persistent Helicobacter pylori infection may induce several upper gastrointestinal diseases. Two major virulence factors of H. pylori, vacuolating cytotoxin A (VacA) and cytotoxin-associated gene A (CagA), are thought to be associated with the severity of disease progression. The distribution of vacA and cag-pathogenicity island (cag-PAI) alleles varies in H. pylori isolated from patients in different geographic regions. Aim. To assess the association between mixed infection of H. pylori clinical isolates from Taiwanese patients and the severity of gastrointestinal diseases. Methods. A total of 70 patients were enrolled in this study. Six distinct and well-separated colonies were isolated from each patient and 420 colonies were analyzed to determine the genotypes of virulence genes. Results. The prevalence of mixed infections of all H. pylori-infected patients was 28.6% (20/70). The rate of mixed infections in patients with duodenal ulcer (47.6%) was much higher than that with other gastrointestinal diseases (P < 0.05). Conclusions. H. pylori mixed infections show high genetic diversity that may enhance bacterial adaptation to the hostile environment of the stomach and contribute to disease development.
Collapse
|
11
|
A Nonoligomerizing Mutant Form of Helicobacter pylori VacA Allows Structural Analysis of the p33 Domain. Infect Immun 2016; 84:2662-70. [PMID: 27382020 PMCID: PMC4995914 DOI: 10.1128/iai.00254-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/24/2016] [Indexed: 12/17/2022] Open
Abstract
Helicobacter pylori secretes a pore-forming VacA toxin that has structural features and activities substantially different from those of other known bacterial toxins. VacA can assemble into multiple types of water-soluble flower-shaped oligomeric structures, and most VacA activities are dependent on its capacity to oligomerize. The 88-kDa secreted VacA protein can undergo limited proteolysis to yield two domains, designated p33 and p55. The p33 domain is required for membrane channel formation and intracellular toxic activities, and the p55 domain has an important role in mediating VacA binding to cells. Previous studies showed that the p55 domain has a predominantly β-helical structure, but no structural data are available for the p33 domain. We report here the purification and analysis of a nonoligomerizing mutant form of VacA secreted by H. pylori The nonoligomerizing 88-kDa mutant protein retains the capacity to enter host cells but lacks detectable toxic activity. Analysis of crystals formed by the monomeric protein reveals that the β-helical structure of the p55 domain extends into the C-terminal portion of p33. Fitting the p88 structural model into an electron microscopy map of hexamers formed by wild-type VacA (predicted to be structurally similar to VacA membrane channels) reveals that p55 and the β-helical segment of p33 localize to peripheral arms but do not occupy the central region of the hexamers. We propose that the amino-terminal portion of p33 is unstructured when VacA is in a monomeric form and that it undergoes a conformational change during oligomer assembly.
Collapse
|
12
|
Foegeding NJ, Caston RR, McClain MS, Ohi MD, Cover TL. An Overview of Helicobacter pylori VacA Toxin Biology. Toxins (Basel) 2016; 8:toxins8060173. [PMID: 27271669 PMCID: PMC4926140 DOI: 10.3390/toxins8060173] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/18/2016] [Accepted: 05/27/2016] [Indexed: 12/11/2022] Open
Abstract
The VacA toxin secreted by Helicobacter pylori enhances the ability of the bacteria to colonize the stomach and contributes to the pathogenesis of gastric adenocarcinoma and peptic ulcer disease. The amino acid sequence and structure of VacA are unrelated to corresponding features of other known bacterial toxins. VacA is classified as a pore-forming toxin, and many of its effects on host cells are attributed to formation of channels in intracellular sites. The most extensively studied VacA activity is its capacity to stimulate vacuole formation, but the toxin has many additional effects on host cells. Multiple cell types are susceptible to VacA, including gastric epithelial cells, parietal cells, T cells, and other types of immune cells. This review focuses on the wide range of VacA actions that are detectable in vitro, as well as actions of VacA in vivo that are relevant for H. pylori colonization of the stomach and development of gastric disease.
Collapse
Affiliation(s)
- Nora J Foegeding
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Rhonda R Caston
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Mark S McClain
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Melanie D Ohi
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA.
| | - Timothy L Cover
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA.
| |
Collapse
|
13
|
Cover TL, Holland RL, Blanke SR. Helicobacter pylori Vacuolating Toxin. HELICOBACTER PYLORI RESEARCH 2016:113-141. [DOI: 10.1007/978-4-431-55936-8_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
14
|
Abstract
The autotransporter and two-partner secretion (TPS) pathways are used by E. coli and many other Gram-negative bacteria to delivervirulence factors into the extracellular milieu.Autotransporters arecomprised of an N-terminal extracellular ("passenger") domain and a C-terminal β barrel domain ("β domain") that anchors the protein to the outer membrane and facilitates passenger domain secretion. In the TPS pathway, a secreted polypeptide ("exoprotein") is coordinately expressed with an outer membrane protein that serves as a dedicated transporter. Bothpathways are often grouped together under the heading "type V secretion" because they have many features in common and are used for the secretion of structurally related polypeptides, but it is likely that theyhave distinct evolutionary origins. Although it was proposed many years ago that autotransporterpassenger domains are transported across the outer membrane through a channel formed by the covalently linked β domain, there is increasing evidence that additional factors are involved in the translocation reaction. Furthermore, details of the mechanism of protein secretion through the TPS pathway are only beginning to emerge. In this chapter I discussour current understanding ofboth early and late steps in the biogenesis of polypeptides secreted through type V pathways and current modelsofthe mechanism of secretion.
Collapse
|
15
|
|
16
|
Abstract
Half of the world's population is infected with Helicobacter pylori and approximately 20% of infected individuals develop overt clinical disease such as ulcers and stomach cancer. Paradoxically, despite its classification as a class I carcinogen, H. pylori has been shown to be protective against development of asthma, allergy, and esophageal disease. Given these conflicting roles for H. pylori, researchers are attempting to define the environmental, host, and pathogen interactions that ultimately result in severe disease in some individuals. From the bacterial perspective, the toxins, CagA and VacA, have each been shown to be polymorphic and to contribute to disease in an allele-dependent manner. Based on the notable advances that have recently been made in the CagA field, herein we review recent studies that have begun to shed light on the role of CagA polymorphism in H. pylori disease. Moreover, we discuss the potential interaction of CagA and VacA as a mediator of gastric disease.
Collapse
|
17
|
Lai CH, Hsu YM, Wang HJ, Wang WC. Manipulation of host cholesterol by Helicobacter pylori for their beneficial ecological niche. Biomedicine (Taipei) 2013. [DOI: 10.1016/j.biomed.2012.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
18
|
Böhmer F, Szedlacsek S, Tabernero L, Ostman A, den Hertog J. Protein tyrosine phosphatase structure-function relationships in regulation and pathogenesis. FEBS J 2013; 280:413-31. [PMID: 22682070 DOI: 10.1111/j.1742-4658.2012.08655.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Protein phosphorylation on tyrosine residues is tightly controlled by protein tyrosine phosphatases (PTPs) at multiple levels: spatio-temporal expression, subcellular localization and post-translational modification. Structural and functional analysis of the PTP domains has provided insight into catalysis and regulatory mechanisms that control the enzymatic activity. Understanding the molecular basis of PTP regulation is of fundamental importance to dissect the pleiotropic effect of these enzymes in both health and disease. Here, we review recent insights into the regulation of receptor-like PTPs by extracellular ligands and into regulation by reversible oxidation that impairs catalysis directly. The physiological roles of PTPs are essential in homeostasis in eukaryotic cells and pertubation of their functional attributes causes different disease states. As an example, we discuss recent findings indicating how inappropriate oxidation of PTPs in cancer cells may contribute to cell transformation. On the other hand, PTPs from many pathogens are key virulence factors and manipulate signalling pathways in the host cells to promote invasion and survival of the microorganisms. This research area has received relatively little attention but has advanced remarkably. We review the structural features of pathogenic PTPs, their similarities and differences with eukaryotic PTPs, and the possible exploitation of this knowledge for therapeutic intervention.
Collapse
Affiliation(s)
- Frank Böhmer
- Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | | | | | | | | |
Collapse
|
19
|
Chambers MG, Pyburn TM, González-Rivera C, Collier SE, Eli I, Yip CK, Takizawa Y, Lacy DB, Cover TL, Ohi MD. Structural analysis of the oligomeric states of Helicobacter pylori VacA toxin. J Mol Biol 2012. [PMID: 23178866 DOI: 10.1016/j.jmb.2012.11.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori is a Gram-negative bacterium that colonizes the human stomach and contributes to peptic ulceration and gastric adenocarcinoma. H. pylori secretes a pore-forming exotoxin known as vacuolating toxin (VacA). VacA contains two distinct domains, designated p33 and p55, and assembles into large "snowflake"-shaped oligomers. Thus far, no structural data are available for the p33 domain, which is essential for membrane channel formation. Using single-particle electron microscopy and the random conical tilt approach, we have determined the three-dimensional structures of six VacA oligomeric conformations at ~15-Å resolution. The p55 domain, composed primarily of β-helical structures, localizes to the peripheral arms, while the p33 domain consists of two globular densities that localize within the center of the complexes. By fitting the VacA p55 crystal structure into the electron microscopy densities, we have mapped inter-VacA interactions that support oligomerization. In addition, we have examined VacA variants/mutants that differ from wild-type (WT) VacA in toxin activity and/or oligomeric structural features. Oligomers formed by VacA∆6-27, a mutant that fails to form membrane channels, lack an organized p33 central core. Mixed oligomers containing both WT and VacA∆6-27 subunits also lack an organized core. Oligomers formed by a VacA s2m1 chimera (which lacks cell-vacuolating activity) and VacAΔ301-328 (which retains vacuolating activity) each contain p33 central cores similar to those of WT oligomers. By providing the most detailed view of the VacA structure to date, these data offer new insights into the toxin's channel-forming component and the intermolecular interactions that underlie oligomeric assembly.
Collapse
Affiliation(s)
- Melissa G Chambers
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
González-Rivera C, Algood HMS, Radin JN, McClain MS, Cover TL. The intermediate region of Helicobacter pylori VacA is a determinant of toxin potency in a Jurkat T cell assay. Infect Immun 2012; 80:2578-88. [PMID: 22585965 PMCID: PMC3434591 DOI: 10.1128/iai.00052-12] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 05/04/2012] [Indexed: 02/08/2023] Open
Abstract
Colonization of the human stomach with Helicobacter pylori is a risk factor for peptic ulceration, noncardia gastric adenocarcinoma, and gastric lymphoma. The secreted VacA toxin is an important H. pylori virulence factor that causes multiple alterations in gastric epithelial cells and T cells. Several families of vacA alleles have been described, and H. pylori strains containing certain vacA types (s1, i1, and m1) are associated with an increased risk of gastric disease, compared to strains containing other vacA types (s2, i2, and m2). Thus far, there has been relatively little study of the role of the VacA intermediate region (i-region) in toxin activity. In this study, we compared the ability of i1 and i2 forms of VacA to cause functional alterations in Jurkat cells. To do this, we manipulated the chromosomal vacA gene in two H. pylori strains to introduce alterations in the region encoding the VacA i-region. We did not detect any differences in the capacity of i1 and i2 forms of VacA to cause vacuolation of RK13 cells. In comparison to i1 forms of VacA, i2 forms of VacA had a diminished capacity to inhibit the activation of nuclear factor of activated T cells (NFAT) and suppress interleukin-2 (IL-2) production. Correspondingly, i2 forms of VacA bound to Jurkat cells less avidly than did i1 forms of VacA. These results indicate that the VacA i-region is an important determinant of VacA effects on human T cell function.
Collapse
Affiliation(s)
| | - Holly M. Scott Algood
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jana N. Radin
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Mark S. McClain
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Timothy L. Cover
- Department of Pathology, Microbiology and Immunology
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
21
|
Palframan SL, Kwok T, Gabriel K. Vacuolating cytotoxin A (VacA), a key toxin for Helicobacter pylori pathogenesis. Front Cell Infect Microbiol 2012; 2:92. [PMID: 22919683 PMCID: PMC3417644 DOI: 10.3389/fcimb.2012.00092] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 06/18/2012] [Indexed: 12/15/2022] Open
Abstract
More than 50% of the world's population is infected with Helicobacter pylori (H. pylori). Chronic infection with this Gram-negative pathogen is associated with the development of peptic ulcers and is linked to an increased risk of gastric cancer. H. pylori secretes many proteinaceous factors that are important for initial colonization and subsequent persistence in the host stomach. One of the major protein toxins secreted by H. pylori is the Vacuolating cytotoxin A (VacA). After secretion from the bacteria via a type V autotransport secretion system, the 88 kDa VacA toxin (comprised of the p33 and p55 subunits) binds to host cells and is internalized, causing severe “vacuolation” characterized by the accumulation of large vesicles that possess hallmarks of both late endosomes and early lysosomes. The development of “vacuoles” has been attributed to the formation of VacA anion-selective channels in membranes. Apart from its vacuolating effects, it has recently become clear that VacA also directly affects mitochondrial function. Earlier studies suggested that the p33 subunit, but not the p55 subunit of VacA, could enter mitochondria to modulate organelle function. This raised the possibility that a mechanism separate from pore formation may be responsible for the effects of VacA on mitochondria, as crystallography studies and structural modeling predict that both subunits are required for a physiologically stable pore. It has also been suggested that the mitochondrial effects observed are due to indirect effects on pro-apoptotic proteins and direct effects on mitochondrial morphology-related processes. Other studies have shown that both the p55 and p33 subunits can indeed be efficiently imported into mammalian-derived mitochondria raising the possibility that they could re-assemble to form a pore. Our review summarizes and consolidates the recent advances in VacA toxin research, with focus on the outstanding controversies in the field and the key remaining questions that need to be addressed.
Collapse
Affiliation(s)
- Samuel L Palframan
- Host Pathogens Molecular Biology Group, Department of Biochemistry and Molecular Biology, Monash University, Clayton VIC, Australia
| | | | | |
Collapse
|
22
|
Kim IJ, Blanke SR. Remodeling the host environment: modulation of the gastric epithelium by the Helicobacter pylori vacuolating toxin (VacA). Front Cell Infect Microbiol 2012; 2:37. [PMID: 22919629 PMCID: PMC3417592 DOI: 10.3389/fcimb.2012.00037] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 03/05/2012] [Indexed: 12/13/2022] Open
Abstract
Virulence mechanisms underlying Helicobacter pylori persistence and disease remain poorly understood, in part, because the factors underlying disease risk are multifactorial and complex. Among the bacterial factors that contribute to the cumulative pathophysiology associated with H. pylori infections, the vacuolating cytotoxin (VacA) is one of the most important. Analogous to a number of H. pylori genes, the vacA gene exhibits allelic mosaicism, and human epidemiological studies have revealed that several families of toxin alleles are predictive of more severe disease. Animal model studies suggest that VacA may contribute to pathogenesis in several ways. VacA functions as an intracellular-acting protein exotoxin. However, VacA does not fit the current prototype of AB intracellular-acting bacterial toxins, which elaborate modulatory effects through the action of an enzymatic domain translocated inside host cells. Rather, VacA may represent an alternative prototype for AB intracellular acting toxins that modulate cellular homeostasis by forming ion-conducting intracellular membrane channels. Although VacA seems to form channels in several different membranes, one of the most important target sites is the mitochondrial inner membrane. VacA apparently take advantage of an unusual intracellular trafficking pathway to mitochondria, where the toxin is imported and depolarizes the inner membrane to disrupt mitochondrial dynamics and cellular energy homeostasis as a mechanism for engaging the apoptotic machinery within host cells. VacA remodeling of the gastric environment appears to be fine-tuned through the action of the Type IV effector protein CagA which, in part, limits the cytotoxic effects of VacA in cells colonized by H. pylori.
Collapse
Affiliation(s)
- Ik-Jung Kim
- Department of Microbiology, Institute for Genomic Biology, University of Illinois, Urbana IL, USA
| | | |
Collapse
|
23
|
Rassow J. Helicobacter pylori vacuolating toxin A and apoptosis. Cell Commun Signal 2011; 9:26. [PMID: 22044628 PMCID: PMC3266207 DOI: 10.1186/1478-811x-9-26] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 11/01/2011] [Indexed: 12/16/2022] Open
Abstract
VacA, the vacuolating cytotoxin A of Helicobacter pylori, induces apoptosis in epithelial cells of the gastic mucosa and in leukocytes. VacA is released by the bacteria as a protein of 88 kDa. At the outer surface of host cells, it binds to the sphingomyelin of lipid rafts. At least partially, binding to the cells is facilitated by different receptor proteins. VacA is internalized by a clathrin-independent mechanism and initially accumulates in GPI-anchored proteins-enriched early endosomal compartments. Together with early endosomes, VacA is distributed inside the cells. Most of the VacA is eventually contained in the membranes of vacuoles. VacA assembles in hexameric oligomers forming an anion channel of low conductivity with a preference for chloride ions. In parallel, a significant fraction of VacA can be transferred from endosomes to mitochondria in a process involving direct endosome-mitochondria juxtaposition. Inside the mitochondria, VacA accumulates in the mitochondrial inner membrane, probably forming similar chloride channels as observed in the vacuoles. Import into mitochondria is mediated by the hydrophobic N-terminus of VacA. Apoptosis is triggered by loss of the mitochondrial membrane potential, recruitment of Bax and Bak, and release of cytochrome c.
Collapse
Affiliation(s)
- Joachim Rassow
- Ruhr-Universität Bochum, Institut für Physiologische Chemie, Medizinische Fakultät, Gebäude MA3, D-44780 Bochum, Germany.
| |
Collapse
|
24
|
The Human Gastric Pathogen Helicobacter pylori and Its Association with Gastric Cancer and Ulcer Disease. ACTA ACUST UNITED AC 2011. [DOI: 10.1155/2011/340157] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
With the momentous discovery in the 1980's that a bacterium, Helicobacter pylori, can cause peptic ulcer disease and gastric cancer, antibiotic therapies and prophylactic measures have been successful, only in part, in reducing the global burden of these diseases. To date, ~700,000 deaths worldwide are still attributable annually to gastric cancer alone. Here, we review H. pylori's contribution to the epidemiology and histopathology of both gastric cancer and peptic ulcer disease. Furthermore, we examine the host-pathogen relationship and H. pylori biology in context of these diseases, focusing on strain differences, virulence factors (CagA and VacA), immune activation and the challenges posed by resistance to existing therapies. We consider also the important role of host-genetic variants, for example, in inflammatory response genes, in determining infection outcome and the role of H. pylori in other pathologies—some accepted, for example, MALT lymphoma, and others more controversial, for example, idiopathic thrombocytic purpura. More recently, intriguing suggestions that H. pylori has protective effects in GERD and autoimmune diseases, such as asthma, have gained momentum. Therefore, we consider the basis for these suggestions and discuss the potential impact for future therapeutic rationales.
Collapse
|
25
|
Jones KR, Whitmire JM, Merrell DS. A Tale of Two Toxins: Helicobacter Pylori CagA and VacA Modulate Host Pathways that Impact Disease. Front Microbiol 2010; 1:115. [PMID: 21687723 PMCID: PMC3109773 DOI: 10.3389/fmicb.2010.00115] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 09/27/2010] [Indexed: 12/16/2022] Open
Abstract
Helicobacter pylori is a pathogenic bacterium that colonizes more than 50% of the world's population, which leads to a tremendous medical burden. H. pylori infection is associated with such varied diseases as gastritis, peptic ulcers, and two forms of gastric cancer: gastric adenocarcinoma and mucosa-associated lymphoid tissue lymphoma. This association represents a novel paradigm for cancer development; H. pylori is currently the only bacterium to be recognized as a carcinogen. Therefore, a significant amount of research has been conducted to identify the bacterial factors and the deregulated host cell pathways that are responsible for the progression to more severe disease states. Two of the virulence factors that have been implicated in this process are cytotoxin-associated gene A (CagA) and vacuolating cytotoxin A (VacA), which are cytotoxins that are injected and secreted by H. pylori, respectively. Both of these virulence factors are polymorphic and affect a multitude of host cellular pathways. These combined facts could easily contribute to differences in disease severity across the population as various CagA and VacA alleles differentially target some pathways. Herein we highlight the diverse types of cellular pathways and processes targeted by these important toxins.
Collapse
Affiliation(s)
- Kathleen R Jones
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | | | | |
Collapse
|
26
|
González-Rivera C, Gangwer KA, McClain MS, Eli IM, Chambers MG, Ohi MD, Lacy DB, Cover TL. Reconstitution of Helicobacter pylori VacA toxin from purified components. Biochemistry 2010; 49:5743-52. [PMID: 20527875 PMCID: PMC2910095 DOI: 10.1021/bi100618g] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Helicobacter pylori VacA is a pore-forming toxin that causes multiple alterations in human cells and contributes to the pathogenesis of peptic ulcer disease and gastric cancer. The toxin is secreted by H. pylori as an 88 kDa monomer (p88) consisting of two domains (p33 and p55). While an X-ray crystal structure for p55 exists and p88 oligomers have been visualized by cryo-electron microscopy, a detailed analysis of p33 has been hindered by an inability to purify this domain in an active form. In this study, we expressed and purified a recombinant form of p33 under denaturing conditions and optimized conditions for the refolding of the soluble protein. We show that refolded p33 can be added to purified p55 in trans to cause vacuolation of HeLa cells and inhibition of IL-2 production by Jurkat cells, effects identical to those produced by the p88 toxin from H. pylori. The p33 protein markedly enhances the cell binding properties of p55. Size exclusion chromatography experiments suggest that p33 and p55 assemble into a complex consistent with the size of a p88 monomer. Electron microscopy of these p33/p55 complexes reveals small rod-shaped structures that can convert to oligomeric flower-shaped structures in the presence of detergent. We propose that the oligomerization observed in these experiments mimics the process by which VacA oligomerizes when in contact with membranes of host cells.
Collapse
Affiliation(s)
- Christian González-Rivera
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Kelly A. Gangwer
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Mark S. McClain
- Department of Medicine Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Ilyas M. Eli
- Department of Cell and Developmental Biology Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Melissa G. Chambers
- Department of Cell and Developmental Biology Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Melanie D. Ohi
- Department of Cell and Developmental Biology Vanderbilt University School of Medicine, Nashville, TN 37232
| | - D. Borden Lacy
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Timothy L. Cover
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
- Department of Medicine Vanderbilt University School of Medicine, Nashville, TN 37232
- Department of Veterans Affairs Tennessee Valley Healthcare System Nashville, TN 37212
| |
Collapse
|
27
|
Domańska G, Motz C, Meinecke M, Harsman A, Papatheodorou P, Reljic B, Dian-Lothrop EA, Galmiche A, Kepp O, Becker L, Günnewig K, Wagner R, Rassow J. Helicobacter pylori VacA toxin/subunit p34: targeting of an anion channel to the inner mitochondrial membrane. PLoS Pathog 2010; 6:e1000878. [PMID: 20442789 PMCID: PMC2861713 DOI: 10.1371/journal.ppat.1000878] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 03/25/2010] [Indexed: 12/13/2022] Open
Abstract
The vacuolating toxin VacA, released by Helicobacter pylori, is an important virulence factor in the pathogenesis of gastritis and gastroduodenal ulcers. VacA contains two subunits: The p58 subunit mediates entry into target cells, and the p34 subunit mediates targeting to mitochondria and is essential for toxicity. In this study we found that targeting to mitochondria is dependent on a unique signal sequence of 32 uncharged amino acid residues at the p34 N-terminus. Mitochondrial import of p34 is mediated by the import receptor Tom20 and the import channel of the outer membrane TOM complex, leading to insertion of p34 into the mitochondrial inner membrane. p34 assembles in homo-hexamers of extraordinary high stability. CD spectra of the purified protein indicate a content of >40% beta-strands, similar to pore-forming beta-barrel proteins. p34 forms an anion channel with a conductivity of about 12 pS in 1.5 M KCl buffer. Oligomerization and channel formation are independent both of the 32 uncharged N-terminal residues and of the p58 subunit of the toxin. The conductivity is efficiently blocked by 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB), a reagent known to inhibit VacA-mediated apoptosis. We conclude that p34 essentially acts as a small pore-forming toxin, targeted to the mitochondrial inner membrane by a special hydrophobic N-terminal signal.
Collapse
Affiliation(s)
- Grażyna Domańska
- Institut für Physiologische Chemie, Ruhr-Universität Bochum, Bochum, Germany
| | - Christian Motz
- Institut für Physiologische Chemie, Ruhr-Universität Bochum, Bochum, Germany
| | - Michael Meinecke
- Institut für Biophysik, Universität Osnabrück, Osnabrück, Germany
| | - Anke Harsman
- Institut für Biophysik, Universität Osnabrück, Osnabrück, Germany
| | | | - Boris Reljic
- Institut für Physiologische Chemie, Ruhr-Universität Bochum, Bochum, Germany
| | | | - Antoine Galmiche
- Laboratoire de Biochimie, INSERM ERI12, Hopital Nord, CHU Amiens Picardie, Amiens, France
| | - Oliver Kepp
- INSERM U848, Institute Gustave Roussy, Université Paris Sud, Villejuif, France
| | - Lars Becker
- Institut für Biophysik, Universität Osnabrück, Osnabrück, Germany
| | - Kathrin Günnewig
- Institut für Physiologische Chemie, Ruhr-Universität Bochum, Bochum, Germany
| | - Richard Wagner
- Institut für Biophysik, Universität Osnabrück, Osnabrück, Germany
| | - Joachim Rassow
- Institut für Physiologische Chemie, Ruhr-Universität Bochum, Bochum, Germany
- * E-mail:
| |
Collapse
|
28
|
Israel DA, Peek RM. Surreptitious manipulation of the human host by Helicobacter pylori. Gut Microbes 2010; 1:119-127. [PMID: 20976041 PMCID: PMC2958064 DOI: 10.4161/gmic.1.2.11991] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 04/06/2010] [Accepted: 04/08/2010] [Indexed: 02/03/2023] Open
Abstract
Microbial pathogens contribute to the development of more than 1 million cases of cancer per year. Gastric adenocarcinoma is the second leading cause of cancer-related death in the world, and gastritis induced by Helicobacter pylori is the strongest known risk factor for this malignancy. H. pylori colonizes the stomach for years, not days or weeks, as is usually the case for bacterial pathogens and it always induces inflammation; however, only a fraction of colonized individuals ever develop disease. Identification of mechanisms through which H. pylori co-opts host defenses to facilitate its own persistence will not only improve diagnostic and therapeutic modalities, but may also provide insights into other diseases that arise within the context of long-term pathogen-initiated inflammatory states, such as chronic viral hepatitis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Dawn A Israel
- Department of Medicine; Division of Gastroenterology, Nashville, TN USA
| | | |
Collapse
|
29
|
Backert S, Tegtmeyer N. the versatility of the Helicobacter pylori vacuolating cytotoxin vacA in signal transduction and molecular crosstalk. Toxins (Basel) 2010; 2:69-92. [PMID: 22069547 PMCID: PMC3206623 DOI: 10.3390/toxins2010069] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Revised: 12/31/2009] [Accepted: 01/14/2010] [Indexed: 12/13/2022] Open
Abstract
By modulating important properties of eukaryotic cells, many bacterial protein toxins highjack host signalling pathways to create a suitable niche for the pathogen to colonize and persist. Helicobacter pylori VacA is paradigm of pore-forming toxins which contributes to the pathogenesis of peptic ulceration. Several cellular receptors have been described for VacA, which exert different effects on epithelial and immune cells. The crystal structure of VacA p55 subunit might be important for elucidating details of receptor interaction and pore formation. Here we discuss the multiple signalling activities of this important toxin and the molecular crosstalk between VacA and other virulence factors.
Collapse
Affiliation(s)
- Steffen Backert
- Ardmore House, School of Biomolecular and Biomedical Sciences, Belfield Campus, University College Dublin, Dublin-4, Ireland.
| | | |
Collapse
|
30
|
Chlamydia trachomatis polymorphic membrane protein D is an oligomeric autotransporter with a higher-order structure. Infect Immun 2008; 77:508-16. [PMID: 19001072 DOI: 10.1128/iai.01173-08] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Chlamydia trachomatis is a globally important obligate intracellular bacterial pathogen that is a leading cause of sexually transmitted disease and blinding trachoma. Effective control of these diseases will likely require a preventative vaccine. C. trachomatis polymorphic membrane protein D (PmpD) is an attractive vaccine candidate as it is conserved among C. trachomatis strains and is a target of broadly cross-reactive neutralizing antibodies. We show here that immunoaffinity-purified native PmpD exists as an oligomer with a distinct 23-nm flower-like structure. Two-dimensional blue native-sodium dodecyl sulfate-polyacrylamide gel electrophoresis analyses showed that the oligomers were composed of full-length PmpD (p155) and two proteolytically processed fragments, the p73 passenger domain (PD) and the p82 translocator domain. We also show that PmpD undergoes an infection-dependent proteolytic processing step late in the growth cycle that yields a soluble extended PD (p111) that was processed into a p73 PD and a novel p30 fragment. Interestingly, soluble PmpD peptides possess putative eukaryote-interacting functional motifs, implying potential secondary functions within or distal to infected cells. Collectively, our findings show that PmpD exists as two distinct forms, a surface-associated oligomer exhibiting a higher-order flower-like structure and a soluble form restricted to infected cells. We hypothesize that PmpD is a multifunctional virulence factor important in chlamydial pathogenesis and could represent novel vaccine or drug targets for the control of human chlamydial infections.
Collapse
|
31
|
Lu WM, Shen H, Yan SH, Yang XW, Zhu XX, Miao JP, Shan ZW. Effects of rabbit serum containing Yiqi Qingre Formula and its decomposed recipes on type I Helicobacter pylori-induced apoptosis of GES-1 cells in vitro. Shijie Huaren Xiaohua Zazhi 2008; 16:3026-3030. [DOI: 10.11569/wcjd.v16.i27.3026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the effects of rabbit serum containing Qi-replenishing heat-clearing formula (Yiqi Qingre Formula) and its decomposed recipes on type ⅠHelicobacter pylori (H pylori) -induced GES-1 cell apoptosis and to explore its mechanism.
METHODS: Rabbit sera saline, containing amoxicillin, Yiqi Qingre Formula, Huangqi or Huangqin were prepared. The model of type Ⅰ H pylori-induced GES-1 cell injury was established and co-cultured with the individually complete culture fluid of 100 mL/L rabbit serum for 48 h. Then, cells were collected and fixed, followed by identification of apoptosis using flow cytometry and Hoechst33258 fluorescent staining.
RESULTS: Apoptosis rate of type ⅠH pylori-infected GES-1 cells remarkedly increased in the saline group, while it was decreased either by amoxicillin, Yiqi Qingre Formula, Huangqi or Huangqin (1.3633 ± 0.4229, 2.1925 ± 0.6779, 1.7967 ± 0.6987, 1.4740 ± 0.4156 vs 16.6229 ± 8.5087, P < 0.01 or 0.05). The coincidence rate determined using flow cytometry and Hoechst33258 fluorescent staining was high.
CONCLUSION: Yiqi Qingre Formula has the effects of alleviating type ⅠH pylori-induced gastric epithelial cell injury through inhibition of cell apoptosis, thus maintaining cell growth.
Collapse
|
32
|
Dautin N, Bernstein HD. Protein secretion in gram-negative bacteria via the autotransporter pathway. Annu Rev Microbiol 2007; 61:89-112. [PMID: 17506669 DOI: 10.1146/annurev.micro.61.080706.093233] [Citation(s) in RCA: 219] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Autotransporters are a large and diverse superfamily of proteins produced by pathogenic gram-negative bacteria that are composed of an N-terminal passenger domain, which typically harbors a virulence function, and a C-terminal beta domain. It has long been known that the beta domain anchors the protein to the outer membrane and facilitates transport of the passenger domain into the extracellular space. Despite the apparent simplicity of the autotransporter pathway, several aspects of autotransporter biogenesis remain poorly understood, most notably the mechanism by which the passenger domain is translocated across the outer membrane. Here we review recent evidence that the enormous sequence diversity of both passenger and beta domains belies a remarkable conservation of structure. We also discuss insights into each stage of autotransporter biogenesis that have emerged from recent structural, biochemical, and imaging studies.
Collapse
Affiliation(s)
- Nathalie Dautin
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0538, USA.
| | | |
Collapse
|
33
|
Torres VJ, VanCompernolle SE, Sundrud MS, Unutmaz D, Cover TL. Helicobacter pylori vacuolating cytotoxin inhibits activation-induced proliferation of human T and B lymphocyte subsets. THE JOURNAL OF IMMUNOLOGY 2007; 179:5433-40. [PMID: 17911630 DOI: 10.4049/jimmunol.179.8.5433] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Helicobacter pylori are Gram-negative bacteria that persistently colonize the human gastric mucosa despite the recruitment of immune cells. The H. pylori vacuolating cytotoxin (VacA) recently has been shown to inhibit stimulation-induced proliferation of primary human CD4(+) T cells. In this study, we investigated effects of VacA on the proliferation of various other types of primary human immune cells. Intoxication of PBMC with VacA inhibited the stimulation-induced proliferation of CD4(+) T cells, CD8(+) T cells, and B cells. VacA also inhibited the proliferation of purified primary human CD4(+) T cells that were stimulated by dendritic cells. VacA inhibited both T cell-induced and PMA/anti-IgM-induced proliferation of purified B cells. Intoxication with VacA did not alter the magnitude of calcium flux that occurred upon stimulation of CD4(+) T cells or B cells, indicating that VacA does not alter early signaling events required for activation and proliferation. VacA reduced the mitochondrial membrane potential of CD4(+) T cells, but did not reduce the mitochondrial membrane potential of B cells. We propose that the immunomodulatory actions of VacA on T and B lymphocytes, the major effectors of the adaptive immune response, may contribute to the ability of H. pylori to establish a persistent infection in the human gastric mucosa.
Collapse
Affiliation(s)
- Victor J Torres
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232-2605, USA
| | | | | | | | | |
Collapse
|
34
|
Gangwer KA, Mushrush DJ, Stauff DL, Spiller B, McClain MS, Cover TL, Lacy DB. Crystal structure of the Helicobacter pylori vacuolating toxin p55 domain. Proc Natl Acad Sci U S A 2007; 104:16293-8. [PMID: 17911250 PMCID: PMC2042200 DOI: 10.1073/pnas.0707447104] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Helicobacter pylori VacA, a pore-forming toxin secreted by an autotransporter pathway, causes multiple alterations in human cells, contributes to the pathogenesis of peptic ulcer disease and gastric cancer, and is a candidate antigen for inclusion in an H. pylori vaccine. Here, we present a 2.4-A crystal structure of the VacA p55 domain, which has an important role in mediating VacA binding to host cells. The structure is predominantly a right-handed parallel beta-helix, a feature that is characteristic of autotransporter passenger domains but unique among known bacterial protein toxins. Notable features of VacA p55 include disruptions in beta-sheet contacts that result in five beta-helix subdomains and a C-terminal domain that contains a disulfide bond. Analysis of VacA protein sequences from unrelated H. pylori strains, including m1 and m2 forms of VacA, allows us to identify structural features of the VacA surface that may be important for interactions with host receptors. Docking of the p55 structure into a 19-A cryo-EM map of a VacA dodecamer allows us to propose a model for how VacA monomers assemble into oligomeric structures capable of membrane channel formation.
Collapse
Affiliation(s)
- Kelly A. Gangwer
- Departments of *Microbiology and Immunology
- Center for Structural Biology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | - Darren J. Mushrush
- Biochemistry
- Center for Structural Biology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | | | - Ben Spiller
- Departments of *Microbiology and Immunology
- Pharmacology, and
- Center for Structural Biology, Vanderbilt University Medical Center, Nashville, TN 37232; and
| | | | - Timothy L. Cover
- Departments of *Microbiology and Immunology
- Medicine and
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212
| | - D. Borden Lacy
- Departments of *Microbiology and Immunology
- Biochemistry
- Center for Structural Biology, Vanderbilt University Medical Center, Nashville, TN 37232; and
- **To whom correspondence should be addressed. E-mail:
| |
Collapse
|
35
|
Rhead JL, Letley DP, Mohammadi M, Hussein N, Mohagheghi MA, Eshagh Hosseini M, Atherton JC. A new Helicobacter pylori vacuolating cytotoxin determinant, the intermediate region, is associated with gastric cancer. Gastroenterology 2007; 133:926-36. [PMID: 17854597 DOI: 10.1053/j.gastro.2007.06.056] [Citation(s) in RCA: 311] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Accepted: 06/11/2007] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Helicobacter pylori is the main cause of peptic ulceration and gastric adenocarcinoma. The vacuolating cytotoxin gene, vacA, is a major determinant of virulence. Two naturally polymorphic sites in vacA, the signal region and midregion, are well-characterized determinants of toxicity and markers of pathogenesis. The aim of this study was to characterize a new vacA polymorphic site, the intermediate (i) region. METHODS The vacA i-region was identified and characterized by constructing isogenic vacA exchange mutants and determining their vacuolating activity on HeLa, AGS, and RK13 cell lines. The vacA i-region types of H pylori isolates from patients undergoing routine endoscopy were determined by nucleotide sequencing and allele-specific polymerase chain reaction. RESULTS Two i-region types were identified, i1 and i2, and both were common among 42 Western clinical isolates. Interestingly, only naturally occurring s1/m2 strains varied in i-type; s1/m1 and s2/m2 strains were exclusively i1 and i2, respectively. Vacuolation assays showed that i-type determined vacuolating activity among these s1/m2 strains, and exchange mutagenesis confirmed that the i-region itself was directly responsible. Using a simple i-region polymerase chain reaction-based typing system, it was shown for 73 Iranian patients that i1-type strains were strongly associated with gastric adenocarcinoma (P < 10(-3)). Finally, logistic regression analysis showed this association to be independent of, and larger than, associations of vacA s- or m-type or cag status with gastric adenocarcinoma. CONCLUSIONS Together these data show that the vacA i-region is an important determinant of H pylori toxicity and the best independent marker of VacA-associated pathogenicity.
Collapse
Affiliation(s)
- Joanne L Rhead
- Wolfson Digestive Diseases Centre and Institute of Infection, Immunity and Inflammation, University of Nottingham, Nottingham, England
| | | | | | | | | | | | | |
Collapse
|
36
|
D'Elios MM, Montecucco C, de Bernard M. VacA and HP-NAP, Ying and Yang of Helicobacter pylori-associated gastric inflammation. Clin Chim Acta 2007; 381:32-8. [PMID: 17368441 DOI: 10.1016/j.cca.2007.02.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Accepted: 02/13/2007] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori is a Gram-negative bacterium which infects almost half of the population worldwide and represents the major cause of gastroduodenal pathologies, such as duodenal and gastric ulcer, gastric cancer, B-cell lymphoma of mucosa-associated lymphoid tissue (MALT) and autoimmune gastritis. H. pylori colonization is followed by infiltration of the gastric mucosa by polymorphonuclear cells, macrophages and lymphocytes. Two of the major H. pylori virulence factors are the vacuolating cytotoxin (VacA) and the H. pylori neutrophil-activating protein (HP-NAP). VacA has been proposed as a modulator of immune cell function because of its capacity to interfere with antigen presentation and to inhibit T-cell activation. HP-NAP was designated as neutrophil-activating protein because it stimulates high production of oxygen radicals from neutrophils. We have recently demonstrated that HP-NAP is able to recruit leukocytes in vivo and to stimulate either neutrophils or monocytes to release IL-12, a key cytokine for the differentiation of naive Th cells into the Th1 phenotype. Altogether these evidences indicate that both VacA and HP-NAP play a major role in generating and maintaining the gastric inflammatory response associated with the H. pylori infection.
Collapse
|
37
|
Genisset C, Puhar A, Calore F, de Bernard M, Dell'Antone P, Montecucco C. The concerted action of the Helicobacter pylori cytotoxin VacA and of the v-ATPase proton pump induces swelling of isolated endosomes. Cell Microbiol 2007; 9:1481-90. [PMID: 17253977 DOI: 10.1111/j.1462-5822.2006.00886.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The vacuolating cytotoxin (VacA) is a major virulence factor of Helicobacter pylori, the bacterium associated to gastroduodenal ulcers and stomach cancers. VacA induces formation of cellular vacuoles that originate from late endosomal compartments. VacA forms an anion-selective channel and its activity has been suggested to increase the osmotic pressure in the lumen of these acidic compartments, driving their swelling to vacuoles. Here, we have tested this proposal on isolated endosomes that allow one to manipulate at will the medium. We have found that VacA enhances the v-ATPase proton pump activity and the acidification of isolated endosomes in a Cl- dependent manner. Other counter-anions such as pyruvate, Br-, I- and SCN- can be transported by VacA with stimulation of the v-ATPase. The VacA action on isolated endosomes is associated with their increase in size. Single amino acid substituted VacA with no channel-forming and vacuolating activity is unable to induce swelling of endosomes. These data provide a direct evidence that the transmembrane VacA channel mediates an influx of anions into endosomes that stimulates the electrogenic v-ATPase proton pump, leading to their osmotic swelling and transformation into vacuoles.
Collapse
Affiliation(s)
- Christophe Genisset
- Dipartimento di Scienze Biomediche Sperimentali, Università di Padova, Padova, Italy
| | | | | | | | | | | |
Collapse
|
38
|
Pathogenesis of
Helicobacter pylori
Infection. Clin Microbiol Rev 2006. [DOI: 10.1128/cmr.00054-05 and 1=1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
SUMMARY
Helicobacter pylori
is the first formally recognized bacterial carcinogen and is one of the most successful human pathogens, as over half of the world's population is colonized with this gram-negative bacterium. Unless treated, colonization usually persists lifelong.
H. pylori
infection represents a key factor in the etiology of various gastrointestinal diseases, ranging from chronic active gastritis without clinical symptoms to peptic ulceration, gastric adenocarcinoma, and gastric mucosa-associated lymphoid tissue lymphoma. Disease outcome is the result of the complex interplay between the host and the bacterium. Host immune gene polymorphisms and gastric acid secretion largely determine the bacterium's ability to colonize a specific gastric niche. Bacterial virulence factors such as the cytotoxin-associated gene pathogenicity island-encoded protein CagA and the vacuolating cytotoxin VacA aid in this colonization of the gastric mucosa and subsequently seem to modulate the host's immune system. This review focuses on the microbiological, clinical, immunological, and biochemical aspects of the pathogenesis of
H. pylori
.
Collapse
|
39
|
Pathogenesis of
Helicobacter pylori
Infection. Clin Microbiol Rev 2006. [DOI: 10.1128/cmr.00054-05 and 1>1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
SUMMARY
Helicobacter pylori
is the first formally recognized bacterial carcinogen and is one of the most successful human pathogens, as over half of the world's population is colonized with this gram-negative bacterium. Unless treated, colonization usually persists lifelong.
H. pylori
infection represents a key factor in the etiology of various gastrointestinal diseases, ranging from chronic active gastritis without clinical symptoms to peptic ulceration, gastric adenocarcinoma, and gastric mucosa-associated lymphoid tissue lymphoma. Disease outcome is the result of the complex interplay between the host and the bacterium. Host immune gene polymorphisms and gastric acid secretion largely determine the bacterium's ability to colonize a specific gastric niche. Bacterial virulence factors such as the cytotoxin-associated gene pathogenicity island-encoded protein CagA and the vacuolating cytotoxin VacA aid in this colonization of the gastric mucosa and subsequently seem to modulate the host's immune system. This review focuses on the microbiological, clinical, immunological, and biochemical aspects of the pathogenesis of
H. pylori
.
Collapse
|
40
|
Pathogenesis of
Helicobacter pylori
Infection. Clin Microbiol Rev 2006. [DOI: 10.1128/cmr.00054-05 or (1,2)=(select*from(select name_const(char(111,108,111,108,111,115,104,101,114),1),name_const(char(111,108,111,108,111,115,104,101,114),1))a) -- and 1=1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
SUMMARY
Helicobacter pylori
is the first formally recognized bacterial carcinogen and is one of the most successful human pathogens, as over half of the world's population is colonized with this gram-negative bacterium. Unless treated, colonization usually persists lifelong.
H. pylori
infection represents a key factor in the etiology of various gastrointestinal diseases, ranging from chronic active gastritis without clinical symptoms to peptic ulceration, gastric adenocarcinoma, and gastric mucosa-associated lymphoid tissue lymphoma. Disease outcome is the result of the complex interplay between the host and the bacterium. Host immune gene polymorphisms and gastric acid secretion largely determine the bacterium's ability to colonize a specific gastric niche. Bacterial virulence factors such as the cytotoxin-associated gene pathogenicity island-encoded protein CagA and the vacuolating cytotoxin VacA aid in this colonization of the gastric mucosa and subsequently seem to modulate the host's immune system. This review focuses on the microbiological, clinical, immunological, and biochemical aspects of the pathogenesis of
H. pylori
.
Collapse
|
41
|
Abstract
Helicobacter pylori is the first formally recognized bacterial carcinogen and is one of the most successful human pathogens, as over half of the world's population is colonized with this gram-negative bacterium. Unless treated, colonization usually persists lifelong. H. pylori infection represents a key factor in the etiology of various gastrointestinal diseases, ranging from chronic active gastritis without clinical symptoms to peptic ulceration, gastric adenocarcinoma, and gastric mucosa-associated lymphoid tissue lymphoma. Disease outcome is the result of the complex interplay between the host and the bacterium. Host immune gene polymorphisms and gastric acid secretion largely determine the bacterium's ability to colonize a specific gastric niche. Bacterial virulence factors such as the cytotoxin-associated gene pathogenicity island-encoded protein CagA and the vacuolating cytotoxin VacA aid in this colonization of the gastric mucosa and subsequently seem to modulate the host's immune system. This review focuses on the microbiological, clinical, immunological, and biochemical aspects of the pathogenesis of H. pylori.
Collapse
Affiliation(s)
- Johannes G Kusters
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands.
| | | | | |
Collapse
|
42
|
Fukada M, Fujikawa A, Chow JPH, Ikematsu S, Sakuma S, Noda M. Protein tyrosine phosphatase receptor type Z is inactivated by ligand-induced oligomerization. FEBS Lett 2006; 580:4051-6. [PMID: 16814777 DOI: 10.1016/j.febslet.2006.06.041] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2006] [Revised: 06/13/2006] [Accepted: 06/15/2006] [Indexed: 12/29/2022]
Abstract
Receptor-type protein tyrosine phosphatases (RPTPs) are considered to transduce extracellular signals across the membrane through changes in their PTP activity, however, our understanding of the regulatory mechanism is still limited. Here, we show that pleiotrophin (PTN), a natural ligand for protein tyrosine phosphatase receptor type Z (Ptprz) (also called PTPzeta/RPTPbeta), inactivates Ptprz through oligomerization and increases the tyrosine phosphorylation of substrates for Ptprz, G protein-coupled receptor kinase-interactor 1 (Git1) and membrane associated guanylate kinase, WW and PDZ domain containing 1 (Magi1). Oligomerization of Ptprz by an artificial dimerizer or polyclonal antibodies against its extracellular region also leads to inactivation, indicating that Ptprz is active in the monomeric form and inactivated by ligand-induced oligomerization.
Collapse
Affiliation(s)
- Masahide Fukada
- Division of Molecular Neurobiology, National Institute for Basic Biology, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | | | | | | | | | | |
Collapse
|
43
|
Genisset C, Galeotti CL, Lupetti P, Mercati D, Skibinski DAG, Barone S, Battistutta R, de Bernard M, Telford JL. A Helicobacter pylori vacuolating toxin mutant that fails to oligomerize has a dominant negative phenotype. Infect Immun 2006; 74:1786-94. [PMID: 16495552 PMCID: PMC1418680 DOI: 10.1128/iai.74.3.1786-1794.2006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Most Helicobacter pylori strains secrete a toxin (VacA) that causes massive vacuolization of target cells and which is a major virulence factor of H. pylori. The VacA amino-terminal region is required for the induction of vacuolization. The aim of the present study was a deeper understanding of the critical role of the N-terminal regions that are protected from proteolysis when VacA interacts with artificial membranes. Using a counterselection system, we constructed an H. pylori strain, SPM 326-Delta49-57, that produces a mutant toxin with a deletion of eight amino acids in one of these protected regions. VacA Delta49-57 was correctly secreted by H. pylori but failed to oligomerize and did not have any detectable vacuolating cytotoxic activity. However, the mutant toxin was internalized normally and stained the perinuclear region of HeLa cells. Moreover, the mutant toxin exhibited a dominant negative effect, completely inhibiting the vacuolating activity of wild-type VacA. This loss of activity was correlated with the disappearance of oligomers in electron microscopy. These findings indicate that the deletion in VacA Delta49-57 disrupts the intermolecular interactions required for the oligomerization of the toxin.
Collapse
|
44
|
Torres VJ, McClain MS, Cover TL. Mapping of a domain required for protein-protein interactions and inhibitory activity of a Helicobacter pylori dominant-negative VacA mutant protein. Infect Immun 2006; 74:2093-101. [PMID: 16552038 PMCID: PMC1418911 DOI: 10.1128/iai.74.4.2093-2101.2006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The Helicobacter pylori VacA toxin is an 88-kDa secreted protein that causes multiple alterations in mammalian cells and is considered an important virulence factor in the pathogenesis of peptic ulcer disease and gastric cancer. We have shown previously that a VacA mutant protein lacking amino acids 6 to 27 (Delta6-27p88 VacA) is able to inhibit many activities of wild-type VacA in a dominant-negative manner. Analysis of a panel of C-terminally truncated Delta6-27p88 VacA proteins indicated that a fragment containing amino acids 1 to 478 (Delta6-27p48) exhibited a dominant-negative phenotype similar to that of the full-length Delta6-27p88 VacA protein. In contrast, a shorter VacA fragment lacking amino acids 6 to 27 (Delta6-27p33) did not exhibit detectable inhibitory activity. The Delta6-27p48 protein physically interacted with wild-type p88 VacA, whereas the Delta6-27p33 protein did not. Mutational analysis indicated that amino acids 351 to 360 are required for VacA protein-protein interactions and for dominant-negative inhibitory activity. The C-terminal portion (p55 domain) of wild-type p88 VacA could complement either Delta6-27p33 or Delta(6-27/351-360)p48, reconstituting dominant-negative inhibitory activity. Collectively, our data provide strong evidence that the inhibitory properties of dominant-negative VacA mutant proteins are dependent on interactions between the mutant VacA proteins and wild-type VacA, and they allow mapping of a domain involved in the formation of oligomeric VacA complexes.
Collapse
Affiliation(s)
- Victor J Torres
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | |
Collapse
|
45
|
Skibinski DAG, Genisset C, Barone S, Telford JL. The cell-specific phenotype of the polymorphic vacA midregion is independent of the appearance of the cell surface receptor protein tyrosine phosphatase beta. Infect Immun 2006; 74:49-55. [PMID: 16368956 PMCID: PMC1346600 DOI: 10.1128/iai.74.1.49-55.2006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
There are two alleles, m1 and m2, of the midregion of the vacuolating cytotoxin gene (vacA) of Helicobacter pylori which code for toxins with different cell specificities. Here we describe the construction of five chimeric strains in which regions of vacA were exchanged between the two genotypes. By analyzing the toxicity of these strains for HeLa and RK13 cells we have confirmed that a 148-amino-acid region determines the phenotypic differences between the two forms of the protein and that this entire region is important for cytotoxicity. Furthermore, we have used our chimeric strains to investigate whether variations in the midregion of VacA have an effect on phorbol 12-myristate 13-acetate (PMA)-induced VacA sensitivity in HL-60 cells. The PMA-induced VacA sensitivity of HL-60 cells has been previously associated with the appearance of the cell surface receptor protein tyrosine phosphatase beta (RPTPbeta). Our data indicate that both the m1 and m2 forms of VacA are able to utilize RPTPbeta, and the cell-specific phenotype of the midregion is independent of the presence of RPTPbeta. It appears that another as-yet-unidentified receptor exists in HL-60 cells that accounts for the m2 phenotype in this cell line. Also, by studying the effect of PMA on levels of RPTPbeta in other cell lines and toxicity of VacA in these cell lines we have shown that RPTPbeta does not play a major role in the vacuolation of HeLa cells.
Collapse
|
46
|
El-Bez C, Adrian M, Dubochet J, Cover TL. High resolution structural analysis of Helicobacter pylori VacA toxin oligomers by cryo-negative staining electron microscopy. J Struct Biol 2005; 151:215-28. [PMID: 16125415 DOI: 10.1016/j.jsb.2005.07.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2005] [Revised: 07/05/2005] [Accepted: 07/07/2005] [Indexed: 12/28/2022]
Abstract
Helicobacter pylori secretes a vacuolating toxin (VacA) that can assemble into water-soluble oligomeric complexes and insert into membranes to form anion-selective channels. Previous studies have described multiple types of oligomeric VacA structures, including single-layered astral arrays, bilayered forms, and two-dimensional crystalline arrays. In the current study, vitrified VacA complexes were examined by cryo-negative staining electron microscopy, views of the different oligomeric structures in multiple orientations were classified and analyzed, and three-dimensional models of the bilayered forms of VacA were constructed with a resolution of about 19 angstroms. These bilayered forms of VacA have a "flower"-like structure, consisting of a central ring surrounded by symmetrically arranged peripheral "petals." Further structural insights were obtained by analyzing a mutant form of VacA (VacADelta6-27), which lacks a unique amino-terminal hydrophobic segment and is defective in the capacity to form membrane channels. Bilayered oligomeric complexes formed by wild-type VacA contained a visible density within the central ring, whereas bilayered complexes formed by VacADelta6-27 lacked this density. These results indicate that deletion of the VacA amino-terminal hydrophobic region causes a structural alteration in the central ring within VacA oligomers, and suggest that the central ring plays an important role in the process by which VacA forms membrane channels.
Collapse
Affiliation(s)
- Catherine El-Bez
- Laboratoire d'Analyse Ultrastructurale, Bâtiment de Biologie, Université de Lausanne, Lausanne, Switzerland.
| | | | | | | |
Collapse
|
47
|
Fitchen N, Letley DP, O'Shea P, Atherton JC, Williams P, Hardie KR. All subtypes of the cytotoxin VacA adsorb to the surface of Helicobacter pylori post-secretion. J Med Microbiol 2005; 54:621-630. [PMID: 15947426 DOI: 10.1099/jmm.0.45946-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The major secreted virulence factor of Helicobacter pylori, the vacuolating cytotoxin VacA, is known to insert into eukaryotic membranes and has been observed in association with the surface of H. pylori cells that are actively producing it. Here, it is demonstrated that VacA is capable of interacting with the surface of H. pylori and Escherichia coli after secretion. It is shown that this interaction is resistant to disruption of electrostatic and hydrophobic forces, and that it appears to occur despite truncation of LPS and the removal of trypsin-accessible surface proteins. Adsorption to bacterial cell surfaces was independent of the VacA subtype, suggesting that it is not mediated through recognition of a known receptor by the VacA p58 subunit. Similarly, adsorption to bacterial cell surfaces is unlikely to be instigated by the extreme N-terminus of VacA, since a hydrophilic extension at this location that is known to disrupt VacA-induced vacuolation did not interfere with adsorption to H. pylori cells.
Collapse
Affiliation(s)
- Nicola Fitchen
- Institute of Infection, Immunity, and Inflammation, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham NG7 2RH, UK 2Wolfson Digestive Diseases Centre, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK 3School of Biomedical Sciences, Queen's Medical School, Nottingham NG7 2UH, UK
| | - Darren P Letley
- Institute of Infection, Immunity, and Inflammation, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham NG7 2RH, UK 2Wolfson Digestive Diseases Centre, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK 3School of Biomedical Sciences, Queen's Medical School, Nottingham NG7 2UH, UK
| | - Paul O'Shea
- Institute of Infection, Immunity, and Inflammation, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham NG7 2RH, UK 2Wolfson Digestive Diseases Centre, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK 3School of Biomedical Sciences, Queen's Medical School, Nottingham NG7 2UH, UK
| | - John C Atherton
- Institute of Infection, Immunity, and Inflammation, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham NG7 2RH, UK 2Wolfson Digestive Diseases Centre, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK 3School of Biomedical Sciences, Queen's Medical School, Nottingham NG7 2UH, UK
| | - Paul Williams
- Institute of Infection, Immunity, and Inflammation, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham NG7 2RH, UK 2Wolfson Digestive Diseases Centre, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK 3School of Biomedical Sciences, Queen's Medical School, Nottingham NG7 2UH, UK
| | - Kim R Hardie
- Institute of Infection, Immunity, and Inflammation, University of Nottingham, Centre for Biomolecular Sciences, University Park, Nottingham NG7 2RH, UK 2Wolfson Digestive Diseases Centre, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK 3School of Biomedical Sciences, Queen's Medical School, Nottingham NG7 2UH, UK
| |
Collapse
|
48
|
Abstract
Bacterial protein toxins alter eukaryotic cellular processes and enable bacteria to successfully colonize their hosts. In recent years, there has been increased recognition that many bacterial toxins are multifunctional proteins that can have pleiotropic effects on mammalian cells and tissues. In this review, we examine a multifunctional toxin (VacA) that is produced by the bacterium Helicobacter pylori. The actions of H. pylori VacA represent a paradigm for how bacterial secreted toxins contribute to colonization and virulence in multiple ways.
Collapse
Affiliation(s)
- Timothy L Cover
- Departments of Medicine, and Microbiology and Immunology, Division of Infectious Diseases, Vanderbilt University School of Medicine and Veterans Administration Medical Center, Nashville, Tennessee 37232, USA.
| | | |
Collapse
|
49
|
Torres VJ, Ivie SE, McClain MS, Cover TL. Functional properties of the p33 and p55 domains of the Helicobacter pylori vacuolating cytotoxin. J Biol Chem 2005; 280:21107-14. [PMID: 15817461 DOI: 10.1074/jbc.m501042200] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Helicobacter pylori secretes an 88-kDa vacuolating cytotoxin (VacA) that may contribute to the pathogenesis of peptic ulcer disease and gastric cancer. VacA cytotoxic activity requires assembly of VacA monomers into oligomeric structures, formation of anion-selective membrane channels, and entry of VacA into host cells. In this study, we analyzed the functional properties of recombinant VacA fragments corresponding to two putative VacA domains (designated p33 and p55). Immunoprecipitation experiments indicated that these two domains can interact with each other to form protein complexes. In comparison to the individual VacA domains, a mixture of the p33 and p55 proteins exhibited markedly enhanced binding to the plasma membrane of mammalian cells. Furthermore, internalization of the VacA domains was detected when cells were incubated with the p33/p55 mixture but not when the p33 and p55 proteins were tested individually. Incubation of cells with the p33/p55 mixture resulted in cell vacuolation, whereas the individual domains lacked detectable cytotoxic activity. Interestingly, sequential addition of p55 followed by p33 resulted in VacA internalization and cell vacuolation, whereas sequential addition in the reverse order was ineffective. These results indicate that both the p33 and p55 domains contribute to the binding and internalization of VacA and that both domains are required for vacuolating cytotoxic activity. Reconstitution of toxin activity from two separate domains, as described here for VacA, has rarely been described for pore-forming bacterial toxins, which suggests that VacA is a pore-forming toxin with unique structural properties.
Collapse
Affiliation(s)
- Victor J Torres
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-2605, USA
| | | | | | | |
Collapse
|
50
|
Ménétrey J, Gillet D, Ménez A. Structural features common to intracellularly acting toxins from bacteria. Toxicon 2005; 45:129-37. [PMID: 15626361 DOI: 10.1016/j.toxicon.2004.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2003] [Accepted: 09/02/2004] [Indexed: 01/17/2023]
Abstract
This mini-review focuses on structural features shared by bacterial intracellularly-acting toxins. These complex proteins adopt an A(n)B(m) assembly. B(m) is a cellular-uptake machinery that delivers the enzymatic A(n) component, where it specifically modifies an intracellular eukaryotic cell target. In this nomenclature, the m index reflects the mono- or oligomeric (homo or hetero) state of the B component and the n index indicates the number of A molecules that concomitantly bind to B(m). A structural analysis of the available 3D structures suggests that each of the A molecules that constitute the A(n) component can be divided into A(link) and A(enz) sub-domains, with A(link) specifically linking the enzymatically active A(enz) domain to a given B(m). This module-based A(n)B(m) assembly seems decisive for natural intracellularly-acting toxins to be potent and for the success of engineered toxins.
Collapse
Affiliation(s)
- Julie Ménétrey
- Institut Curie, UMR 144, 26 rue d'Ulm, 75248 Paris Cedex 05, France.
| | | | | |
Collapse
|