101
|
Abstract
The pathogenicity of many bacteria colonizing the gastrointestinal tract often depends on their ability to gain access to cells that are normally non-phagocytic. Helicobacter pylori colonizes the stomach of over half the world population and is the main cause of peptic ulcer disease and gastric cancer. It is generally considered to be a non-invasive pathogen present only in the lumen of the stomach and attached to gastric epithelial cells although a number of in vivo and in vitro studies have demonstrated that H. pylori is in fact invasive. In addition, H. pylori can repopulate the extracellular environment after complete elimination of extracellular bacteria with gentamicin, suggesting it may be considered a facultative intracellular bacterium. This review examines the validity of these observations and describes the evidence suggesting that the intracellular presence of H. pylori plays a role in the induction of diseases, in immune evasion, and in life-long persistence of the bacterium in the stomach of a majority of humans.
Collapse
Affiliation(s)
- Andre Dubois
- Laboratory of Gastrointestinal and Liver Studies, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4799, USA.
| | | |
Collapse
|
102
|
Abstract
Helicobacter pylori is a spiral-shaped, flagellated, microaerophilic Gram-negative bacterium that colonizes the gastric epithelium of humans. All persons infected with H. pylori have gastritis, and some will develop severe disease such as peptic ulcers or gastric cancer. A characteristic feature of this infection is the pronounced accumulation of phagocytes, particularly neutrophils, in the gastric mucosa. H. pylori thrives in a phagocyte-rich environment, and we describe here how this organism uses an array of novel virulence factors to manipulate chemotaxis, phagocytosis, membrane trafficking and the respiratory burst as a means to evade elimination by the innate immune response.
Collapse
Affiliation(s)
- Lee-Ann H Allen
- Inflammation Program and the Department of Medicine, University of Iowa and the VA Medical Center, Iowa City, IA 52242, USA.
| |
Collapse
|
103
|
Abstract
Neutrophils are short-lived granulocytes essential for innate host defense. We describe here methods for analysis of resting and activated cells using immunofluorescence and confocal microscopy. Procedures for stimulation of adherent and suspended cells are provided along with protocols for particle opsonization and synchronized phagocytosis. Most importantly, we describe in detail methods for rapid and efficient cell fixation and permeabilization that optimize detection of granule proteins and NADPH oxidase components. Variables that impact antigen detection (such as cell spreading, degranulation, and phagocytosis) are discussed as are methods for image acquisition and analysis.
Collapse
|
104
|
Robinson K, Argent RH, Atherton JC. The inflammatory and immune response to Helicobacter pylori infection. Best Pract Res Clin Gastroenterol 2007; 21:237-59. [PMID: 17382275 DOI: 10.1016/j.bpg.2007.01.001] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Lifelong Helicobacter pylori infection and its associated gastric inflammation underlie peptic ulceration and gastric carcinogenesis. The immune and inflammatory responses to H. pylori are doubly responsible: gastric inflammation is the main mediator of pathology, and the immune and inflammatory response is ineffective, allowing lifelong bacterial persistence. However, despite inducing gastric inflammation, most infections do not cause disease, and bacterial, host and environmental factors determine individual disease risk. Although H. pylori avoids many innate immune receptors, specific virulence factors (including those encoded on the cag pathogenicity island) stimulate innate immunity to increase gastric inflammation and increase disease risk. An acquired T helper 1 response upregulates local immune effectors. The extent to which environmental factors (including parasite infection), host factors and H. pylori itself influence T-helper differentiation and regulatory T-cell responses remains controversial. Finally, effective vaccines have still not been developed: a better understanding of the immune response to H. pylori may help.
Collapse
Affiliation(s)
- Karen Robinson
- Wolfson Digestive Diseases Centre, University of Nottingham, C Floor, South Block, Queen's Medical Centre Campus, Nottingham University Hospital NHS Trust, Nottingham NG7 2UH, UK.
| | | | | |
Collapse
|
105
|
Algood HMS, Cover TL. Helicobacter pylori persistence: an overview of interactions between H. pylori and host immune defenses. Clin Microbiol Rev 2006; 19:597-613. [PMID: 17041136 PMCID: PMC1592695 DOI: 10.1128/cmr.00006-06] [Citation(s) in RCA: 183] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori is a gram-negative bacterium that persistently colonizes more than half of the global human population. In order to successfully colonize the human stomach, H. pylori must initially overcome multiple innate host defenses. Remarkably, H. pylori can persistently colonize the stomach for decades or an entire lifetime despite development of an acquired immune response. This review focuses on the immune response to H. pylori and the mechanisms by which H. pylori resists immune clearance. Three main sections of the review are devoted to (i) analysis of the immune response to H. pylori in humans, (ii) analysis of interactions of H. pylori with host immune defenses in animal models, and (iii) interactions of H. pylori with immune cells in vitro. The topics addressed in this review are important for understanding how H. pylori resists immune clearance and also are relevant for understanding the pathogenesis of diseases caused by H. pylori (peptic ulcer disease, gastric adenocarcinoma, and gastric lymphoma).
Collapse
Affiliation(s)
- Holly M Scott Algood
- Division of Infectious Diseases, A2200 Medical Center North, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | |
Collapse
|
106
|
Kyei GB, Vergne I, Chua J, Roberts E, Harris J, Junutula JR, Deretic V. Rab14 is critical for maintenance of Mycobacterium tuberculosis phagosome maturation arrest. EMBO J 2006; 25:5250-9. [PMID: 17082769 PMCID: PMC1636625 DOI: 10.1038/sj.emboj.7601407] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Accepted: 10/05/2006] [Indexed: 01/12/2023] Open
Abstract
Mycobacterium tuberculosis arrests phagosomal maturation in infected macrophage, and, apart from health significance, provides a superb model system to dissect the phagolysosomal biogenesis pathway. Here, we demonstrate a critical role for the small GTPase Rab14 in maintaining mycobacterial phagosome maturation block. Four-dimensional microscopy showed that phagosomes containing live mycobacteria accumulated Rab14 following phagocytosis. The recruitment of Rab14 had strong functional consequence, as a knockdown of endogenous Rab14 by siRNA or overexpression of Rab14 dominant-negative mutants (Rab14S25N and Rab14N125I) released the maturation block and allowed phagosomes harboring live mycobacteria to progress into phagolysosomes. Conversely, overexpression of the wild-type Rab14 and the constitutively active mutant Rab14Q70L prevented phagosomes with dead mycobacteria from undergoing default maturation into phagolysosomal organelles. Mechanistic studies demonstrated a role for Rab14 in stimulating organellar fusion between phagosomes and early endosomes but not with late endosomes. Rab14 enables mycobacterial phagosomes to maintain early endosomal characteristics and avoid late endosomal/lysosomal degradative components.
Collapse
Affiliation(s)
- George B Kyei
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Isabelle Vergne
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Jennifer Chua
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Esteban Roberts
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - James Harris
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | | | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
- Departments of Molecular Genetics & Microbiology, and Cell Biology and Physiology, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, NM 87131-001, USA. Tel.: +1 505 272 0291; Fax: +1 505 272 5309; E-mail:
| |
Collapse
|
107
|
Wunder C, Churin Y, Winau F, Warnecke D, Vieth M, Lindner B, Zähringer U, Mollenkopf HJ, Heinz E, Meyer TF. Cholesterol glucosylation promotes immune evasion by Helicobacter pylori. Nat Med 2006; 12:1030-8. [PMID: 16951684 DOI: 10.1038/nm1480] [Citation(s) in RCA: 215] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Accepted: 08/01/2006] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori infection causes gastric pathology such as ulcer and carcinoma. Because H. pylori is auxotrophic for cholesterol, we have explored the assimilation of cholesterol by H. pylori in infection. Here we show that H. pylori follows a cholesterol gradient and extracts the lipid from plasma membranes of epithelial cells for subsequent glucosylation. Excessive cholesterol promotes phagocytosis of H. pylori by antigen-presenting cells, such as macrophages and dendritic cells, and enhances antigen-specific T cell responses. A cholesterol-rich diet during bacterial challenge leads to T cell-dependent reduction of the H. pylori burden in the stomach. Intrinsic alpha-glucosylation of cholesterol abrogates phagocytosis of H. pylori and subsequent T cell activation. We identify the gene hp0421 as encoding the enzyme cholesterol-alpha-glucosyltransferase responsible for cholesterol glucosylation. Generation of knockout mutants lacking hp0421 corroborates the importance of cholesteryl glucosides for escaping phagocytosis, T cell activation and bacterial clearance in vivo. Thus, we propose a mechanism regulating the host-pathogen interaction whereby glucosylation of a lipid tips the scales towards immune evasion or response.
Collapse
Affiliation(s)
- Christian Wunder
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
108
|
McCaffrey RL, Allen LAH. Francisella tularensis LVS evades killing by human neutrophils via inhibition of the respiratory burst and phagosome escape. J Leukoc Biol 2006; 80:1224-30. [PMID: 16908516 PMCID: PMC1828114 DOI: 10.1189/jlb.0406287] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Francisella tularensis is a Gram-negative bacterium and the causative agent of tularemia. Recent data indicate that F. tularensis replicates inside macrophages, but its fate in other cell types, including human neutrophils, is unclear. We now show that F. tularensis live vaccine strain (LVS), opsonized with normal human serum, was rapidly ingested by neutrophils but was not eliminated. Moreover, evasion of intracellular killing can be explained, in part, by disruption of the respiratory burst. As judged by luminol-enhanced chemiluminescence and nitroblue tetrazolium staining, neutrophils infected with live F. tularensis did not generate reactive oxygen species. Confocal microscopy demonstrated that NADPH oxidase assembly was disrupted, and LVS phagosomes did not acquire gp91/p22(phox) or p47/p67(phox). At the same time, F. tularensis also impaired neutrophil activation by heterologous stimuli such as phorbol esters and opsonized zymosan particles. Later in infection, LVS escaped the phagosome, and live organisms persisted in the neutrophil cytosol for at least 12 h. To our knowledge, our data are the first demonstration of a facultative intracellular pathogen, which disrupts the oxidative burst and escapes the phagosome to evade elimination inside neutrophils, and as such, our data define a novel mechanism of virulence.
Collapse
Affiliation(s)
- Ramona L. McCaffrey
- Inflammation Program, University of Iowa and the VA Medical Center, Iowa City, Iowa, USA
- Departments of Medicine, University of Iowa and the VA Medical Center, Iowa City, Iowa, USA
| | - Lee-Ann H. Allen
- Inflammation Program, University of Iowa and the VA Medical Center, Iowa City, Iowa, USA
- Departments of Medicine, University of Iowa and the VA Medical Center, Iowa City, Iowa, USA
- Microbiology, University of Iowa and the VA Medical Center, Iowa City, Iowa, USA
- Correspondence: Inflammation Program, University of Iowa, 2501 Crosspark Rd., MTF-D154, Coralville, IA 52241, USA. E-mail:
| |
Collapse
|
109
|
Schulert GS, Allen LAH. Differential infection of mononuclear phagocytes by Francisella tularensis: role of the macrophage mannose receptor. J Leukoc Biol 2006; 80:563-71. [PMID: 16816147 PMCID: PMC1865506 DOI: 10.1189/jlb.0306219] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Francisella tularensis (Ft) is a Gram-negative bacterium and the causative agent of tularemia. It is well established that this organism replicates inside macrophages, but we are only beginning to understand this interface at the molecular level. Herein, we compared directly the ability of Ft subspecies holarctica live-vaccine strain to infect freshly isolated human peripheral blood monocytes, monocyte-derived macrophages (MDM), and cells of the murine macrophage cell line J774A.1 (J774). We now show that unopsonized bacteria infected human MDM fivefold more efficiently than monocytes or J774 cells in standard media. Moreover, enhanced infection of MDM was mediated, in part, by the macrophage mannose receptor (MR). Forming Ft phagosomes accumulated MR, and infection was inhibited by MR-blocking antibody or soluble mannan but not by the dectin-1 ligand laminarin. Up-regulation of MR in MDM (by exposure to interleukin-4) increased Ft phagocytosis, as did expression of MR in J774 cells. Conversely, opsonized Ft were ingested readily by monocytes and MDM. Medium supplementation with 2.5% fresh autologous serum was sufficient to confer opsonophagocytosis and CD11b accumulated in the membrane at sites of Ft engulfment. Infection of monocytes by opsonized Ft was nearly ablated by complement receptor 3 (CR3) blockade. Conversely, MDM used MR and CD11b/CD18 to ingest opsonized organisms. Altogether, our data demonstrate differential infection of mononuclear phagocytes by Ft and define distinct roles for MR and CR3 in phagocytosis.
Collapse
Affiliation(s)
- Grant S. Schulert
- Inflammation Program, University of Iowa and the VA Medical Center, Iowa City
- Department of Microbiology, University of Iowa and the VA Medical Center, Iowa City
| | - Lee-Ann H. Allen
- Inflammation Program, University of Iowa and the VA Medical Center, Iowa City
- Department of Microbiology, University of Iowa and the VA Medical Center, Iowa City
- Department of Internal Medicine, University of Iowa and the VA Medical Center, Iowa City
- Correspondence: Inflammation Program, University of Iowa, 2501 Crosspark Rd., MTF D154, Coralville, IA 52241. E-mail:
| |
Collapse
|
110
|
Schmees C, Gerhard M, Treptau T, Voland P, Schwendy S, Rad R, Prinz C. VacA-associated inhibition of T-cell function: reviewed and reconsidered. Helicobacter 2006; 11:144-6. [PMID: 16684260 DOI: 10.1111/j.1523-5378.2006.00393.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Chronic Helicobacter pylori infection is characterized by dense infiltration of the mucosa with neutrophilic granulocytes, lymphocytes, and monocytes/macrophages. Among these different cell types, T-lymphocytes are the most intriguing and crucial cells for the elimination of the bacteria. Previous studies have elucidated possible mechanisms on how bacteria could interfere with the human immune response and claimed that especially the secreted vacuolating toxin VacA may be responsible for the chronic persistence of the bacteria. Some of these results have to be interpreted with caution and may just describe in vitro phenomena; others may reveal promising facts.
Collapse
Affiliation(s)
- Christian Schmees
- II. Medical Department, Technical University of Munich, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
111
|
Schwartz JT, Allen LAH. Role of urease in megasome formation and Helicobacter pylori survival in macrophages. J Leukoc Biol 2006; 79:1214-25. [PMID: 16543403 PMCID: PMC1868427 DOI: 10.1189/jlb.0106030] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Previous studies have demonstrated that Helicobacter pylori (Hp) delays its entry into macrophages and persists inside megasomes, which are poorly acidified and accumulate early endosome autoantigen 1. Herein, we explored the role of Hp urease in bacterial survival in murine peritoneal macrophages and J774 cells. Plasmid-free mutagenesis was used to replace ureA and ureB with chloramphenicol acetyltransferase in Hp Strains 11637 and 11916. ureAB null Hp lacked detectable urease activity and did not express UreA or UreB as judged by immunoblotting. Deletion of ureAB had no effect on Hp binding to macrophages or the rate or extent of phagocytosis. However, intracellular survival of mutant organisms was impaired significantly. Immunofluorescence microscopy demonstrated that (in contrast to parental organisms) mutant Hp resided in single phagosomes, which were acidic and accumulated the lysosome marker lysosome-associated membrane protein-1 but not early endosome autoantigen 1. A similar phenotype was observed for spontaneous urease mutants derived from Hp Strain 60190. Treatment of macrophages with bafilomycin A1, NH4Cl, or chloroquine prevented acidification of phagosomes containing mutant Hp. However, only ammonium chloride enhanced bacterial viability significantly. Rescue of ureAB null organisms was also achieved by surface adsorption of active urease. Altogether, our data indicate a role for urease and urease-derived ammonia in megasome formation and Hp survival.
Collapse
Affiliation(s)
- Justin T. Schwartz
- Department of Medicine, University of Iowa and the VA Medical Center, Iowa City
- Department of Microbiology, University of Iowa and the VA Medical Center, Iowa City
| | - Lee-Ann H. Allen
- Department of Medicine, University of Iowa and the VA Medical Center, Iowa City
- Department of Microbiology, University of Iowa and the VA Medical Center, Iowa City
- Inflammation Program, University of Iowa and the VA Medical Center, Iowa City
| |
Collapse
|
112
|
Portal-Celhay C, Perez-Perez GI. Immune responses to Helicobacter pylori colonization: mechanisms and clinical outcomes. Clin Sci (Lond) 2006; 110:305-14. [PMID: 16464172 DOI: 10.1042/cs20050232] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori colonizes the stomachs of half of the world's population and usually persists in the gastric mucosa of human hosts for decades or life. Although most H. pylori-positive people are asymptomatic, the presence of H. pylori is associated with increased risk for the development of peptic ulcer disease, gastric adenocarcinoma and gastric lymphoma. The development of a sustained gastric inflammatory and immune response to infection appears to be pivotal for the development of disease. During its long co-existence with humans, H. pylori has evolved complex strategies to maintain a mild inflammation of the gastric epithelium while limiting the extent of immune effector activity. In this review, the nature of the host immune response to H. pylori infection and the mechanism employed by the bacterium to evade them is considered. Understanding the mechanisms of colonization, persistence and virulence factors of the bacterium as well as the innate and adaptive immune responses of the host are critically important for the development of new strategies to prevent the development of H. pylori-induced gastroduodenal disease.
Collapse
Affiliation(s)
- Cynthia Portal-Celhay
- Department of Microbiology, NYU School of Medicine, VA Medical Center, 423 East 23rd Street, New York, NY 10010, USA
| | | |
Collapse
|
113
|
Bergman M, Del Prete G, van Kooyk Y, Appelmelk B. Helicobacter pylori phase variation, immune modulation and gastric autoimmunity. Nat Rev Microbiol 2006; 4:151-9. [PMID: 16415930 DOI: 10.1038/nrmicro1344] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Helicobacter pylori can be regarded as a model pathogen for studying persistent colonization of humans. Phase-variable expression of Lewis blood-group antigens by H. pylori allows this microorganism to modulate the host T-helper-1-cell versus T-helper-2-cell response. We describe a model in which interactions between host lectins and pathogen carbohydrates facilitate asymptomatic persistence of H. pylori. This delicate balance, favourable for both the pathogen and the host, could lead to gastric autoimmunity in genetically susceptible individuals.
Collapse
Affiliation(s)
- Mathijs Bergman
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Room L-253, 3015 GD Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|
114
|
Srinivasan V, Morowitz HJ. Ancient genes in contemporary persistent microbial pathogens. THE BIOLOGICAL BULLETIN 2006; 210:1-9. [PMID: 16501059 DOI: 10.2307/4134531] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Autotrophs, the earliest prokaryotes, use CO(2) as the sole or the key source in the reductive citric acid cycle for carbon fixation. This pathway, also known as the reductive tricarboxylic acid (rTCA) cycle, has as its center the Krebs cycle running in the reductive direction, using reduced cofactors for energy. During the infection process, persistent pathogenic bacteria like Mycobacterium tuberculosis, Helicobacter pylori, and Salmonella typhi experience diverse and hostile environments both intracellularly (in macrophages) and extracellularly. M. tuberculosis, for example, must adapt to nutrient-deprived, hypoxic conditions in the granuloma. Genomic annotations reveal the presence of the key enzymes of the rTCA cycle--citrate lyase (Enzyme Commission number EC 4.1.3.6) and 2-oxoglutarate synthase (EC 1.2.7.3)--along with the rest of the TCA cycle enzymes. It is possible that there is a metabolic switch to anaerobic respiration in which a complete or a partial TCA cycle may operate in the reductive mode. This switch would both facilitate carbon fixation and restore the balance of oxidative and reductive reactions during environmental transitions, thus enabling the pathogen to survive, grow, and persist. Verification of enzyme function by biochemical investigations and validation of gene essentiality by knockout studies may reveal these enzymes to be rational drug targets for treatment of persistent microbial infections in mechanism-based drug discovery processes.
Collapse
|
115
|
Ernst PB, Peura DA, Crowe SE. The translation of Helicobacter pylori basic research to patient care. Gastroenterology 2006; 130:188-206; quiz 212-3. [PMID: 16401482 DOI: 10.1053/j.gastro.2005.06.032] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Accepted: 06/09/2005] [Indexed: 12/14/2022]
Abstract
In 1984, Barry Marshall and Robin Warren proposed a role for bacterial infections in the pathogenesis of gastroduodenal disease, which triggered an avalanche of research intended to prove or disprove their theory. The result has been a series of advances that have enhanced our understanding of these diseases and completely modernized the clinical approach to their management. In just over 20 years, many aspects of the immunopathogenesis of these diseases have been dissected at the molecular level, with key pathogenic mechanisms being validated by the identification of genes that are associated with the development of gastric cancer. There has been particular emphasis on understanding the molecular structures associated with Helicobacter pylori and their role in modifying the host responses. Gastric immune and inflammatory responses have emerged as key elements in the pathogenesis of gastritis and epithelial cell damage. This review summarizes important findings emanating from basic research primarily related to the immunopathogenesis of H pylori that have advanced the practice of medicine or our understanding of gastroduodenal disease.
Collapse
Affiliation(s)
- Peter B Ernst
- Digestive Health Center of Excellence, Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia 22908-0708, USA.
| | | | | |
Collapse
|
116
|
Cooke CL, Huff JL, Solnick JV. The role of genome diversity and immune evasion in persistent infection with Helicobacter pylori. ACTA ACUST UNITED AC 2005; 45:11-23. [PMID: 15949928 DOI: 10.1016/j.femsim.2005.04.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2005] [Revised: 04/14/2005] [Accepted: 04/15/2005] [Indexed: 02/08/2023]
Abstract
Helicobacter pylori is an important human pathogen that chronically colonizes the stomach of half the world's population. Infection typically occurs in childhood and persists for decades, if not for the lifetime of the host. How is bacterial persistence possible despite a vigorous innate and adaptive immune response? Here we describe the complex role of bacterial diversity and specific mechanisms to avoid or subvert host immunity in bacterial persistence. We suggest that H. pylori finely modulates the extent to which it interacts with the host in order to promote chronic infection, and that it uses diverse mechanisms to do so.
Collapse
Affiliation(s)
- Cara L Cooke
- Departments of Internal Medicine and Medical Microbiology and Immunology, Center for Comparative Medicine, University of California, Davis, CA 95616, USA
| | | | | |
Collapse
|
117
|
Duménil G, Nassif X. Extracellular bacterial pathogens and small GTPases of the Rho family: an unexpected combination. Curr Top Microbiol Immunol 2005; 291:11-28. [PMID: 15981457 DOI: 10.1007/3-540-27511-8_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Even in the case of extracellular bacterial pathogens, it is becoming increasingly clear that successful colonization does not limit itself to passive attachment on the surface of human cells; a dialogue takes place between bacteria and infected cells. These pathogens modulate cellular functions to their advantage, leading to survival and proliferation at the cell surface. Furthermore, there is increasing evidence that a variety of extracellular pathogens activate small GTPases of the Rho family during adhesion, placing these regulators at the center of the interaction between these bacteria and their infected host.
Collapse
Affiliation(s)
- G Duménil
- INSERM Unité 570, Faculté de Médecine Necker-Enfants Malades, 156 rue de Vaugirard, 75015 Paris, France.
| | | |
Collapse
|
118
|
Elshal MF, Elsayed IH, El Kady IM, Badra G, El-Refaei A, El-Batanony M, Hendy OM. Role of concurrent S. mansoni infection in H. pylori-associated gastritis: a flow cytometric DNA-analysis and oxyradicals correlations. Clin Chim Acta 2005; 346:191-8. [PMID: 15256320 DOI: 10.1016/j.cccn.2004.03.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Revised: 02/21/2004] [Accepted: 03/09/2004] [Indexed: 01/03/2023]
Abstract
BACKGROUND/AIM Helicobacter pylori infection is associated with the development of atrophic gastritis and increased gastric epithelial proliferation that is important in developing gastric carcinoma. Some countries with a high prevalence of H. pylori infection have high gastric cancer rates, whereas in others these rates are low. Several theories have been advanced to explain this phenomenon. One of these explanations is that the concurrent parasitic infection that is common in the African population might alter the immune response to H. pylori infection and reduce the incidence of atrophic gastritis. The aim of the present study was to assess whether concurrent Schistosoma mansoni infection with H. pylori has an effect on gastric mucosal injury in view of cell proliferation, apoptosis, pathological changes, nitric oxide (NO), oxyradicals and antioxidant capacity status. PATIENTS/METHODS Between April 2001 and March 2002, 73 patients were subjected to upper gastrointestinal endoscopy for dyspepsia and liver cirrhosis in the National Liver Institute, Menoufiya University. Biopsies were obtained from any lesion as well as from apparently healthy mucosa. Specimens were preserved in RNA later solution, and then kept at -80 degrees C until utilized for estimation of DNA-flow cytometric assay, reduced glutathione (GSH), catalase (CAT), superoxide dismutase (SOD), NO and lipid peroxidation (LPO) product--malondialdehyde (MDA). Diagnosis of bilharziasis was done by stool analysis, or by sigmoidoscopy and rectal snip. RESULTS Of the 73 patients, 48 were H. pylori-positive, 34 of them were positive and 14 were negative for S. mansoni. Of the 25 H. pylori-negative cases, 18 were positive and 7 were negative for S. mansoni. Concurrent infection with S. mansoni occurred in 34 patients and they had reduced DNA S-phase (7.57 +/- 4.99 vs. 14.5 +/- 3.11, P = 0.001), reduced proliferation activity (9.95 +/- 3.95 vs. 16.78, P < 0.004) and reduced apoptosis (21.83 +/- 11.64 vs. 26.0 +/- 8.31, P > 0.05) compared with H. pylori infected patients alone. CONCLUSIONS The results demonstrate that concurrent helminthes infection may modify the inflammatory response to gastric H. pylori infection manifested by the reduction of oxyradical-induced DNA-damage, apoptosis and cellular proliferation activity, and the increase in antioxidant production. Concurrent S. mansoni infection may have a protective effect against the possible progression of H. pylori-induced gastritis towards gastric carcinoma.
Collapse
Affiliation(s)
- M F Elshal
- Genetic Engineering and Biotechnology Institute, Menoufiya University, Egypt.
| | | | | | | | | | | | | |
Collapse
|
119
|
Chui SY, Clay TM, Lyerly HK, Morse MA. The Development of Therapeutic and Preventive Vaccines for Gastric Cancer and Helicobacter pylori. Cancer Epidemiol Biomarkers Prev 2005; 14:1883-9. [PMID: 16103431 DOI: 10.1158/1055-9965.epi-04-0775] [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: 11/16/2022] Open
Abstract
Gastric cancer is one of the most important worldwide public health problems. Convincing epidemiologic and etiologic associations have been made between the development of gastric cancer and infection with Helicobacter pylori. H. pylori not only has adapted to survive within the harsh environment of the stomach but also is able to modulate and avoid endogenous immune responses. The design and creation of efficacious vaccine strategies against H. pylori requires an understanding of the complex interactions that make up mucosal immunity. An effective vaccine strategy against H. pylori has the potential to affect significantly on population health worldwide.
Collapse
Affiliation(s)
- Stephen Y Chui
- Duke University Medical Center, Box 2606, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
120
|
Kranzer K, Söllner L, Aigner M, Lehn N, Deml L, Rehli M, Schneider-Brachert W. Impact of Helicobacter pylori virulence factors and compounds on activation and maturation of human dendritic cells. Infect Immun 2005; 73:4180-9. [PMID: 15972508 PMCID: PMC1168582 DOI: 10.1128/iai.73.7.4180-4189.2005] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Recently, we and others have shown that Helicobacter pylori induces dendritic cell (DC) activation and maturation. However, the impact of virulence factors on the interplay between DCs and H. pylori remains elusive. Therefore, we investigated the contribution of cag pathogenicity island (PAI) and VacA status on cytokine release and up-regulation of costimulatory molecules in H. pylori-treated DCs. In addition, to characterize the stimulatory capacity of H. pylori compounds in more detail, we studied the effect of formalin-inactivated and sonicated H. pylori, as well as secreted H. pylori molecules, on DCs. Incubation of DCs with viable or formalin-inactivated H. pylori induced comparable secretion of interleukin-6 (IL-6), IL-8, IL-10, IL-12, IL-1beta, and tumor necrosis factor (TNF). In contrast, IL-12 and IL-1beta release was significantly reduced in DCs treated with sonicated bacteria and secreted bacterial molecules. Treatment of sonicated H. pylori preparations with polymyxin B resulted in a significant reduction of IL-8 and IL-6 secretion, suggesting that H. pylori-derived lipopolysaccharide at least partially contributes to activation of immature DCs. In addition, the capacity of H. pylori-pulsed DCs to activate allogeneic T cells was not affected by cag PAI and VacA. Pretreatment of DC with cytochalasin D significantly inhibited secretion of IL-12, IL-1beta, and TNF, indicating that phagocytosis of H. pylori contributes to maximal activation of DCs. Taken together, our results suggest that DC activation and maturation, as well as DC-mediated T-cell activation, are independent of the cag PAI and VacA status of H. pylori.
Collapse
Affiliation(s)
- Katharina Kranzer
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
121
|
Allen LAH, Allgood JA, Han X, Wittine LM. Phosphoinositide3-kinase regulates actin polymerization during delayed phagocytosis of Helicobacter pylori. J Leukoc Biol 2005; 78:220-30. [PMID: 15809290 PMCID: PMC1868428 DOI: 10.1189/jlb.0205091] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
We have shown previously that ulcerogenic (type I) strains of Helicobacter pylori (Hp) retard their entry into macrophages. However, the signaling pathways that regulate Hp phagocytosis are largely undefined. We show here that Hp strongly activated class IA phosphoinositide3-kinases (PI3Ks) in macrophages, coincident with phagocytosis, and endogenous p85 and active protein kinase Balpha accumulated on forming phagosomes. PI3K inhibitors, wortmannin and LY294002, inhibited phagocytosis of Hp in a dose-dependent manner, and blockade of engulfment correlated directly with loss of 3'-phosphoinositides in the membrane subjacent to attached bacteria. During uptake of large immunoglobulin G (IgG)-coated particles, PI3Ks regulate pseudopod extension and phagosome closure. In marked contrast, we show here that 3'-phosphoinositides regulated actin polymerization at sites of Hp uptake. Moreover, Hp and IgG beads activated distinct PI3K isoforms. Phagosomes containing IgG-coated particles accumulated 3'-phosphatase and tensin homologue deleted on chromosome 10 and Src homology 2 domain-containing inositol 5'-phosphatase, yet Hp phagosomes did not. Finally, rapid uptake of IgG-opsonized Hp or a less-virulent type II Hp was PI3K-independent. We conclude that Hp and IgG beads are ingested by distinct mechanisms and that PI3Ks regulate the actin cytoskeleton during slow phagocytosis of ulcerogenic Hp.
Collapse
Affiliation(s)
- Lee-Ann H Allen
- Department of Medicine, University of Iowa, Coralville, 52241, USA.
| | | | | | | |
Collapse
|
122
|
Abstract
To maintain prolonged colonization of the human gastric mucosa, Helicobacter pylori must avoid both innate and adaptive immune responses. During its long coexistence with humans, it has evolved complex strategies to maintain a mild inflammation of the gastric epithelium while limiting the extent of immune effector activity. Severe disease, associated with bacterial colonization, might reflect loss of this control. Several mechanisms and the bacterial factors involved in immune subversion have, in recent years, been elucidated, thus opening the possibility of a better understanding of the pathogenicity of this microorganism.
Collapse
Affiliation(s)
- Cosima T Baldari
- Department of Evolutionary Biology, University of Siena, Via A. Moro 2, I-53100 Siena, Italy.
| | | | | |
Collapse
|
123
|
Gerhard M, Schmees C, Voland P, Endres N, Sander M, Reindl W, Rad R, Oelsner M, Decker T, Mempel M, Hengst L, Prinz C. A secreted low-molecular-weight protein from Helicobacter pylori induces cell-cycle arrest of T cells. Gastroenterology 2005; 128:1327-39. [PMID: 15887115 DOI: 10.1053/j.gastro.2005.03.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Although Helicobacter pylori is recognized by the human immune system, the bacteria are not eliminated and lead to a chronic inflammation of the gastric mucosa. METHODS We investigated the interaction of H. pylori with human lymphocytes. T and B lymphocytes were isolated from H. pylori-infected patients and stimulated with anti-CD3/CD28 or interleukin-6. RESULTS Proliferation of lymphocytes was abolished on co-incubation with different H. pylori strains (1-5 bacteria/cell) or with protein extracts of culture supernatants. Inhibition of proliferation was independent of known virulence factors. The factor is a protein or protein complex with an apparent molecular weight between 30 and 60 kilodaltons, clearly distinct from VacA. Although antigen-specific activation of T cells (as shown by nuclear factor of activated T cells [NFAT]-activation, interferon-gamma production, and CD25 or CD69 up-regulation) remained intact, cell-cycle analysis showed that S-phase entry of T cells was inhibited completely by H. pylori. Consequently, stimulated T cells arrested in the G1 phase of the cell cycle. Western blot analysis showed markedly reduced phosphorylation of the retinoblastoma protein (pRb), suggesting inhibition of G1 cyclin-dependent kinase activity. In line with this, activities of cyclin D3 and cyclin E were down-regulated, and levels of the cyclin-dependent kinase inhibitor p27Kip1 were increased. Mouse embryonic fibroblasts deficient in p27 showed a decrease in H. pylori-induced inhibition of cell proliferation, suggesting a central role for p27 in mediating H. pylori-induced G1 arrest. CONCLUSIONS Induction of cell-cycle arrest in lymphocytes may be of major significance for the chronic persistence of bacteria in the human stomach.
Collapse
Affiliation(s)
- Markus Gerhard
- Department of Medicine II, Technical University, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
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
|
125
|
Allen LAH, Beecher BR, Lynch JT, Rohner OV, Wittine LM. Helicobacter pylori disrupts NADPH oxidase targeting in human neutrophils to induce extracellular superoxide release. THE JOURNAL OF IMMUNOLOGY 2005; 174:3658-67. [PMID: 15749904 DOI: 10.4049/jimmunol.174.6.3658] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Helicobacter pylori (Hp) infection triggers a chronic influx of polymorphonuclear leukocyte neutrophils (PMNs) into the gastric mucosa. Although Hp reside in a neutrophil-rich environment, how these organisms evade phagocytic killing is largely unexplored. We now show that live Hp (strains 11637, 60190, DT61A, and 11916) are readily ingested by PMNs and induce a rapid and strong respiratory burst that is comparable to PMA. Relative to other particulate stimuli, Hp are more potent activators of PMNs than opsonized zymosan, Staphylococcus aureus, or Salmonella. Strikingly, biochemical and microscopic analyses demonstrate that Hp disrupt NADPH oxidase targeting such that superoxide anions are released into the extracellular milieu and do not accumulate inside Hp phagosomes. Specifically, nascent Hp phagosomes acquire flavocytochrome b558 but do not efficiently recruit or retain p47phox or p67phox. Superoxide release peaks at 16 min coincident with the appearance of assembled oxidase complexes in patches at the cell surface. Oxidant release is regulated by formalin-resistant and heat-sensitive bacterial surface factors distinct from urease and Hp(2-20). Following opsonization with fresh serum, Hp triggers a modest respiratory burst that is confined to the phagosome, and ingested bacteria are eliminated. We conclude that disruption of NADPH oxidase targeting allows unopsonized Hp to escape phagocytic killing, and our findings support the hypothesis that bacteria and PMNs act in concert to damage the gastric mucosa.
Collapse
Affiliation(s)
- Lee-Ann H Allen
- Department of Medicine and Inflammation Program, University of Iowa, Coralville, IA 52241, USA.
| | | | | | | | | |
Collapse
|
126
|
Unemo M, Aspholm-Hurtig M, Ilver D, Bergström J, Borén T, Danielsson D, Teneberg S. The sialic acid binding SabA adhesin of Helicobacter pylori is essential for nonopsonic activation of human neutrophils. J Biol Chem 2005; 280:15390-7. [PMID: 15689619 DOI: 10.1074/jbc.m412725200] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Infiltration of neutrophils and monocytes into the gastric mucosa is a hallmark of chronic gastritis caused by Helicobacter pylori. Certain H. pylori strains nonopsonized stimulate neutrophils to production of reactive oxygen species causing oxidative damage of the gastric epithelium. Here, the contribution of some H. pylori virulence factors, the blood group antigen-binding adhesin BabA, the sialic acid-binding adhesin SabA, the neutrophil-activating protein HP-NAP, and the vacuolating cytotoxin VacA, to the activation of human neutrophils in terms of adherence, phagocytosis, and oxidative burst was investigated. Neutrophils were challenged with wild type bacteria and isogenic mutants lacking BabA, SabA, HP-NAP, or VacA. Mutant and wild type strains lacking SabA had no neutrophil-activating capacity, demonstrating that binding of H. pylori to sialylated neutrophil receptors plays a pivotal initial role in the adherence and phagocytosis of the bacteria and the induction of the oxidative burst. The link between receptor binding and oxidative burst involves a G-protein-linked signaling pathway and downstream activation of phosphatidylinositol 3-kinase as shown by experiments using signal transduction inhibitors. Collectively our data suggest that the sialic acid-binding SabA adhesin is a prerequisite for the nonopsonic activation of human neutrophils and, thus, is a virulence factor important for the pathogenesis of H. pylori infection.
Collapse
Affiliation(s)
- Magnus Unemo
- Department of Clinical Microbiology, Orebro University Hospital
| | | | | | | | | | | | | |
Collapse
|
127
|
Brest P, Bétis F, Cuburu N, Selva E, Herrant M, Servin A, Auberger P, Hofman P. Increased rate of apoptosis and diminished phagocytic ability of human neutrophils infected with Afa/Dr diffusely adhering Escherichia coli strains. Infect Immun 2004; 72:5741-9. [PMID: 15385473 PMCID: PMC517549 DOI: 10.1128/iai.72.10.5741-5749.2004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The proinflammatory effect of Afa/Dr diffusely adhering Escherichia coli (Afa/Dr DAEC) strains have been recently demonstrated in vitro by showing that polymorphonuclear leukocyte (PMN) transepithelial migration is induced after bacterial colonization of apical intestinal monolayers. The effect of Afa/Dr DAEC-PMN interaction on PMN behavior has been not investigated. Because of the putative virulence mechanism of PMN apoptosis during infectious diseases and taking into account the high level of expression of the decay-accelerating factor (DAF, or CD55), the receptor of Afa/Dr DAEC on PMNs, we sought to determine whether infection of PMNs by Afa/Dr DAEC strains could promote cell apoptosis. We looked at the behavior of PMNs incubated with Afa/Dr DAEC strains once they had transmigrated across polarized monolayers of intestinal (T84) cells. Infection of PMNs by Afa/Dr DAEC strains induced PMN apoptosis characterized by morphological nuclear changes, DNA fragmentation, caspase activation, and a high level of annexin V expression. However, transmigrated and nontransmigrated PMNs incubated with Afa/Dr DAEC strains showed similar elevated global caspase activities. PMN apoptosis depended on their agglutination, induced by Afa/Dr DAEC, and was still observed after preincubation of PMNs with anti-CD55 and/or anti-CD66 antibodies. Low levels of phagocytosis of Afa/Dr DAEC strains were observed both in nontransmigrated and in transmigrated PMNs compared to that observed with the control E. coli DH5alpha strain. Taken together, these data strongly suggest that interaction of Afa/Dr DAEC with PMNs may increase the bacterial virulence both by inducing PMN apoptosis through an agglutination process and by diminishing their phagocytic capacity.
Collapse
|
128
|
Monack DM, Mueller A, Falkow S. Persistent bacterial infections: the interface of the pathogen and the host immune system. Nat Rev Microbiol 2004; 2:747-65. [PMID: 15372085 DOI: 10.1038/nrmicro955] [Citation(s) in RCA: 381] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Persistent bacterial infections involving Mycobacterium tuberculosis, Salmonella enterica serovar Typhi (S. typhi) and Helicobacter pylori pose significant public-health problems. Multidrug-resistant strains of M. tuberculosis and S. typhi are on the increase, and M. tuberculosis and S. typhi infections are often associated with HIV infection. This review discusses the strategies used by these bacteria during persistent infections that allow them to colonize specific sites in the host and evade immune surveillance. The nature of the host immune response to this type of infection and the balance between clearance of the pathogen and avoidance of damage to host tissues are also discussed.
Collapse
Affiliation(s)
- Denise M Monack
- Department of Microbiology and Immunology, Stanford School of Medicine, Stanford University, Stanford, California 94305, USA.
| | | | | |
Collapse
|
129
|
Zabaleta J, McGee DJ, Zea AH, Hernández CP, Rodriguez PC, Sierra RA, Correa P, Ochoa AC. Helicobacter pylori arginase inhibits T cell proliferation and reduces the expression of the TCR zeta-chain (CD3zeta). THE JOURNAL OF IMMUNOLOGY 2004; 173:586-93. [PMID: 15210820 DOI: 10.4049/jimmunol.173.1.586] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Helicobacter pylori infects approximately half the human population. The outcomes of the infection range from gastritis to gastric cancer and appear to be associated with the immunity to H. pylori. Patients developing nonatrophic gastritis present a Th1 response without developing protective immunity, suggesting that this bacterium may have mechanisms to evade the immune response of the host. Several H. pylori proteins can impair macrophage and T cell function in vitro through mechanisms that are poorly understood. We tested the effect of H. pylori extracts and live H. pylori on Jurkat cells and freshly isolated human normal T lymphocytes to identify possible mechanisms by which the bacteria might impair T cell function. Jurkat cells or activated T lymphocytes cultured with an H. pylori sonicate had a reduced proliferation that was not caused by T cell apoptosis or impairment in the early T cell signaling events. Instead, both the H. pylori sonicate and live H. pylori induced a decreased expression of the CD3zeta-chain of the TCR. Coculture of live H. pylori with T cells demonstrated that the wild-type strain, but not the arginase mutant rocF(-), depleted L-arginine and caused a decrease in CD3zeta expression. Furthermore, arginase inhibitors reversed these events. These results suggest that H. pylori arginase is not only important for urea production, but may also impair T cell function during infection.
Collapse
Affiliation(s)
- Jovanny Zabaleta
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
130
|
Gebert B, Fischer W, Haas R. The Helicobacter pylori vacuolating cytotoxin: from cellular vacuolation to immunosuppressive activities. Rev Physiol Biochem Pharmacol 2004; 152:205-20. [PMID: 15549607 DOI: 10.1007/s10254-004-0027-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Helicobacter pylori is a highly successful bacterial pathogen of humans, infecting the stomach of more than half of the world's population. The H. pylori infection results in chronic gastritis, eventually followed by peptic ulceration and, more rarely, gastric cancer. H. pylori has developed a unique set of virulence factors, actively supporting its survival in the special ecological niche of the human stomach. Vacuolating cytotoxin (VacA) and cytotoxin-associated antigen A (CagA) are two major bacterial virulence factors involved in host cell modulation. VacA, so far mainly regarded as a cytotoxin of the gastric epithelial cell layer, now turns out to be a potent immunomodulatory toxin, targeting the adapted immune system. Thus, in addition to the well-known vacuolating activity, VacA has been reported to induce apoptosis in epithelial cells, to affect B lymphocyte antigen presentation, to inhibit the activation and proliferation of T lymphocytes, and to modulate the T cell-mediated cytokine response.
Collapse
Affiliation(s)
- B Gebert
- Max von Pettenkofer Institut für Hygiene und Medizinische Mikrobiologie, LMU München Pettenkoferstr., München, Germany
| | | | | |
Collapse
|
131
|
Basu M, Czinn SJ, Blanchard TG. Absence of catalase reduces long-term survival of Helicobacter pylori in macrophage phagosomes. Helicobacter 2004; 9:211-6. [PMID: 15165256 DOI: 10.1111/j.1083-4389.2004.00226.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Some Helicobacter pylori strains can survive within macrophage phagosomes for up to 24 hours. The factors that play a role in this survival remain ill-defined. Therefore, the contribution of catalase in mediating the survival of H. pylori following phagocytosis was investigated in vitro. METHODS An isogenic, catalase-deficient strain of H. pylori was generated and tested for sensitivity to hydrogen peroxide and susceptibility to macrophage-mediated killing. RESULTS The isogenic, catalase-deficient strain of H. pylori was effectively killed by hydrogen peroxide within 3 minutes compared to wild-type H. pylori which maintained 100% survival up to 21 minutes. The catalase-deficient mutant was also significantly more susceptible to macrophage-mediated killing than the parent strain, even when the ratio of bacteria to macrophage was increased. CONCLUSION These results indicate that although some strains of H. pylori are capable of survival within the macrophage phagosome, survival is dependent on virulence factors such as catalase for evasion of innate host defense.
Collapse
Affiliation(s)
- Malini Basu
- Department of Pediatrics, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
132
|
Blaser MJ, Atherton JC. Helicobacter pylori persistence: biology and disease. J Clin Invest 2004. [PMID: 14755326 DOI: 10.1172/jci200420925] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Helicobacter pylori are bacteria that have coevolved with humans to be transmitted from person to person and to persistently colonize the stomach. Their population structure is a model for the ecology of the indigenous microbiota. A well-choreographed equilibrium between bacterial effectors and host responses permits microbial persistence and health of the host but confers risk of serious diseases, including peptic ulceration and gastric neoplasia.
Collapse
Affiliation(s)
- Martin J Blaser
- Department of Medicine, New York University School of Medicine, and New York Harbor Veterans Affairs Medical Center, New York 10016, USA.
| | | |
Collapse
|
133
|
Rittig MG, Shaw B, Letley DP, Thomas RJ, Argent RH, Atherton JC. Helicobacter pylori-induced homotypic phagosome fusion in human monocytes is independent of the bacterial vacA and cag status. Cell Microbiol 2004; 5:887-99. [PMID: 14641174 DOI: 10.1046/j.1462-5822.2003.00328.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Following reports that a VacA+cag+ toxigenic but not a VacA-cag- non-toxigenic Helicobacter pylori strain induced homotypic phagosome fusion in murine macrophages, we addressed that phenomenon in human cells. Mononuclear phagocytes and epitheloid cells were challenged with H. pylori strains of different vacA and cag genotypes and with VacA- and Cag- isogenic mutants, and chased in the absence or presence of signal transduction modulators. Electron microscopy revealed that, in monocytes: (i) homotypic phagosome fusion was frequently induced by all live H. pylori strains investigated but not by exogenous VacA; (ii) phagosomes containing bacteria fused, but not those containing latex beads; (iii) fusion resulted in communal compartments resembling giant multivesicular bodies; and (iv) formation of these compartments was blocked by inhibiting the host cell regulators PI 3-kinase, phospholipase C and p42 MAP kinase. Whereas some internalized bacteria remained viable 1 h after uptake, none survived a 24 h period. In contrast to monocytes, infected epitheloid cells rarely developed communal compartments. In combination, these results demonstrate that, in human monocytes, the H. pylori-induced homotypic phagosome fusion depends on neither the vacuolating cytotoxin VacA nor the cag pathogenicity island of H. pylori and does not result in prolonged intracellular survival.
Collapse
Affiliation(s)
- M G Rittig
- Centre for Biochemistry and Cell Biology, Institute of Infections, Immunity and Inflammation, University of Nottingham Meeical School, UK.
| | | | | | | | | | | |
Collapse
|
134
|
Abstract
Helicobacter pylori are bacteria that have coevolved with humans to be transmitted from person to person and to persistently colonize the stomach. Their population structure is a model for the ecology of the indigenous microbiota. A well-choreographed equilibrium between bacterial effectors and host responses permits microbial persistence and health of the host but confers risk of serious diseases, including peptic ulceration and gastric neoplasia.
Collapse
Affiliation(s)
- Martin J Blaser
- Department of Medicine, New York University School of Medicine, and New York Harbor Veterans Affairs Medical Center, New York 10016, USA.
| | | |
Collapse
|
135
|
Abstract
PURPOSE OF REVIEW Helicobacter pylori remains one of the world's most prevalent bacterial pathogens, often causing gastritis, peptic ulcer disease, gastric mucosa-associated lymphatic tissue lymphoma, or gastric adenocarcinoma. Elucidation of H. pylori virulence mechanisms and characteristics of the host that contribute to pathogenesis will facilitate the development of both pharmacologic and immunologic therapies. RECENT FINDINGS The functional status of the outer inflammatory protein A may have predictive value for duodenal ulcer, and host alleles for interleukin-1beta, interleukin-1R, tumor necrosis factor-alpha, and interleukin-10 correlate with increased risk for gastric cancer. H. pylori vacuolating cytotoxin A and cytotoxin-associated gene A protein interact with multiple host proteins, although downstream signaling events need further characterization. It does appear however, that CagA may participate in a negative feedback loop on Src family kinases to prevent further phosphorylation of CagA. Several models, including delayed type hypersensitivity in immune mice, and spontaneous clearance of H. pylori from interleukin-10 and phagocyte oxidase mice, provide evidence that severe inflammation may be sufficient to eradicate H. pylori. The strategies used by H. pylori to avoid the inflammatory response are also becoming clearer. H. pylori remains viable when internalized by epithelial cells, and it remains viable in macrophage phagosomes by inhibiting phagosome maturation. Additionally, H. pylori may regulate the host immune response through activation of dendritic cells and CD25 regulatory T cells, and it may direct immunosuppression of T cells. SUMMARY Helicobacter pylori virulence is accomplished through many mechanisms, including vacuolating cytotoxin A and CagA activities, and may be predicted based on bacterial and host genotypes. Ultimately, H. pylori persistence may depend on its success in downregulating the inflammatory response.
Collapse
Affiliation(s)
- Thomas G Blanchard
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.
| | | | | |
Collapse
|
136
|
Abstract
Helicobacter pylori is a global pathogen that causes severe gastrointestinal diseases leading to a significant morbidity and mortality. There is an effective treatment for peptic ulcer disease, however, this is being compromised by an increase in the prevalence of antibiotic resistance. Although alternative rescue regimens have been advocated, the best strategy would be to prevent disease, especially in the case of gastric cancer for which there is still no treatment. One approach is to inhibit the first step in the pathogenic process - adhesion of the organism to the host tissue. Another and probably a better approach is vaccination, but clinical trials have so far been unsuccessful. There is still a large uncertainty in relation to how H. pylori causes disease. Knowledge from genomics, proteomics, and the relationship between polymorphism of the bacterium and the host, as well as the continuing investigation of the role played by important virulence factors in the outcome of the disease, will help both in understanding pathogenesis of disease and in the design of the best vaccine.
Collapse
Affiliation(s)
- Rachel O'Mahony
- Centre for Infectious Diseases & International Health, RF&UCL Medical School, UK
| | | | | | | |
Collapse
|
137
|
Rittig MG, Kaufmann A, Robins A, Shaw B, Sprenger H, Gemsa D, Foulongne V, Rouot B, Dornand J. Smooth and rough lipopolysaccharide phenotypes of Brucella induce different intracellular trafficking and cytokine/chemokine release in human monocytes. J Leukoc Biol 2003; 74:1045-55. [PMID: 12960272 DOI: 10.1189/jlb.0103015] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Virulence of the intracellular pathogen Brucella for humans is mainly associated with its lipopolysaccharide (LPS) phenotype, with smooth LPS phenotypes generally being virulent and rough ones not. The reason for this association is not quite understood. We now demonstrate by flow cytometry, electron microscopy, and ELISA that human peripheral blood monocytes interact both quantitatively and qualitatively different with smooth and rough Brucella organisms in vitro. We confirm that considerably higher numbers of rough than smooth brucellae attach to and enter the monocytes in nonopsonic conditions; but only smooth brucellae replicate in the host cells. We show for the first time that rough brucellae induce higher amounts than smooth brucellae of several CXC (GRO-alpha, IL-8) and CC (MIP-1alpha, MIP-1beta, MCP-1, RANTES) chemokines, as well as pro- (IL-6, TNF-alpha) and anti-inflammatory (IL-10) cytokines released by challenged monocytes. Upon uptake, phagosomes containing rough brucellae develop selective fusion competence to form spacious communal compartments, whereas phagosomes containing smooth brucellae are nonfusiogenic. Collectively, our data suggest that rough brucellae attract and infect monocytes more effectively than smooth brucellae, but only smooth LPS phenotypes establish a specific host cell compartment permitting successful parasitism. These novel findings link the LPS phenotype of Brucella and its virulence for humans at the level of the infected host cells. Whether this is due to a direct effect of the LPS molecules or to upstream bacterial mechanisms remains to be established.
Collapse
Affiliation(s)
- Michael G Rittig
- School of Biomedical Sciences, University of Nottingham Medical School, Nottingham NG7 2UH, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Sukumaran SK, Shimada H, Prasadarao NV. Entry and intracellular replication of Escherichia coli K1 in macrophages require expression of outer membrane protein A. Infect Immun 2003; 71:5951-61. [PMID: 14500515 PMCID: PMC201085 DOI: 10.1128/iai.71.10.5951-5961.2003] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interactions between Escherichia coli K1, which causes meningitis in neonates, and macrophages have not been explored well. In this study we found that E. coli K1 was able to enter, survive, and replicate intracellularly in both murine and human macrophage cell lines, as well as in monocytes and macrophages of newborn rats. In addition, we demonstrated that OmpA (+) E. coli also enters and replicates in human peripheral blood monocytes in vitro. Outer membrane protein A (OmpA) expression on E. coli contributes to binding to macrophages, phagocytosis, and survival within macrophages. Opsonization with either complement proteins or antibody is not required for uptake and survival of the bacteria within the macrophages. Transmission electron microscopy and immunocytochemistry studies with the infected macrophages indicated that OmpA(+) E. coli multiplies enormously in a single phagosome and bursts the cell. Internalization of OmpA(+) E. coli by RAW 264.7 cells occurred by both actin- and microtubule-dependent processes, which are independent of RGD-mediated integrin receptors. Internalization and intracellular survival within phagocytic cells thus may play an important role in the development of bacteremia, which is crucial for E. coli crossing of the blood-brain barrier.
Collapse
Affiliation(s)
- Sunil K Sukumaran
- Division of Infectious Diseases, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, 4650 Sunset Boulevard, Los Angeles, CA 90027, USA
| | | | | |
Collapse
|
139
|
Abstract
Few microorganisms evade killing by neutrophils. Summarized here are the mechanisms used by Yersinia, group A streptococci, Helicobacter, Ehrlichia and Francisella to block phagocytosis, disrupt phagosome maturation or perturb the respiratory burst. Also discussed are mechanisms used by neutrophils to control organisms that replicate inside macrophages.
Collapse
Affiliation(s)
- Lee-Ann H Allen
- Department of Medicine and the Inflammation Program, MTF D154, University of Iowa and the VA Medical Center, 2501 Crosspark Road, Coralville, Iowa City, IA 55241, USA.
| |
Collapse
|
140
|
Abstract
Macropinocytosis (fluid uptake) and phagocytosis (particle uptake) are processes that result in the formation of intracellular membrane enclosed vacuoles termed macropinosomes and phagosomes, respectively. Macropinosomes and phagosomes are modified by fission and fusion reactions with the endo-lysosomal pathway that eventually transform these vacuoles into a lysosomal environment. Many human bacterial pathogens, including species of Mycobacteria, Legionella, and Chlamydia, are thought to survive by disrupting the normal membrane trafficking events that usually result in the formation of phago-lysosomes and death of the microorganism. In addition, a number of important pathogens facilitate homotypic phagosome fusion in order to generate an intracellular environment conducive for survival. A greater understanding of the regulation of phagosomal maturation and fusion will be critical in designing new therapies to treat infections caused by intracellular pathogens. The genetically tractable phagocyte, D. discoideum, has proven extremely useful in dissecting the signaling pathways regulating macropinocytosis, phagocytosis, phagosomal maturation and phagosome-phagosome fusion. A body of knowledge has accumulated and demonstrates important roles for Rab GTPases, the cytoskeleton, phosphoinositide metabolism and pH regulation in regulating phagosome maturation. This review will summarize the current state of knowledge.
Collapse
Affiliation(s)
- Damian Duhon
- Department of Microbiology and Immunology, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, USA
| | | |
Collapse
|
141
|
Guiney DG, Hasegawa P, Cole SP. Helicobacter pylori preferentially induces interleukin 12 (IL-12) rather than IL-6 or IL-10 in human dendritic cells. Infect Immun 2003; 71:4163-6. [PMID: 12819109 PMCID: PMC161996 DOI: 10.1128/iai.71.7.4163-4166.2003] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Dendritic cells are potent antigen-presenting cells that are present in the gastrointestinal tract and are required for the induction of a Th1 T-cell acquired immune response. Since infection with the gastric pathogen Helicobacter pylori elicits a Th1 cell response, the interaction of these organisms with dendritic cells should reflect the Th1 bias. We incubated H. pylori with cultured human dendritic cells and measured the cytokine induction profile, comparing the response to that induced by Salmonella enterica serovar Typhimurium. We found that H. pylori induced little interleukin 6 (IL-6) and essentially no IL-10 in contrast to S. enterica. However, H. pylori induced levels of IL-12 that were 30% of those induced by S. enterica, indicating a Th1 response. An isogenic cagE mutant of H. pylori lost about 50% of its IL-12-inducing ability, suggesting a role for the cag type IV secretion system in the stimulation of dendritic cells.
Collapse
Affiliation(s)
- Donald G Guiney
- Department of Medicine, University of California, San Diego, La Jolla, California 92093-0640, USA.
| | | | | |
Collapse
|
142
|
Petersen AM, Krogfelt KA. Helicobacter pylori: an invading microorganism? A review. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2003; 36:117-26. [PMID: 12738380 DOI: 10.1016/s0928-8244(03)00020-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this review we evaluate the pros and cons of Helicobacter pylori invasion of epithelial cells as part of the natural history of H. pylori infection. H. pylori is generally considered an extracellular microorganism. However, a growing body of evidence supports the controversial hypothesis that at least a subset of H. pylori microorganisms has an intracellular (intraepithelial) location. Most significant is the fact that H. pylori invades cultured epithelial cells with invasion frequencies similar to Yersinia enterocolitica and better than Shigella flexneri; furthermore, studies of invasion mechanisms suggest that H. pylori invasion of and survival within epithelial cells is not merely a passive event, but requires active participation of the microorganism. Although many studies of human gastric biopsy specimens have failed to demonstrate any intracellular H. pylori, some studies have revealed a minor fraction of H. pylori inside gastric epithelial cells, with possible linkage to peptic ulceration and epithelial cell damage. In conclusion, these data encourage further research to establish whether intracellular H. pylori does play a role in H. pylori colonization of the human stomach and in peptic ulcer pathogenesis.
Collapse
Affiliation(s)
- Andreas Munk Petersen
- Department of Gastrointestinal Infections, Statens Serum Institut, 5 Artillerivej, 2300 Copenhagen S, Denmark
| | | |
Collapse
|
143
|
Zheng PY, Jones NL. Helicobacter pylori strains expressing the vacuolating cytotoxin interrupt phagosome maturation in macrophages by recruiting and retaining TACO (coronin 1) protein. Cell Microbiol 2003; 5:25-40. [PMID: 12542468 DOI: 10.1046/j.1462-5822.2003.00250.x] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recent evidence suggests that persistence of Helicobacter pylori can be explained, at least in part, by the failure of macrophages to kill bacteria. The fate of type 1 H. pylori strain LC11, which expresses the cag pathogenicity island (PAI) and the vacuolating cytotoxin, and type 2 strain LC20, which lacks both these virulence factors, was determined following infection of the murine macrophage cell line RAW 264.7 or the human macrophage-like cell line THP-1. Helicobacter pylori strain LC11 displayed enhanced survival in macrophages in comparison with strain LC20 (4.0 +/- 0.2 versus 2.1 +/- 0.6 log CFU ml-1, P < 0.01) at 24 h. Phagosomes containing strain LC11 showed reduced co-localization with LysoTracker Red, higher levels of expression of the early endosome marker EEA1 expression and lower expression of the late endosome/lysosome marker LAMP1 relative to internalized strain LC20, both at 2 h and 24 h. These findings indicate that, in contrast to strain LC20, strain LC11 resides in a compartment with early endosome properties and does not fuse with lysosomes. In addition, phagosomes containing LC11 recruited and retained a higher percentage of TACO (coronin 1) protein in comparison with phagosomes containing strain LC20. Furthermore, IFN-gamma stimulation facilitated maturation of phagosomes containing strain LC11 in association with the release of TACO and a reduction in bacterial survival. We have demonstrated through the use of isogenic cagA-, cagE-/picB- and vacA- mutant strains, that VacA plays a significant role in the interruption of the phagosome maturation. Taken together, these results indicate that, following phagocytosis, H. pylori strains expressing the vacuolating cytotoxin arrest phagosome maturation in association with the retention of TACO.
Collapse
Affiliation(s)
- Peng-Yuan Zheng
- Research Institute, The Hospital for Sick Children, Departments of Pediatrics and Physiology, University of Toronto, 555 University Avenue, Toronto, Ontario, M5G 1X8 Canada
| | | |
Collapse
|
144
|
Allen LAH, Allgood JA. Atypical protein kinase C-zeta is essential for delayed phagocytosis of Helicobacter pylori. Curr Biol 2002; 12:1762-6. [PMID: 12401171 DOI: 10.1016/s0960-9822(02)01216-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Phagocytosis is a rapid actin-dependent endocytic process used by macrophages and neutrophils to ingest and kill microorganisms. Perturbation of phagocytosis is central to the ability of some pathogenic microbes to cause disease, and we demonstrated previously that the ulcerogenic bacterium Helicobacter pylori (Hp) actively retards its uptake by macrophages and subsequently persists inside novel vacuoles called megasomes. Neither the receptor that mediates Hp binding nor the signaling pathways that regulate bacterial engulfment have been defined. Nevertheless, the fact that other phagocytic stimuli do not exhibit delayed phagocytosis suggests that Hp may be ingested by a unique mechanism. We now show that Hp transiently activated protein kinase C (PKC) in macrophages and that atypical PKCzeta and novel PKC(epsilon), but not conventional PKC(alpha), accumulated on forming phagosomes. Pharmacologic agents, isoform-selective pseudosubstrate peptides, and antisense oligonucleotides demonstrated that PKC(zeta) regulated local actin polymerization and bacterial engulfment, whereas other PKC isoforms did not. In contrast, opsonization of Hp with immunoglobulin G (IgG) induced rapid PKC(zeta)-independent uptake and enhanced killing of ingested bacteria. A role for atypical PKCs in phagocytosis has not been described. We conclude that Hp defines a new phagocytic pathway in macrophages that is regulated by PKC(zeta).
Collapse
Affiliation(s)
- Lee Ann H Allen
- Department of Medicine and The Inflammation Program, The University of Iowa and The Veterans Affairs Medical Center, Iowa City, IA 52242, USA.
| | | |
Collapse
|
145
|
Amieva MR, Salama NR, Tompkins LS, Falkow S. Helicobacter pylori enter and survive within multivesicular vacuoles of epithelial cells. Cell Microbiol 2002; 4:677-90. [PMID: 12366404 DOI: 10.1046/j.1462-5822.2002.00222.x] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Although intracellular Helicobacter pylori have been described in biopsy specimens and in cultured epithelial cells, the fate of these bacteria is unknown. Using differential interference contrast (DIC) video and immunofluorescence microscopy, we document that a proportion of cell-associated H. pylori enter large cytoplasmic vacuoles, where they remain viable and motile and can survive lethal concentrations of extracellular gentamicin. Entry into vacuoles occurs in multiple epithelial cell lines including AGS gastric adenocarcinoma, Caco-2 colon adenocarcinoma and MDCK kidney cell line, and depends on the actin cytoskeleton. Time-lapse microscopy over several hours was used to follow the movement of live H. pylori within vacuoles of a single cell. Pulsed, extracellular gentamicin treatments show that the half-life of intravacuolar bacteria is on the order of 24 h. Viable H. pylori repopulate the extracellular environment in parallel with the disappearance of intravacuolar bacteria, suggesting release from the intravacuolar niche. Using electron microscopy and live fluorescent staining with endosomal dyes, we observe that H. pylori-containing vacuoles are similar in morphology to late endosomal multivesicular bodies. VacA is not required for these events, as isogenic vacA- mutants still enter and survive within the intravacuolar niche. The exploitation of an intravacuolar niche is a new aspect of the biological life cycle of H. pylori that could explain the difficulties in eradicating this infection.
Collapse
Affiliation(s)
- Manuel R Amieva
- Department of Microbiology and Immunology, Stanford University School of Medicine, 299 Campus Drive, Stanford, CA 94305, USA.
| | | | | | | |
Collapse
|
146
|
Harris E, Cardelli J. RabD, a Dictyostelium Rab14-related GTPase, regulates phagocytosis and homotypic phagosome and lysosome fusion. J Cell Sci 2002; 115:3703-13. [PMID: 12186956 DOI: 10.1242/jcs.00050] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
RabD, a Dictyostelium Rab14-related GTPase, localizes in the endo-lysosomal pathway and contractile vacuole system of membranes. Cell lines expressing dominant-negative RabD were defective in endocytosis, endosomal membrane flow and homotypic lysosome fusion. In support of a role for RabD in fusion, cells overexpressing constitutively active RabD(Q67L) accumulated enlarged hydrolase-rich acidic vesicles ringed with GFP-RabD, consistent with RabD directly regulating lysosome fusion. To determine whether RabD also regulated phagocytosis and/or homotypic phagosome fusion (a process stimulated by many intracellular pathogens), cells overexpressing dominant-active (RabD(Q67L)) or dominant-negative (Rab(N121I)) RabD were analyzed microscopically and biochemically. The rate of phagocytosis was increased two-fold in RabD(Q67L)-expressing cells and reduced by 50% in RabD(N121I)-expressing cells compared with control cells. To examine the role of RabD in the formation of multiparticle phagosomes, we performed a series of pulse-chase experiments using fluorescently labeled bacteria and fluorescent latex beads. The rate of fusion of newly formed phagosomes was five times higher in the RabD(Q67L)-expressing cells and reduced by over 50% in RabD(N121I)-expressing cells as compared with control cells. GFP-RabD(Q67L) was found to ring multiparticle spacious phagosomes, which supports a direct role for this protein in regulating fusion. Inhibition of PI 3-kinase activity, which is known to regulate phagosome fusion in the wild-type cells, reduced the rate of phagosome fusion in RabD(Q67L+) cells, indicating that RabD acted upstream of or parallel with PI 3-kinase. We hypothesize that RabD and, possibly, Rab14, a related GTPase that associates with phagosomes in mammalian cells, are important regulators of homotypic phagosome and endo-lysosome fusion.
Collapse
Affiliation(s)
- Edward Harris
- Department of Microbiology and Immunology and Feist-Weiller Cancer Center, LSU Health Sciences Center, Shreveport, LA 71130, USA
| | | |
Collapse
|
147
|
Fischer W, Haas R, Odenbreit S. Type IV secretion systems in pathogenic bacteria. Int J Med Microbiol 2002; 292:159-68. [PMID: 12398207 DOI: 10.1078/1438-4221-00199] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Wolfgang Fischer
- Max von Pettenkofer-Institut für Hygiene und Mikrobiologie, Ludwig-Maximilians-Universität München, Germany.
| | | | | |
Collapse
|
148
|
Moese S, Selbach M, Meyer TF, Backert S. cag+ Helicobacter pylori induces homotypic aggregation of macrophage-like cells by up-regulation and recruitment of intracellular adhesion molecule 1 to the cell surface. Infect Immun 2002; 70:4687-91. [PMID: 12117984 PMCID: PMC128178 DOI: 10.1128/iai.70.8.4687-4691.2002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Infection with cag+ but not cag-negative Helicobacter pylori leads to the formation of large homotypic aggregates of macrophage-like cells. Intracellular adhesion molecule 1 is up-regulated and recruited to the cell surface of infected cells and mediates the aggregation via lymphocyte function-associated molecule 1. This signaling may regulate cell-cell interactions and inflammatory responses.
Collapse
Affiliation(s)
- Stefan Moese
- Abteilung Molekulare Biologie, Max-Planck-Institut für Infektionsbiologie, D-10117 Berlin, Germany
| | | | | | | |
Collapse
|
149
|
Watarai M, Makino SI, Fujii Y, Okamoto K, Shirahata T. Modulation of Brucella-induced macropinocytosis by lipid rafts mediates intracellular replication. Cell Microbiol 2002; 4:341-55. [PMID: 12067319 DOI: 10.1046/j.1462-5822.2002.00195.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Intracellular replication of Brucella requires the VirB complex, which is highly similar to conjugative DNA transfer systems. In this study, we show that Brucella internalizes into macrophages by swimming on the cell surface with generalized membrane ruffling for several minutes, after which the bacteria are enclosed by macropinosomes. Lipid raft-associated molecules such as glycosylphosphatidylinositol (GPI)-anchored proteins, GM1 gangliosides and cholesterol were selectively incorporated into macropinosomes containing Brucella. In contrast, lysosomal glycoprotein LAMP-1 and host cell transmembrane protein CD44 were excluded from the macropinosomes. Removing GPI-anchored proteins from the macrophage surface and cholesterol sequestration markedly inhibited the VirB-dependent macropinocytosis and intracellular replication. Our results suggest that the entry route of Brucella into the macrophage determines the intracellular fate of the bacteria that is modulated by lipid raft microdomains.
Collapse
Affiliation(s)
- Masahisa Watarai
- Department of Veterinary Microbiology, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan.
| | | | | | | | | |
Collapse
|
150
|
Abstract
Phagocytosis constitutes the primary line of host innate and adaptive defence against incoming microbial pathogens, providing an efficient means for their removal and destruction. However, several virulent bacteria that do not function as intracellular pathogens have evolved mechanisms to avoid and prevent phagocytosis that constitute an essential part of their pathogenic capacity. Some of these mechanisms include preventing recognition by phagocytic receptors or blocking uptake by professional phagocytes. Recently, the molecular mechanisms of such antiphagocytic properties have been elucidated for some pathogens. Such mechanisms illustrate the diversity of mechanisms bacterial pathogens use to avoid phagocytic uptake.
Collapse
Affiliation(s)
- Jean Celli
- Biotechnology Laboratory, University of British Columbia, Room 237, 6174 University Boulevard, Vancouver, BC, Canada V6T 1Z3.
| | | |
Collapse
|