1
|
Interaction of Macrophages and Cholesterol-Dependent Cytolysins: The Impact on Immune Response and Cellular Survival. Toxins (Basel) 2020; 12:toxins12090531. [PMID: 32825096 PMCID: PMC7551085 DOI: 10.3390/toxins12090531] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) are key virulence factors involved in many lethal bacterial infections, including pneumonia, necrotizing soft tissue infections, bacterial meningitis, and miscarriage. Host responses to these diseases involve myeloid cells, especially macrophages. Macrophages use several systems to detect and respond to cholesterol-dependent cytolysins, including membrane repair, mitogen-activated protein (MAP) kinase signaling, phagocytosis, cytokine production, and activation of the adaptive immune system. However, CDCs also promote immune evasion by silencing and/or destroying myeloid cells. While there are many common themes between the various CDCs, each CDC also possesses specific features to optimally benefit the pathogen producing it. This review highlights host responses to CDC pathogenesis with a focus on macrophages. Due to their robust plasticity, macrophages play key roles in the outcome of bacterial infections. Understanding the unique features and differences within the common theme of CDCs bolsters new tools for research and therapy.
Collapse
|
2
|
Shewell LK, Day CJ, Jen FEC, Haselhorst T, Atack JM, Reijneveld JF, Everest-Dass A, James DBA, Boguslawski KM, Brouwer S, Gillen CM, Luo Z, Kobe B, Nizet V, von Itzstein M, Walker MJ, Paton AW, Paton JC, Torres VJ, Jennings MP. All major cholesterol-dependent cytolysins use glycans as cellular receptors. SCIENCE ADVANCES 2020; 6:eaaz4926. [PMID: 32494740 PMCID: PMC7244308 DOI: 10.1126/sciadv.aaz4926] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/17/2020] [Indexed: 05/03/2023]
Abstract
Cholesterol-dependent cytolysins (CDCs) form pores in cholesterol-rich membranes, but cholesterol alone is insufficient to explain their cell and host tropism. Here, we show that all eight major CDCs have high-affinity lectin activity that identifies glycans as candidate cellular receptors. Streptolysin O, vaginolysin, and perfringolysin O bind multiple glycans, while pneumolysin, lectinolysin, and listeriolysin O recognize a single glycan class. Addition of exogenous carbohydrate receptors for each CDC inhibits toxin activity. We present a structure for suilysin domain 4 in complex with two distinct glycan receptors, P1 antigen and αGal/Galili. We report a wide range of binding affinities for cholesterol and for the cholesterol analog pregnenolone sulfate and show that CDCs bind glycans and cholesterol independently. Intermedilysin binds to the sialyl-TF O-glycan on its erythrocyte receptor, CD59. Removing sialyl-TF from CD59 reduces intermedilysin binding. Glycan-lectin interactions underpin the cellular tropism of CDCs and provide molecular targets to block their cytotoxic activity.
Collapse
Affiliation(s)
- Lucy K. Shewell
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia
| | - Christopher J. Day
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia
| | - Freda E.-C. Jen
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia
| | - Thomas Haselhorst
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia
| | - John M. Atack
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia
| | | | - Arun Everest-Dass
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia
| | - David B. A. James
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | | | - Stephan Brouwer
- School of Chemistry and Molecular Biosciences and the Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Christine M. Gillen
- School of Chemistry and Molecular Biosciences and the Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Zhenyao Luo
- School of Chemistry and Molecular Biosciences and the Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences and the Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia
| | - Mark J. Walker
- School of Chemistry and Molecular Biosciences and the Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Adrienne W. Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide 5005, Australia
| | - James C. Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide 5005, Australia
| | - Victor J. Torres
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Michael P. Jennings
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia
| |
Collapse
|
3
|
Cavaillon JM. Exotoxins and endotoxins: Inducers of inflammatory cytokines. Toxicon 2018; 149:45-53. [PMID: 29056305 DOI: 10.1016/j.toxicon.2017.10.016] [Citation(s) in RCA: 232] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 12/26/2022]
Abstract
Endotoxins and exotoxins are among the most potent bacterial inducers of cytokines. During infectious processes, the production of inflammatory cytokines including tumor necrosis factor (TNF), interleukin-1β (IL-1β), gamma interferon (IFNγ) and chemokines orchestrates the anti-infectious innate immune response. However, an overzealous production, leading up to a cytokine storm, can be deleterious and contributes to mortality consecutive to sepsis or toxic shock syndrome. Endotoxins of Gram-negative bacteria (lipopolysaccharide, LPS) are particularly inflammatory because they generate auto-amplificatory loops after activation of monocytes/macrophages. LPS and numerous pore-forming exotoxins also activate the inflammasome, the molecular platform that allows the release of mature IL-1β and IL-18. Among exotoxins, some behave as superantigens, and as such activate the release of cytokines by T-lymphocytes. In most cases, pre-exposure to exotoxins enhances the cytokine production induced by LPS and its lethality, whereas pre-exposure to endotoxin usually results in tolerance. In this review we recall the various steps, which, from the very early discovery of pyrogenicity induced by bacterial products, ended to the discovery of the endogenous pyrogen. Furthermore, we compare the specific characteristics of endotoxins and exotoxins in their capacity to induce inflammatory cytokines.
Collapse
Affiliation(s)
- Jean-Marc Cavaillon
- Unit Cytokines & Inflammation, Institut Pasteur, 28 Rue Dr. Roux, 75015, Paris, France.
| |
Collapse
|
4
|
An H, Wang Z, Chen H, Wang T, Wang X, Liu L, Liu X, Xu J, He L, Zhang K, Zhang H, Liu X. Clinical efficacy of short-course chemotherapy combined with topical injection therapy in treatment of superficial lymph node tuberculosis. Oncotarget 2017; 8:109889-109893. [PMID: 29299116 PMCID: PMC5746351 DOI: 10.18632/oncotarget.22492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/28/2017] [Indexed: 11/25/2022] Open
Abstract
To evaluate the clinical efficacy and safety of short-course chemotherapy combined with regional injection therapy in the treatment of superficial lymph node tuberculosis. 201 patients diagnosed with superficial lymph node tuberculosis were retrospectively analyzed. All patients were randomly divided into the study (n = 100) and control groups (n = 101). In the study group, the patients received 6-month chemotherapy with isoniazid (H), rifampin (R) and ethambutol (E) (6HRE) in combination with regional injection of streptomycin, and their counterparts in the control group underwent systemic regime of 3HRZE/6HRE. In the study group, the overall cure rate was calculated as 98% and the recurrence rate was 2%. Twenty-four of 25 nodular type patients and 36 among 37 inflammatory type patients were recovered and discharged. One patient with huge nodular type mass was treated for 4 months and the mass size was slightly reduced. In the control group, the overall cure rate was 48.5% and the recurrence rate was 7.9%. The recurrent patients were further administered with regional injection of streptomycin based upon the chemotherapy regime until they were recovered. Combined therapy of systemic chemotherapy and regional injection of streptomycin is probably an efficacious and safe approach in the treatment of superficial lymph node tuberculosis, which remains to be validated by more investigations.
Collapse
Affiliation(s)
- Huiru An
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Zhongyuan Wang
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Hongbing Chen
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Tao Wang
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Xinjing Wang
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Lin Liu
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Xiao Liu
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Jing Xu
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Luxing He
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Kai Zhang
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Hongyan Zhang
- Military Institute of Tuberculosis, Beijing, 100091, China
| | - Xinying Liu
- Military Institute of Tuberculosis, Beijing, 100091, China
| |
Collapse
|
5
|
Huang T, Song X, Zhao K, Jing J, Shen Y, Zhang X, Yue B. Quorum-sensing molecules N-acyl homoserine lactones inhibit Trueperella pyogenes infection in mouse model. Vet Microbiol 2017; 213:89-94. [PMID: 29292009 DOI: 10.1016/j.vetmic.2017.11.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/19/2017] [Accepted: 11/19/2017] [Indexed: 11/24/2022]
Abstract
Trueperella pyogenes is a gram-positive opportunistic pathogen normally causes mastitis, liver abscesses and pneumonia of economically important livestock. It has been suggested that gram-negative bacteria can suppress the growth and virulence of T. pyogenes in vitro by using the quorum-sensing (QS) signal molecules and cause the transition of predominant species. However, whether these QS signals can be used as potential anti-virulence drugs against T. pyogenes infection is unclear. In this study, the in vivo inhibitory effect N-acyl homoserine lactones (AHLs) from Escherichia coli and Pseudomonas aeruginosa on T. pyogenes was tested by using mouse model. Mice were first peritoneally infected with T. pyogenes followed by intravenous injection of N-Octanoyl-DL-homoserine lactone (C8HSL) or N-(3-oxododecanoyl) homoserine-l-lactone (C12HSL). The results showed that C8HSL and C12HSL significantly reduced bacterial load and increased the survival rate of mice against T. pyogenes challenge. Additionally, the treatment of AHLs promoted the secretion of IL-1β, IL-6, IL-8 and TNF-α in mouse peritoneal fluid, and significantly decreased the expression levels of virulence genes of residual T. pyogenes. Importantly, murine macrophages rapidly phagocytosed bacteria when they were treated with AHLs compared to untreated cells. Collectively, our findings provide a major advance in understanding the inhibitory effect of AHLs in vivo and a promise for developing new clinical or veterinary treatments of T. pyogenes-related infection.
Collapse
Affiliation(s)
- Ting Huang
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Xuhao Song
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, China
| | - Kelei Zhao
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Jie Jing
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Yongmei Shen
- Sichuan Engineering Technology Research Center of Medical Animal, Chengdu, China
| | - Xiuyue Zhang
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, China
| | - Bisong Yue
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Sowa JE, Ślusarczyk J, Trojan E, Chamera K, Leśkiewicz M, Regulska M, Kotarska K, Basta-Kaim A. Prenatal stress affects viability, activation, and chemokine signaling in astroglial cultures. J Neuroimmunol 2017; 311:79-87. [DOI: 10.1016/j.jneuroim.2017.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/26/2017] [Accepted: 08/18/2017] [Indexed: 01/04/2023]
|
7
|
Hernández-Flores KG, Calderón-Garcidueñas AL, Mellado-Sánchez G, Ruiz-Ramos R, Sánchez-Vargas LA, Thomas-Dupont P, Izaguirre-Hernández IY, Téllez-Sosa J, Martínez-Barnetche J, Wood L, Paterson Y, Cedillo-Barrón L, López-Franco O, Vivanco-Cid H. Evaluation of the safety and adjuvant effect of a detoxified listeriolysin O mutant on the humoral response to dengue virus antigens. Clin Exp Immunol 2017; 188:109-126. [PMID: 27886660 DOI: 10.1111/cei.12906] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2016] [Indexed: 01/14/2023] Open
Abstract
Listeriolysin O (LLO) has been proposed as a potential carrier or adjuvant molecule in the vaccination field. However, the cytotoxic and pro-apoptotic effects of LLO are the major limitations for this purpose. Here, we have performed a preclinical safety evaluation and characterized a new potential adjuvant application for a non-cytolytic LLO mutant (dtLLO) to enhance and modulate the immune response against the envelope (E) protein from dengue virus. In addition, we have studied the adjuvant effects of dtLLO on human immune cells and the role of membrane cholesterol for the binding and proinflammatory property of the toxoid. Our in-vivo results in the murine model confirmed that dtLLO is a safer molecule than wild-type LLO (wtLLO), with a significantly increased survival rate for mice challenged with dtLLO compared with mice challenged with wtLLO (P < 0·001). Histopathological analysis showed non-toxic effects in key target organs such as brain, heart, liver, spleen, kidney and lung after challenge with dtLLO. In vitro, dtLLO retained the capacity of binding to plasma membrane cholesterol on the surface of murine and human immune cells. Immunization of 6-8-week-old female BALB/c mice with a combination of dtLLO mixed with E protein elicited a robust specific humoral response with isotype diversification of immunoglobulin (Ig)G antibodies (IgG1 and IgG2a). Finally, we demonstrated that cholesterol and lipid raft integrity are required to induce a proinflammatory response by human cells. Taken together, these findings support a potential use of the dtLLO mutant as a safe and effective adjuvant molecule in vaccination.
Collapse
Affiliation(s)
- K G Hernández-Flores
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz City, Veracruz, México.,Doctorado en Ciencias Biomédicas, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa Veracruz, México
| | | | - G Mellado-Sánchez
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz City, Veracruz, México
| | - R Ruiz-Ramos
- Instituto de Medicina Forense, Universidad Veracruzana, Boca del Río Veracruz, México
| | - L A Sánchez-Vargas
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz City, Veracruz, México.,Doctorado en Ciencias Biomédicas, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa Veracruz, México
| | - P Thomas-Dupont
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz City, Veracruz, México.,Doctorado en Ciencias Biomédicas, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa Veracruz, México
| | - I Y Izaguirre-Hernández
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz City, Veracruz, México.,Doctorado en Ciencias Biomédicas, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa Veracruz, México
| | - J Téllez-Sosa
- Departamento de Inmunología, Instituto Nacional de Salud Pública (INSP), Centro de Investigación Sobre Enfermedades Infecciosas (CISEI), Cuernavaca, México
| | - J Martínez-Barnetche
- Departamento de Inmunología, Instituto Nacional de Salud Pública (INSP), Centro de Investigación Sobre Enfermedades Infecciosas (CISEI), Cuernavaca, México
| | - L Wood
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Y Paterson
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - L Cedillo-Barrón
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados IPN, Ciudad de México, México
| | - O López-Franco
- Centro de Estudios y Servicios en Salud. Universidad Veracruzana, Veracruz City, Veracruz, México
| | - H Vivanco-Cid
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz City, Veracruz, México
| |
Collapse
|
8
|
Meng R, Zhao Z, Guo N, Liu Z, Zhao X, Li W, Li X, Shi C, Nie D, Wang W, Liu T, Ma W, Yu L, Li J. Effect of honokiol on exotoxin proteins listeriolysin O and p60 secreted by Listeria monocytogenes. J Med Microbiol 2015; 64:1474-1480. [DOI: 10.1099/jmm.0.000183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Rizeng Meng
- Public Health College, Jilin University, 130062 Changchun, PR China
- Jilin Enrty-exit Inspection and Quarantine Bureau, Changchun, 130062, PR China
| | - Ziwen Zhao
- Public Health College, Jilin University, 130062 Changchun, PR China
- Department of Food Quality and Safety, Jilin University, 130062 Changchun, PR China
| | - Na Guo
- Public Health College, Jilin University, 130062 Changchun, PR China
- Department of Food Quality and Safety, Jilin University, 130062 Changchun, PR China
| | - Zonghui Liu
- Department of Food Quality and Safety, Jilin University, 130062 Changchun, PR China
| | - Xingchen Zhao
- Department of Food Quality and Safety, Jilin University, 130062 Changchun, PR China
| | - Wenli Li
- Department of Food Quality and Safety, Jilin University, 130062 Changchun, PR China
| | - Xiaoxu Li
- Department of Food Quality and Safety, Jilin University, 130062 Changchun, PR China
| | - Ce Shi
- Department of Food Quality and Safety, Jilin University, 130062 Changchun, PR China
| | - Dandan Nie
- Jilin Enrty-exit Inspection and Quarantine Bureau, Changchun, 130062, PR China
| | - Weilin Wang
- Jilin Enrty-exit Inspection and Quarantine Bureau, Changchun, 130062, PR China
| | - Tao Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, 130062 Changchun, PR China
| | - Wenchen Ma
- Jilin Enrty-exit Inspection and Quarantine Bureau, Changchun, 130062, PR China
| | - Lu Yu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, 130062 Changchun, PR China
- Public Health College, Jilin University, 130062 Changchun, PR China
| | - Juan Li
- Public Health College, Jilin University, 130062 Changchun, PR China
| |
Collapse
|
9
|
Rosenheinrich M, Heine W, Schmühl CM, Pisano F, Dersch P. Natural Killer Cells Mediate Protection against Yersinia pseudotuberculosis in the Mesenteric Lymph Nodes. PLoS One 2015; 10:e0136290. [PMID: 26296209 PMCID: PMC4546584 DOI: 10.1371/journal.pone.0136290] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 08/02/2015] [Indexed: 01/11/2023] Open
Abstract
Natural killer cells play a crucial role in the initial defense against bacterial pathogens. The crosstalk between host cells infected with intracellular pathogens and NK cells has been studied intensively, but not much attention has been given to characterize the role of NK cells in the response to extracellular bacterial pathogens such as yersiniae. In this study we used antibody-mediated NK cell depletion to address the importance of this immune cell type in controlling a Y. pseudotuberculosis infection. Analysis of the bacterial counts was used to follow the infection and flow cytometry was performed to characterize the composition and dynamic of immune cells. Depletion of NK cells led to higher bacterial loads within the mesenteric lymph nodes. We further show that in particular CD11b+ CD27+ NK cells which express higher levels of the activation marker CD69 increase within the mesenteric lymph nodes during a Y. pseudotuberculosis infection. Moreover, in response to the activation NK cells secrete higher levels of IFNy, which in turn triggers the production of the proinflammatory cytokine TNFα. These results suggest, that NK cells aid in the clearance of Y. pseudotuberculosis infections mainly by triggering the expression of proinflammatory cytokines manipulating the host immune response.
Collapse
MESH Headings
- Animals
- Antibodies/pharmacology
- Antigens, CD/genetics
- Antigens, CD/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/microbiology
- B-Lymphocytes/pathology
- Female
- Gene Expression
- Immunophenotyping
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/microbiology
- Killer Cells, Natural/pathology
- Lymph Nodes/immunology
- Lymph Nodes/microbiology
- Lymph Nodes/pathology
- Lymphocyte Count
- Lymphocyte Depletion
- Macrophages/immunology
- Macrophages/microbiology
- Macrophages/pathology
- Mesentery/immunology
- Mesentery/microbiology
- Mesentery/pathology
- Mice
- Mice, Inbred C57BL
- Neutrophils/immunology
- Neutrophils/microbiology
- Neutrophils/pathology
- Spleen/immunology
- Spleen/microbiology
- Spleen/pathology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/microbiology
- T-Lymphocytes, Cytotoxic/pathology
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/metabolism
- Yersinia pseudotuberculosis/immunology
- Yersinia pseudotuberculosis Infections/immunology
- Yersinia pseudotuberculosis Infections/microbiology
- Yersinia pseudotuberculosis Infections/pathology
Collapse
Affiliation(s)
- Maik Rosenheinrich
- Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Wiebke Heine
- Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carina M. Schmühl
- Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Fabio Pisano
- Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Petra Dersch
- Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
10
|
Cajnko MM, Marušić M, Kisovec M, Rojko N, Benčina M, Caserman S, Anderluh G. Listeriolysin O Affects the Permeability of Caco-2 Monolayer in a Pore-Dependent and Ca2+-Independent Manner. PLoS One 2015; 10:e0130471. [PMID: 26087154 PMCID: PMC4472510 DOI: 10.1371/journal.pone.0130471] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 05/20/2015] [Indexed: 12/20/2022] Open
Abstract
Listeria monocytogenes is a food and soil-borne pathogen that secretes a pore-forming toxin listeriolysin O (LLO) as its major virulence factor. We tested the effects of LLO on an intestinal epithelial cell line Caco-2 and compared them to an unrelated pore-forming toxin equinatoxin II (EqtII). Results showed that apical application of both toxins causes a significant drop in transepithelial electrical resistance (TEER), with higher LLO concentrations or prolonged exposure time needed to achieve the same magnitude of response than with EqtII. The drop in TEER was due to pore formation and coincided with rearrangement of claudin-1 within tight junctions and associated actin cytoskeleton; however, no significant increase in permeability to fluorescein or 3 kDa FITC-dextran was observed. Influx of calcium after pore formation affected the magnitude of the drop in TEER. Both toxins exhibit similar effects on epithelium morphology and physiology. Importantly, LLO action upon the membrane is much slower and results in compromised epithelium on a longer time scale at lower concentrations than EqtII. This could favor listerial invasion in hosts resistant to E-cadherin related infection.
Collapse
Affiliation(s)
- Miša Mojca Cajnko
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Maja Marušić
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Matic Kisovec
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Nejc Rojko
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Mojca Benčina
- Laboratory for Biotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Simon Caserman
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Gregor Anderluh
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|
11
|
Biological effects of listeriolysin O: implications for vaccination. BIOMED RESEARCH INTERNATIONAL 2015; 2015:360741. [PMID: 25874208 PMCID: PMC4385656 DOI: 10.1155/2015/360741] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 02/13/2015] [Accepted: 02/14/2015] [Indexed: 12/13/2022]
Abstract
Listeriolysin O (LLO) is a thiol-activated cholesterol-dependent pore-forming toxin and the major virulence factor of Listeria monocytogenes (LM). Extensive research in recent years has revealed that LLO exerts a wide array of biological activities, during the infection by LM or by itself as recombinant antigen. The spectrum of biological activities induced by LLO includes cytotoxicity, apoptosis induction, endoplasmic reticulum stress response, modulation of gene expression, intracellular calcium oscillations, and proinflammatory activity. In addition, LLO is a highly immunogenic toxin and the major target for innate and adaptive immune responses in different animal models and humans. Recently, the crystal structure of LLO has been published in detail. Here, we review the structure-function relationship for this fascinating microbial molecule, highlighting the potential uses of LLO in the fields of biomedicine and biotechnology, particularly in vaccination.
Collapse
|
12
|
The cholesterol-dependent cytolysins pneumolysin and streptolysin O require binding to red blood cell glycans for hemolytic activity. Proc Natl Acad Sci U S A 2014; 111:E5312-20. [PMID: 25422425 DOI: 10.1073/pnas.1412703111] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The cholesterol-dependent cytolysin (CDC) pneumolysin (Ply) is a key virulence factor of Streptococcus pneumoniae. Membrane cholesterol is required for the cytolytic activity of this toxin, but it is not clear whether cholesterol is the only cellular receptor. Analysis of Ply binding to a glycan microarray revealed that Ply has lectin activity and binds glycans, including the Lewis histo-blood group antigens. Surface plasmon resonance analysis showed that Ply has the highest affinity for the sialyl LewisX (sLeX) structure, with a K(d) of 1.88 × 10(-5) M. Ply hemolytic activity against human RBCs showed dose-dependent inhibition by sLeX. Flow cytometric analysis and Western blots showed that blocking binding of Ply to the sLeX glycolipid on RBCs prevents deposition of the toxin in the membrane. The lectin domain responsible for sLeX binding is in domain 4 of Ply, which contains candidate carbohydrate-binding sites. Mutagenesis of these predicted carbohydrate-binding residues of Ply resulted in a decrease in hemolytic activity and a reduced affinity for sLeX. This study reveals that this archetypal CDC requires interaction with the sLeX glycolipid cellular receptor as an essential step before membrane insertion. A similar analysis conducted on streptolysin O from Streptococcus pyogenes revealed that this CDC also has glycan-binding properties and that hemolytic activity against RBCs can be blocked with the glycan lacto-N-neotetraose by inhibiting binding to the cell surface. Together, these data support the emerging paradigm shift that pore-forming toxins, including CDCs, have cellular receptors other than cholesterol that define target cell tropism.
Collapse
|
13
|
Abstract
The cell membrane is crucial for protection of the cell from its environment. MACPF/CDC proteins are a large superfamily known to be essential for bacterial pathogenesis and proper functioning of the immune system. The three most studied groups of MACPF/CDC proteins are cholesterol-dependent cytolysins from bacteria, the membrane attack complex of complement and human perforin. Their primary function is to form transmembrane pores in target cell membranes. The common mechanism of action comprises water-soluble monomeric proteins binding to the host cell membrane, oligomerization, and formation of a functional pore. This causes a disturbance in gradients of ions and other molecules across the membrane and can lead to cell death. Cells react to this form of attack in a complex manner. Responses can be general, like removing the perforated part of the membrane, or more specific, in many cases depending on binding of proteins to specific receptors to trigger various signalling cascades.
Collapse
|
14
|
Köster S, Hudel M, Chakraborty T, Yildiz Ö. Crystallization and X-ray crystallographic analysis of the cholesterol-dependent cytolysin listeriolysin O from Listeria monocytogenes. Acta Crystallogr Sect F Struct Biol Cryst Commun 2013; 69:1212-5. [PMID: 24192351 PMCID: PMC3818035 DOI: 10.1107/s1744309113025761] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/17/2013] [Indexed: 01/23/2023]
Abstract
The secreted pore-forming toxin listeriolysin O (LLO) from the intracellular pathogen Listeria monocytogenes is a member of the family of cholesterol-dependent cytolysins (CDC) with broad properties in pathogenesis. Its role as a virulence factor is enigmatic: it disrupts membranes and acts as an inductor of both pro- and anti-inflammatory responses in infected cells. In addition, LLO is also a potent target for immunogenicity during infection. Natively secreted LLO from a recombinant L. innocua strain was crystallized in its water-soluble monomeric form. The crystals obtained belonged to the orthorhombic space group P2(1)2(1)2(1), with unit-cell parameters a = 26.7, b = 85.1, c = 230.0 Å, and diffracted to beyond 2.2 Å resolution. The Matthews coefficient and the solvent content were estimated to be 2.4 Å(3) Da(-1) and 49.2%, respectively. The structure with one molecule in the asymmetric unit was solved using Phaser employing the structure of the previously characterized CDC toxin perfringolysin O as a search model.
Collapse
Affiliation(s)
- Stefan Köster
- Department of Structural Biology, Max Planck Institute of Biophysics, Max-von-Laue Strasse 3, 60438 Frankfurt am Main, Germany
- Division of Infectious Diseases, Department of Medicine, New York University School of Medicine, 550 First Avenue, Smilow 907, New York, NY 10016, USA
| | - Martina Hudel
- Institute of Medical Microbiology, Justus-Liebig University Giessen, Schubertstrasse 81, 35392 Giessen, Germany
| | - Trinad Chakraborty
- Institute of Medical Microbiology, Justus-Liebig University Giessen, Schubertstrasse 81, 35392 Giessen, Germany
| | - Özkan Yildiz
- Department of Structural Biology, Max Planck Institute of Biophysics, Max-von-Laue Strasse 3, 60438 Frankfurt am Main, Germany
| |
Collapse
|
15
|
Koch I, Dach K, Heesemann J, Hoffmann R. Yersinia enterocolitica inactivates NK cells. Int J Med Microbiol 2013; 303:433-42. [PMID: 23810728 DOI: 10.1016/j.ijmm.2013.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 05/07/2013] [Accepted: 05/20/2013] [Indexed: 12/16/2022] Open
Abstract
Natural Killer (NK) cells serve as an important source of proinflammatory cytokines early during infection. Hypothesizing that Yersinia enterocolitica might interact with and inactivate NK cells, we examined NK cell-Y. enterocolitica interactions in vitro and in vivo. Y. enterocolitica adheres to NK cells in an Invasin dependent manner and inhibits NK cell cytotoxicity and IFN-γ production induced by IL-12+IL-18 or IL-12 alone. YopP, an acetyltransferase known to inhibit MAPK and NFκB signaling, suppresses IL-12 and IL-12+IL-18 mediated IFN-γ production in NK cells by inhibiting phosphorylation of Tyk2 and STAT4 in addition to MAPK. YopP inhibits induction of all genes whose expression is induced by IL-12+IL-18 in NK cells. Y. enterocolitica-mediated adherence to and inactivation of NK cells also occurs after infection in vivo. Thus, we present the first report of a bacterial pathogen inactivating NK cells, and report interaction with Tyk2-STAT4 signaling as a novel function of YopP.
Collapse
Affiliation(s)
- Isabel Koch
- Ludwig Maximilians University, Max von Pettenkofer Institut, Department of Bacteriology, 80336 Munich, Germany
| | | | | | | |
Collapse
|
16
|
Sun R, Liu Y. Listeriolysin O as a strong immunogenic molecule for the development of new anti-tumor vaccines. Hum Vaccin Immunother 2013; 9:1058-68. [PMID: 23399758 PMCID: PMC3899140 DOI: 10.4161/hv.23871] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/23/2013] [Accepted: 02/03/2013] [Indexed: 11/19/2022] Open
Abstract
The pore-forming toxin listeriolysin O (LLO), which is produced by Listeria monocytogenes, mediates bacterial phagosomal escape and facilitates bacterial multiplication during infection. This toxin has recently gained attention because of its confirmed role in the controlled and specific modulation of the immune response. Currently, cancer immunotherapies are focused on conquering the immune tolerance induced by poorly immunogenic tumor antigens and eliciting strong, lasting immunological memory. An effective way to achieve these goals is the co-administration of potent immunomodulatory adjuvant components with vaccine vectors. LLO, a toxin that belongs to the family of cholesterol-dependent cytolysins (CDCs), exhibits potent cell type-non-specific toxicity and is a source of dominant CD4(+) and CD8(+) T cell epitopes. According to recent research, in addition to its effective cytotoxicity as a cancer immunotherapeutic drug, the non-specific adjuvant property of LLO makes it promising for the development of efficacious anti-tumor vaccines.
Collapse
Affiliation(s)
- Rui Sun
- Department of Pathology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; School of Basic Medicine; Peking Union Medical College; Beijing, P.R. China
| | - Yuqin Liu
- Department of Pathology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; School of Basic Medicine; Peking Union Medical College; Beijing, P.R. China
- Cell Resource Center; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; School of Basic Medicine; Peking Union Medical College; Beijing, P.R. China
| |
Collapse
|
17
|
Lm-LLO-Based Immunotherapies and HPV-Associated Disease. JOURNAL OF ONCOLOGY 2012; 2012:542851. [PMID: 22481930 PMCID: PMC3307007 DOI: 10.1155/2012/542851] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 10/09/2011] [Indexed: 01/06/2023]
Abstract
HPV infection is a direct cause of neoplasia and malignancy. Cellular immunologic activity against cells expressing HPV E6 and E7 is sufficient to eliminate the presence of dysplastic or neoplastic tissue driven by HPV infection. Live attenuated Listeria monocytogenes- (Lm-) based immunotherapy (ADXS11-001) has been developed for the treatment of HPV-associated diseases. ADXS11-001 secretes an antigen-adjuvant fusion (Lm-LLO) protein consisting of a truncated fragment of the Lm protein listeriolysin O (LLO) fused to HPV-16 E7. In preclinical models, this construct has been found to stimulate immune responses and affect therapeutic outcome. ADXS11-001 is currently being evaluated in Phase 2 clinical trials for cervical intraepithelial neoplasia, cervical cancer, and HPV-positive head and neck cancer. The use of a live attenuated bacterium is a more complex and complete method of cancer immunotherapy, as over millennia Lm has evolved to infect humans and humans have evolved to prevent and reject this infection over millennia. This evolution has resulted in profound pathogen-associated immune mechanisms which are genetically conserved, highly efficacious, resistant to tolerance, and can be uniquely invoked using this novel platform technology.
Collapse
|
18
|
Horowitz A, Stegmann KA, Riley EM. Activation of natural killer cells during microbial infections. Front Immunol 2012; 2:88. [PMID: 22566877 PMCID: PMC3342047 DOI: 10.3389/fimmu.2011.00088] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 12/16/2011] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are large granular lymphocytes that express a diverse array of germline encoded inhibitory and activating receptors for MHC Class I and Class I-like molecules, classical co-stimulatory ligands, and cytokines. The ability of NK cells to be very rapidly activated by inflammatory cytokines, to secrete effector cytokines, and to kill infected or stressed host cells, suggests that they may be among the very early responders during infection. Recent studies have also identified a small number of pathogen-derived ligands that can bind to NK cell surface receptors and directly induce their activation. Here we review recent studies that have begun to elucidate the various pathways by which viral, bacterial, and parasite pathogens activate NK cells. We also consider two emerging themes of NK cell–pathogen interactions, namely their contribution to adaptive immune responses and their potential to take on regulatory and immunomodulatory functions.
Collapse
Affiliation(s)
- Amir Horowitz
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine London, UK
| | | | | |
Collapse
|
19
|
Jahangiri A, Rasooli I, Gargari SLM, Owlia P, Rahbar MR, Amani J, Khalili S. An in silico DNA vaccine against Listeria monocytogenes. Vaccine 2011; 29:6948-58. [PMID: 21791233 DOI: 10.1016/j.vaccine.2011.07.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Revised: 07/09/2011] [Accepted: 07/11/2011] [Indexed: 01/14/2023]
Abstract
Listeria monocytogenes causes listeriosis with mortality rate >20%. Listeriolysin-O (LLO), a pore-forming hemolysin, belongs to the family of cholesterol-dependent toxins (CDTX) and plays roles in the pathogenicity. In this study bioinformatic analyses were carried out on LLO sequence as a major immunodominant listerial antigen toward designing a DNA vaccine stimulating cytotoxic T-lymphocytes (CTLs). Mouse and human constructs were designed based on predicted T cell epitopes and MHC class I binders, which were then tandemly fused together. LLO-derived construct codons and a variety of critical gene expression efficiency parameters were optimized. Post-translational modifications such as glycosylation, phosphorylation were analysed. The constructs corresponded to LLO sequences of L. monocytogenes in BLAST search. Neither human nor mouse construct was allergen. Secretory pathway was location of the human construct that enhances immune induction and contribute to the efficacy of the vaccine candidate. mRNAs from optimized DNA sequences of both human and mouse constructs are more stable than the native and are suitable for initiation of translation. The constructs contain several sites for phosphorylation that could improve its degradation and subsequent entry into the MHC class I pathway. Addition of GPI anchor, myristoylation and ubiquitin signals or proline (P), glutamic acid (E), serine (S), threonine (T) (PEST)-like motifs at the N-terminal of constructs increase efficacy of the DNA vaccine. Close physical contact between the favorable immunogen and the suitable CpG oligodeoxynucleotides (CpG ODN) promotes immune response. Vectors for checking the expression of constructs in mammalian cells and for harboring the foreign genes as DNA vaccine are suggested.
Collapse
Affiliation(s)
- Abolfazl Jahangiri
- Department of Biology, Shahed University, Tehran-Qom Express Way, Opposite Imam Khomeini's Shrine, Tehran-3319118651, Iran
| | | | | | | | | | | | | |
Collapse
|
20
|
Stavru F, Archambaud C, Cossart P. Cell biology and immunology of Listeria monocytogenes infections: novel insights. Immunol Rev 2011; 240:160-84. [DOI: 10.1111/j.1600-065x.2010.00993.x] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
21
|
Shahabi V, Maciag PC, Rivera S, Wallecha A. Live, attenuated strains of Listeria and Salmonella as vaccine vectors in cancer treatment. Bioeng Bugs 2010; 1:235-43. [PMID: 21327055 DOI: 10.4161/bbug.1.4.11243] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Revised: 12/28/2009] [Accepted: 01/04/2010] [Indexed: 02/07/2023] Open
Abstract
Live, attenuated strains of many bacteria that synthesize and secrete foreign antigens are being developed as vaccines for a number of infectious diseases and cancer. Bacterial-based vaccines provide a number of advantages over other antigen delivery strategies including low cost of production, the absence of animal products, genetic stability and safety. In addition, bacterial vaccines delivering a tumor-associated antigen (TAA) stimulate innate immunity and also activate both arms of the adaptive immune system by which they exert efficacious anti-tumor effects. Listeria monocytogenes and several strains of Salmonella have been most extensively studied for this purpose. A number of attenuated strains have been generated and used to deliver antigens associated with infectious diseases and cancer. Although both bacteria are intracellular, the immune responses invoked by Listeria and Salmonella are different due to their sub-cellular locations. Upon entering antigen-presenting cells by phagocytosis, Listeria is capable of escaping from the phagosomal compartment and thus has direct access to the cell cytosol. Proteins delivered by this vector behave as endogenous antigens, are presented on the cell surface in the context of MHC class I molecules, and generate strong cell-mediated immune responses. In contrast, proteins delivered by Salmonella, which lacks a phagosomal escape mechanism, are treated as exogenous antigens and presented by MHC class II molecules resulting predominantly in Th2 type immune responses. This fundamental disparity between the life cycles of the two vectors accounts for their differential application as antigen delivery vehicles. The present paper includes a review of the most recent advances in the development of these two bacterial vectors for treatment of cancer. Similarities and differences between the two vectors are discussed.
Collapse
Affiliation(s)
- Vafa Shahabi
- Advaxis Inc., Research and Development, North Brunswick, NJ, USA
| | | | | | | |
Collapse
|
22
|
The natural killer cell: a further innate mediator of gouty inflammation? Immunol Cell Biol 2009; 88:24-31. [DOI: 10.1038/icb.2009.91] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
23
|
Chu J, Thomas LM, Watkins SC, Franchi L, Núñez G, Salter RD. Cholesterol-dependent cytolysins induce rapid release of mature IL-1beta from murine macrophages in a NLRP3 inflammasome and cathepsin B-dependent manner. J Leukoc Biol 2009; 86:1227-38. [PMID: 19675207 DOI: 10.1189/jlb.0309164] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
CDC are exotoxins secreted by many Gram-positive bacteria that bind cholesterol and oligomerize to form pores in eukaryotic cell membranes. We demonstrate that CDC TLO induces caspase-1 cleavage and the rapid release of IL-1beta from LPS-primed murine BMDM. IL-1beta secretion depends on functional toxin pore formation, as free cholesterol, which prevents TLO binding to cell membranes, blocks the cytokine release. Secretion of the mature forms of IL-1beta and caspase-1 occurs only at lower TLO doses, whereas at a higher concentration, cells release the biologically inactive proforms. IL-1beta release at a low TLO dose requires potassium efflux, calcium influx, and the activities of calcium-independent PLA(2), caspase-1, and cathepsin B. Additionally, mature IL-1beta release induced by a low TLO dose is dependent on the NLRP3 inflammasome, and pro-IL-1beta release induced by a high TLO dose occurs independently of NLRP3. These results further elucidate a mechanism of CDC-induced IL-1beta release and suggest a novel, immune evasion strategy in which IL-1beta-containing macrophages might release primarily inactive cytokine following exposure to high doses of these toxins.
Collapse
Affiliation(s)
- Jessica Chu
- Immunology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
24
|
Multiple mechanisms contribute to the robust rapid gamma interferon response by CD8+ T cells during Listeria monocytogenes infection. Infect Immun 2009; 77:1492-501. [PMID: 19179413 DOI: 10.1128/iai.01207-08] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A subset of CD8+ T cells can rapidly secrete gamma interferon (IFN-gamma) in an antigen-independent and interleukin-12 (IL-12)- and IL-18-dependent manner within 16 h of infection with the intracellular bacterial pathogen Listeria monocytogenes. This rapid IFN-gamma response is robust enough to be detected directly ex vivo and is not observed following infection with intracellular bacterial pathogens that remain sequestered within host cell vacuoles. We demonstrate here that three distinct pathways can lead to rapid secretion of IFN-gamma by CD8+ T cells during L. monocytogenes infection: (i) a direct cytokine-inducing activity encoded by the cholesterol-dependent cytolysin (CDC) listeriolysin O (LLO) acts within the infected cell, (ii) the pore-forming activity of LLO promotes cytosolic localization of bacterial products that trigger cytosol-specific signaling pathways, and (iii) the sustained presence of high concentrations of bacterial products can exogenously trigger cytokine production. Although it has been suggested that CDC protein toxins may act as Toll-like receptor 4 (TLR4) agonists to trigger proinflammatory cytokine secretion, we show in this report that TLR4 signaling is not required to induce a maximal rapid IFN-gamma response by CD8+ T cells. The results presented here indicate that multiple mechanisms contribute to the induction of rapid IFN-gamma secretion by CD8+ T cells during Listeria infection and that care must be taken when interpreting the results of in vitro assays, since the contribution of each pathway can vary depending on how the assay is performed.
Collapse
|
25
|
Donaldson DS, Williams NA. Bacterial toxins as immunomodulators. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 666:1-18. [PMID: 20054971 DOI: 10.1007/978-1-4419-1601-3_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bacterial toxins are the causative agent at pathology in a variety of diseases. Although not always the primary target of these toxins, many have been shown to have potent immunomodulatory effects, for example, inducing immune responses to co-administered antigens and suppressing activation of immune cells. These abilities of bacterial toxins can be harnessed and used in a therapeutic manner, such as in vaccination or the treatment of autoimmune diseases. Furthermore, the ability of toxins to gain entry to cells can be used in novel bacterial toxin based immuno-therapies in order to deliver antigens into MHC Class I processing pathways. Whether the immunomodulatory properties of these toxins arose in order to enhance bacterial survival within hosts, to aid spread within the population or is pure serendipity, it is interesting to think that these same toxins potentially hold the key to preventing or treating human disease.
Collapse
Affiliation(s)
- David S Donaldson
- Department of Cellular and Molecular Medicine, School of Medicine Sciences, University of Bristol, Bristol, UK
| | | |
Collapse
|
26
|
Wood LM, Guirnalda PD, Seavey MM, Paterson Y. Cancer immunotherapy using Listeria monocytogenes and listerial virulence factors. Immunol Res 2008; 42:233-45. [PMID: 19018479 PMCID: PMC2763613 DOI: 10.1007/s12026-008-8087-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our laboratory is interested in how immunogenicity may be modulated in vivo in order to better design more effective immunotherapeutics against cancer. Our main approach is to use a facultative intracellular bacterium, Listeria monocytogenes, which has the unusual ability to live and grow in the cytoplasm of the cell and is thus an excellent vector for targeting passenger antigens to the major histocompatibility complex (MHC) class I pathway of antigen processing with the generation of authentic CTL epitopes. We have used this approach to target tumor antigens expressed on breast, melanoma and cervical cancer. We are also exploring the role of Listerial virulence factors in potentiating adaptive immune responses by activating innate immunity. Specifically, we are using these proteins as adjuvants for B cell lymphomas.
Collapse
Affiliation(s)
- Laurence M. Wood
- Department of Microbiology, University of Pennsylvania School of Medicine, 323 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104-6076, USA
| | - Patrick D. Guirnalda
- Department of Microbiology, University of Pennsylvania School of Medicine, 323 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104-6076, USA
| | - Matthew M. Seavey
- Department of Microbiology, University of Pennsylvania School of Medicine, 323 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104-6076, USA
| | - Yvonne Paterson
- Department of Microbiology, University of Pennsylvania School of Medicine, 323 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104-6076, USA
| |
Collapse
|
27
|
Listeria monocytogenes desensitizes immune cells to subsequent Ca2+ signaling via listeriolysin O-induced depletion of intracellular Ca2+ stores. Infect Immun 2007; 76:857-62. [PMID: 18056478 DOI: 10.1128/iai.00622-07] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Listeriolysin O (LLO), the pore-forming toxin of Listeria monocytogenes, is a prototype of the cholesterol-dependent cytolysins (CDCs) secreted by several pathogenic and nonpathogenic gram-positive bacteria. In addition to mediating the escape of the bacterium into the cytosol, this toxin is generally believed to be a central player in host-pathogen interactions during L. monocytogenes infection. LLO triggers the influx of Ca(2+) into host cells as well as the release of Ca(2+) from intracellular stores. Thus, many of the cellular responses induced by LLO are related to calcium signaling. Interestingly, in this study, we report that prolonged exposure to LLO desensitizes cells to Ca(2+) mobilization upon subsequent stimulations with LLO. Cells preexposed to LLO-positive L. monocytogenes but not to the LLO-deficient Deltahly mutant were found to be highly refractory to Ca(2+) induction in response to receptor-mediated stimulation. Such cells also exhibited diminished Ca(2+) signals in response to stimulation with LLO and thapsigargin. The presented results suggest that this phenomenon is due to the depletion of intracellular Ca(2+) stores. The ability of LLO to desensitize immune cells provides a significant hint about the possible role played by CDCs in the evasion of the immune system by bacterial pathogens.
Collapse
|
28
|
Föller M, Shumilina E, Lam R, Mohamed W, Kasinathan R, Huber S, Chakraborty T, Lang F. Induction of suicidal erythrocyte death by listeriolysin from Listeria monocytogenes. Cell Physiol Biochem 2007; 20:1051-60. [PMID: 17975307 DOI: 10.1159/000110715] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Listeriolysin, the secreted cytolysin of the facultative intracellular bacterium Listeria monocytogenes, is its major virulence factor. Previously, non-lytic concentrations of listeriolysin were shown to induce Ca2+-permeable nonselective cation channels in human embryonic kidney cells. In erythrocytes, Ca2+ entry is followed by activation of K+ channels resulting in K+-exit as well as by membrane scrambling resulting in phosphatidylserine exposure at the cell surface. Phosphatidylserine-exposing erythrocytes are recognized by macrophages, engulfed, degraded and thus cleared from circulating blood. Phosphatidylserine exposure is a key event of eryptosis, the suicidal death of erythrocytes. The present study utilized patch-clamp technique, Fluo3-fluorescence, and annexin V-binding in FACS analysis to determine the effect of listeriolysin on cell membrane conductance, cytosolic free Ca2+ concentration, and phosphatidylserine exposure, respectively. Within 30 minutes, exposure of human peripheral blood erythrocytes to low concentrations of listeriolysin (which were non-hemolytic for the majority of cells) induced a Ca2+-permeable cation conductance in the erythrocyte cell membrane, increased cytosolic Ca2+ concentration, and triggered annexin V-binding. Increase of extracellular K+ concentration blunted, but did not prevent, listeriolysin-induced annexin V-binding. In conclusion, listeriolysin triggers suicidal death of erythrocytes, an effect at least partially due to depletion of intracellular K+. Listeriolysin induced suicidal erythrocyte death could well contribute to the pathophysiology of L. monocytogenes infection.
Collapse
Affiliation(s)
- Michael Föller
- Department of Physiology, Eberhard Karls University, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Newman KC, Riley EM. Whatever turns you on: accessory-cell-dependent activation of NK cells by pathogens. Nat Rev Immunol 2007; 7:279-91. [PMID: 17380157 DOI: 10.1038/nri2057] [Citation(s) in RCA: 188] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Natural killer (NK) cells have a crucial role in combating infections and cancers and their surface receptors can directly recognize and respond to damaged, transformed or non-self cells. Whereas some virus-infected cells are recognized by this same route, NK-cell responses to many pathogens are triggered by a different mechanism. Activation of NK cells by these pathogens requires the presence of accessory cells such as monocytes, macrophages and dendritic cells. Recent studies have identified numerous pathogen-recognition receptors that enable accessory cells to recognize different pathogens and subsequently transmit signals--both soluble and contact-dependent--to NK cells, which respond by upregulating their cytotoxic potential and the production of inflammatory cytokines.
Collapse
Affiliation(s)
- Kirsty C Newman
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | | |
Collapse
|
30
|
Hara H, Kawamura I, Nomura T, Tominaga T, Tsuchiya K, Mitsuyama M. Cytolysin-dependent escape of the bacterium from the phagosome is required but not sufficient for induction of the Th1 immune response against Listeria monocytogenes infection: distinct role of Listeriolysin O determined by cytolysin gene replacement. Infect Immun 2007; 75:3791-801. [PMID: 17517863 PMCID: PMC1951982 DOI: 10.1128/iai.01779-06] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Listeria monocytogenes evades the antimicrobial mechanisms of macrophages by escaping from the phagosome into the cytosolic space via a unique cytolysin that targets the phagosomal membrane, listeriolysin O (LLO), encoded by hly. Gamma interferon (IFN-gamma), which is known to play a pivotal role in the induction of Th1-dependent protective immunity in mice, appears to be produced, depending on the bacterial virulence factor. To determine whether the LLO molecule (the major virulence factor of L. monocytogenes) is indispensable or the escape of bacteria from the phagosome is sufficient to induce IFN-gamma production, we first constructed an hly-deleted mutant of L. monocytogenes and then established isogenic L. monocytogenes mutants expressing LLO or ivanolysin O (ILO), encoded by ilo from Listeria ivanovii. LLO-expressing L. monocytogenes was highly capable of inducing IFN-gamma production and Listeria-specific protective immunity, while the hly-deleted mutant was not. In contrast, the level of IFN-gamma induced by ILO-expressing L. monocytogenes was significantly lower both in vitro and in vivo, despite the ability of this strain to escape the phagosome and the intracellular multiplication at a level equivalent to that of LLO-expressing L. monocytogenes. Only a negligible level of protective immunity was induced in mice against challenge with LLO- and ILO-expressing L. monocytogenes. These results clearly show that escape of the bacterium from the phagosome is a prerequisite but is not sufficient for the IFN-gamma-dependent Th1 response against L. monocytogenes, and some distinct molecular nature of LLO is indispensable for the final induction of IFN-gamma that is essentially required to generate a Th1-dependent immune response.
Collapse
Affiliation(s)
- Hideki Hara
- Department of Microbiology, Kyoto University Graduate School of Medicine, Yoshida-konoecho, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | |
Collapse
|
31
|
Yamamoto K, Kawamura I, Tominaga T, Nomura T, Ito J, Mitsuyama M. Listeriolysin O derived from Listeria monocytogenes inhibits the effector phase of an experimental allergic rhinitis induced by ovalbumin in mice. Clin Exp Immunol 2006; 144:475-84. [PMID: 16734617 PMCID: PMC1941979 DOI: 10.1111/j.1365-2249.2006.03092.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Listeriolysin O (LLO) derived from Listeria monocytogenes is highly capable of inducing interleukin (IL)-12, IL-18 and interferon (IFN)-gamma, and facilitates the generation of Th1 cells. We have recently shown that recombinant LLO (rLLO) inhibits generation of ovalbumin (OVA)-specific Th2 immune response by skewing maturation of antigen-specific T cells into Th1 cells. In the present study, we investigated the effect of rLLO on the effector phase of Th2-dependent allergic rhinitis in BALB/c mice sensitized with OVA. In mice sensitized intraperitoneally and challenged intranasally with OVA, nasal allergic symptoms such as sneezing and nose-scratching were observed at a high frequency. A high titre of anti-OVA IgE antibody was detected in sera and a large number of eosinophils migrated into the nasal tissue. However, rLLO treatment during the intranasal challenge inhibited the allergic symptoms, production of anti-OVA IgE antibody and eosinophil infiltration. Though rLLO did not affect antigen-specific cytokine production from splenic CD4(+) T cells, rLLO significantly suppressed OVA-specific IL-4 and IL-5 production from nasal mononuclear cells. We further found that rLLO inhibited the recruitment of CD4(+) T cells in nasal mucosa, and diminished the transcription and cell surface expression of CCR4 on splenic CD4(+) T cells. Moreover, rLLO was able to inhibit the passive cutaneous anaphylaxis reaction mediated by anaphylactic antibodies (IgE and IgG(1)) and mast cells. Taken together, these data showed that rLLO suppresses the effector phase of allergic rhinitis by inhibition of Th2 cell recruitment to nasal mucosa and type I allergic reaction.
Collapse
Affiliation(s)
- K Yamamoto
- Department of Microbiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
32
|
Yamamoto K, Kawamura I, Tominaga T, Nomura T, Kohda C, Ito J, Mitsuyama M. Listeriolysin O, a cytolysin derived from Listeria monocytogenes, inhibits generation of ovalbumin-specific Th2 immune response by skewing maturation of antigen-specific T cells into Th1 cells. Clin Exp Immunol 2005; 142:268-74. [PMID: 16232213 PMCID: PMC1809525 DOI: 10.1111/j.1365-2249.2005.02922.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Listeriolysin O (LLO), a cholesterol-dependent cytolysin derived from Listeria monocytogenes, is a potent inducer of interleukin (IL)-12, IL-18 and interferon (IFN)-gamma. We have shown that LLO facilitates development of T cells mediating protective immunity against L. monocytogenes through the induction of IFN-gamma production at an early stage. Based on this finding, it is postulated that LLO inhibits differentiation of Th2 cells and the Th2 immune response. By using a murine model of ovalbumin (OVA)-induced allergic rhinitis, we investigated whether LLO has an ability to modulate the Th2-type immune disorder. In mice sensitized intraperitoneally with ovalbumin (OVA)/alum and challenged intranasally with OVA, a large number of eosinophils migrated into the nasal tissue, and high titres of anti-OVA IgE and IgG(1) antibodies were detected in sera. However, LLO treatment during sensitization markedly inhibited the eosinophil infiltration and production of these anti-OVA antibodies. A large number of T cells from mice sensitized and challenged with OVA produced high level of IL-4 and IL-5 but not IFN-gamma after stimulation with OVA. In contrast, OVA-specific IFN-gamma-producing T cells were preferentially induced in mice treated with LLO at the time of sensitization. In the absence of LLO administration, the expression level of GATA-3 and SOCS-3 in CD4(+) T cells was enhanced after sensitization with OVA. LLO treatment resulted in a reduction of GATA-3 and SOCS-3 expressions but induced the transcription of T-bet instead. Taken together, these data show clearly that LLO is capable of inhibiting Th2 immune response by skewing differentiation of antigen-specific T cells into Th1 cells.
Collapse
Affiliation(s)
- K Yamamoto
- Department of Microbiology, Kyoto University Graduate School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Jost BH, Billington SJ. Arcanobacterium pyogenes: molecular pathogenesis of an animal opportunist. Antonie van Leeuwenhoek 2005; 88:87-102. [PMID: 16096685 DOI: 10.1007/s10482-005-2316-5] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2004] [Accepted: 02/15/2005] [Indexed: 10/25/2022]
Abstract
Arcanobacterium pyogenes is a commensal and an opportunistic pathogen of economically important livestock, causing diseases as diverse as mastitis, liver abscessation and pneumonia. This organism possesses a number of virulence factors that contribute to its pathogenic potential. A. pyogenes expresses a cholesterol-dependent cytolysin, pyolysin, which is a haemolysin and is cytolytic for immune cells, including macrophages. Expression of pyolysin is required for virulence and this molecule is the most promising vaccine candidate identified to date. A. pyogenes also possesses a number of adherence mechanisms, including two neuraminidases, the action of which are required for full adhesion to epithelial cells, and several extracellular matrix-binding proteins, including a collagen-binding protein, which may be required for adhesion to collagen-rich tissue. A. pyogenes also expresses fimbriae, which are similar to the type 2 fimbriae of Actinomyces naeslundii, and forms biofilms. However, the role of these factors in the pathogenesis of A. pyogenes infections remains to be elucidated. A. pyogenes also invades and survives within epithelial cells and can survive within J774A.1 macrophages for up to 72 h, suggesting an important role for A. pyogenes interaction with host cells during pathogenesis. The two component regulatory system, PloSR, up-regulates pyolysin expression and biofilm formation but down-regulates expression of proteases, suggesting that it may act as a global regulator of A. pyogenes virulence. A. pyogenes is a versatile pathogen, with an arsenal of virulence determinants. However, most aspects of the pathogenesis of infection caused by this important opportunistic pathogen remain poorly characterized.
Collapse
Affiliation(s)
- B Helen Jost
- Department of Veterinary Science and Microbiology, University of Arizona, 1117 East Lowell Street, Tucson, AZ 85721, USA.
| | | |
Collapse
|
34
|
Ito Y, Kawamura I, Kohda C, Tsuchiya K, Nomura T, Mitsuyama M. Seeligeriolysin O, a protein toxin of Listeria seeligeri, stimulates macrophage cytokine production via Toll-like receptors in a profile different from that induced by other bacterial ligands. Int Immunol 2005; 17:1597-606. [PMID: 16291660 DOI: 10.1093/intimm/dxh341] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Seeligeriolysin O (LSO), a member of cholesterol-dependent cytolysins of Listeria seeligeri, exhibits cytokine-inducing activity. In this study, we examined the profile of cytokines expressed in macrophages of mice after stimulation with full-length form of recombinant LSO (rLSO530), C-terminal-truncated protein (rLSO483) and two authentic cytokine-inducing Toll-like receptor (TLR) ligands from bacteria, peptidoglycan (PGN) and LPS. Both rLSO530 and rLSO483 were able to induce IL-12 p40 and IL-12 p70 more strongly in macrophages than PGN or LPS. In contrast, IFN-beta and nitric oxide were induced by LPS but not by rLSO530, rLSO483 or PGN. In the presence of exogenously added IFN-beta, IL-12 p40 and IL-12 p70 production was inhibited after LSO stimulation, but IL-12 p70 production was enhanced after PGN stimulation. Although LSO signaling appeared to be associated with both TLR2 and TLR4, the profile of cytokine production by LSO stimulation was distinct from those by stimulation with PGN or LPS. Thus, it was shown that LSO is a unique bacterial ligand that induces macrophage cytokine production in a manner different from PGN or LPS.
Collapse
Affiliation(s)
- Yutaka Ito
- Department of Microbiology, Kyoto University Graduate School of Medicine, Japan.
| | | | | | | | | | | |
Collapse
|
35
|
Takeuchi O, Suzuki T, Kawamura I, Kobayashi N, Takizawa-Hashimoto A, Mitsuyama M. Involvement of the virulence gene products of Yersinia enterocolitica in the immune response of infected mice. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2005; 45:321-9. [PMID: 16019194 DOI: 10.1016/j.femsim.2005.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2005] [Revised: 05/26/2005] [Accepted: 05/27/2005] [Indexed: 11/26/2022]
Abstract
The virulence of Yersinia enterocolitica is known to be highly dependent on its virulence plasmid. However, it remains unclear whether the virulence plasmid is engaged also in the induction of cell-mediated immunity that is essential for protective immunity in the host. In this study, we have compared the induction of type 1 helper T cell immunity against Y. enterocolitica using a virulent strain (P+) harboring the pYV plasmid and an avirulent strain (P-) harboring no pYV. Spleen cells from both groups of mice immunized with 1/10 LD50 of P+ strain and those with 1/10 LD50 of P- strain produced a high level of gamma interferon (IFN-gamma) upon stimulation with heat-killed bacteria, and CD4+ T cells were exclusively responsible for IFN-gamma production. When crude Yersinia outer proteins (Yops) were used for antigenic stimulation, IFN-gamma response of immune spleen cells against crude Yops was observed only in mice immunized with P+ strain. Flowcytometric analysis revealed a significant level of increase in IFN-gamma-producing CD8+ T cells as well as the increase in IFN-gamma-producing CD4+ T cells against crude Yops. These results suggest that the virulence plasmid of Y. enterocolitica is involved in the induction of Th1-type of possibly protective T cells in infected mice.
Collapse
Affiliation(s)
- Osamu Takeuchi
- Department of Microbiology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Smith C, Cheers C. Synergism between active listeriolysin O and dimethyldioctadecylammonium bromide to activate CD8+ T cells. Vaccine 2005; 23:4481-8. [PMID: 15927321 DOI: 10.1016/j.vaccine.2005.04.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 03/05/2005] [Accepted: 04/22/2005] [Indexed: 11/16/2022]
Abstract
Purified recombinant listeriolysin O (LLO) was assessed for its ability to induce T cell responses in mice. Intraperitoneal immunisation with LLO, as a fusion with glutathione-S-transferase (GST), induced the production of LLO-specific CD8(+) T cells, but not LLO-specific CD4(+) T cells. The generation of this response could be blocked by pre-treatment with cholesterol, indicating a requirement for LLO pore formation. An increase in the LLO-specific response of both CD8(+) and CD4(+) T cells could be detected following the addition of dimethyldioctadecylammonium bromide (DDA), although the generation of this response was not dependent upon LLO pore formation, suggesting that DDA might change the presentation pathway of LLO leading to activation of the CD8(+) T cells. However, this response was dependent upon the presence of structurally intact LLO, suggesting a requirement for the innate recognition of LLO in the activation of the CD4(+) and CD8(+) T cells. Therefore, DDA and LLO can act synergistically to induce the production of a CD8(+) T cell response.
Collapse
Affiliation(s)
- Corey Smith
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, Vic. 3010, Australia.
| | | |
Collapse
|
37
|
Nöllmann M, Gilbert R, Mitchell T, Sferrazza M, Byron O. The role of cholesterol in the activity of pneumolysin, a bacterial protein toxin. Biophys J 2004; 86:3141-51. [PMID: 15111427 PMCID: PMC1304179 DOI: 10.1016/s0006-3495(04)74362-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The mechanism via which pneumolysin (PLY), a toxin and major virulence factor of the bacterium Streptococcus pneumoniae, binds to its putative receptor, cholesterol, is still poorly understood. We present results from a series of biophysical studies that shed light on the interaction of PLY with cholesterol in solution and in lipid bilayers. PLY lyses cells whose walls contain cholesterol. Using standard hemolytic assays we have demonstrated that the hemolytic activity of PLY is inhibited by cholesterol, partially by ergosterol but not by lanosterol and that the functional stoichiometry of the cholesterol-PLY complex is 1:1. Tryptophan (Trp) fluorescence data recorded during PLY-cholesterol titration studies confirm this ratio, reveal a significant blue shift in the Trp fluorescence peak with increasing cholesterol concentrations indicative of increasing nonpolarity in the Trp environment, consistent with cholesterol binding by the tryptophans, and provide a measure of the affinity of cholesterol binding: K(d) = 400 +/- 100 nM. Finally, we have performed specular neutron reflectivity studies to observe the effect of PLY upon lipid bilayer structure.
Collapse
Affiliation(s)
- Marcelo Nöllmann
- Division of Infection and Immunity, Institute of Biomedical and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | | | | | | | | |
Collapse
|
38
|
Abstract
Listeria monocytogenes is the etiological agent of listeriosis, a severe human foodborne infection characterized by gastroenteritis, meningitis, encephalitis, abortions, and perinatal infections. This gram-positive bacterium is a facultative intracellular pathogen that induces its own uptake into nonphagocytic cells and spreads from cell to cell using an actin-based motility process. This review covers both well-established and recent advances in the characterization of L. monocytogenes virulence determinants and their role in the pathophysiology of listeriosis.
Collapse
Affiliation(s)
- Olivier Dussurget
- Unité des Interactions Bactéries-Cellules, Institut Pasteur, INSERM U604, 75015 Paris, France.
| | | | | |
Collapse
|
39
|
Kimoto T, Kawamura I, Kohda C, Nomura T, Tsuchiya K, Ito Y, Watanabe I, Kaku T, Setianingrum E, Mitsuyama M. Differences in gamma interferon production induced by listeriolysin O and ivanolysin O result in different levels of protective immunity in mice infected with Listeria monocytogenes and Listeria ivanovii. Infect Immun 2003; 71:2447-54. [PMID: 12704115 PMCID: PMC153848 DOI: 10.1128/iai.71.5.2447-2454.2003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Two pathogenic species in the genus Listeria, Listeria monocytogenes and Listeria ivanovii, are characterized by the production of hemolysins belonging to cholesterol-dependent cytolysins, listeriolysin O (LLO) and ivanolysin O (ILO), respectively. LLO, produced by L. monocytogenes, is able to induce gamma interferon (IFN-gamma) production and contributes to the generation of Th1-dependent protective immunity. On the other hand, nothing is known about the role of ILO, produced by L. ivanovii, in this regard. In this study, we immunized mice with 0.1 50% lethal dose (LD(50)) of L. monocytogenes and L. ivanovii. Protective immunity against a challenge with 10 LD(50) was generated in mice infected with L. monocytogenes, whereas L. ivanovii infection did not induce protection. After immunization, the level of IFN-gamma in serum samples was increased in mice given L. monocytogenes but not in those given L. ivanovii. To determine the IFN-gamma-inducing activity of cytolysins, recombinant protein was constructed. Recombinant ILO exhibited significantly lower IFN-gamma-inducing activity than LLO. By comparing the IFN-gamma-inducing activity of a chimera incorporating LLO and ILO, it was found that domains 1 to 3 of LLO were critical for IFN-gamma-inducing activity while the counterpart in ILO was unable to induce cytokine production. These results suggested that the weak ability of ILO to induce IFN-gamma production is responsible for the failure of L. ivanovii to generate effective protective immunity.
Collapse
Affiliation(s)
- Terumi Kimoto
- Department of Microbiology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Jost BH, Trinh HT, Songer JG, Billington SJ. Immunization with genetic toxoids of the Arcanobacterium pyogenes cholesterol-dependent cytolysin, pyolysin, protects mice against infection. Infect Immun 2003; 71:2966-9. [PMID: 12704180 PMCID: PMC153263 DOI: 10.1128/iai.71.5.2966-2969.2003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pyolysin (PLO), a cholesterol-dependent cytolysin expressed by Arcanobacterium pyogenes, is an important host-protective antigen. However, this molecule is toxic and requires inactivation prior to its use as a vaccine. Three genetically toxoided, nonhemolytic PLO molecules, HIS-PLO.F(497), HIS-PLO.Delta P(499), and HIS-PLO.A(522), were found to be nontoxic, and vaccinated mice were protected from infection, indicating the potential of these toxoids as vaccines. Furthermore, in a mouse model of infection, A. pyogenes carrying the F(497) mutation was as attenuated as a PLO-deficient strain, indicating that the cytolytic activity of PLO is important in virulence.
Collapse
Affiliation(s)
- B Helen Jost
- Department of Veterinary Science and Microbiology, The University of Arizona, Tucson, Arizona 85721, USA.
| | | | | | | |
Collapse
|
41
|
Ito Y, Kawamura I, Kohda C, Baba H, Nomura T, Kimoto T, Watanabe I, Mitsuyama M. Seeligeriolysin O, a cholesterol-dependent cytolysin of Listeria seeligeri, induces gamma interferon from spleen cells of mice. Infect Immun 2003; 71:234-41. [PMID: 12496171 PMCID: PMC143279 DOI: 10.1128/iai.71.1.234-241.2003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Seeligeriolysin O (LSO), one of the cholesterol-dependent cytolysins produced by Listeria seeligeri, shows 80% homology to listeriolysin O (LLO) produced by Listeria monocytogenes at the amino acid sequence level. In addition to cytolytic activity, LLO has been shown to exhibit cytokine-inducing activity. In order to determine whether LSO is also capable of exhibiting these two different activities, we constructed a recombinant full-length LSO (rLSO530) and a noncytolytic truncated derivative with a C-terminal deletion (rLSO483) and compared these molecules with recombinant LLO. The cytolytic rLSO530 molecule could induce gamma interferon (IFN-gamma) production in spleen cells when the cytolytic activity was blocked by treatment with cholesterol. The noncytolytic truncated rLSO483 molecule also induced IFN-gamma production. Anti-LLO polyclonal antibody inhibited not only LLO-induced IFN-gamma production but also LSO-induced IFN-gamma production. Both NK cells and CD11b(+) cells were required for LSO-induced IFN-gamma production. Among the various cytokines expressed in CD11b(+) cells, interleukin-12 (IL-12) and IL-18 appeared to be essential. We concluded that LSO exhibits the same biological activity as LLO.
Collapse
Affiliation(s)
- Yutaka Ito
- Department of Microbiology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Mitsui KI, Takano K, Nakatani S, Nambu H, Shibata F, Nakagawa H. Chemokine production by rat macrophages stimulated with streptolysin O from Streptococcus pyogenes. Microbiol Immunol 2002; 46:37-45. [PMID: 11911187 DOI: 10.1111/j.1348-0421.2002.tb02674.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The contribution of streptolysin O (SLO) from Streptococcus pyogenes to neutrophil infiltration in inflammatory lesions was determined by production of cytokine-induced neutrophil chemoattractant (CINC)-1, -2 and -3, and macrophage inflammatory protein (MIP)-1alpha by rat macrophages stimulated with SLO in culture. Active SLO induced the production of CINCs and MIP-1alpha in dose- and time-dependent manners. These inductions were ascertained by chemokine mRNA expression in macrophages. Streptolysin S was without effect. The SLO-cholesterol complex induced the chemokine production in proportion to the residual hemolytic activity of the complex. In addition, the effects of SLO on the chemokine production were confirmed by the injection of active SLO into the preformed air pouch on the back of rats. The infiltration of neutrophils into the pouch fluid (exudate) increased steadily with a lag phase of about 2 hr. The major chemokine found in exudates was MIP-1alpha but not CINCs. In this study, it became clear that active SLO, but not the inactive one, contributed to the production of MIP-1alpha and CINCs in the conditioned medium and in exudates.
Collapse
Affiliation(s)
- Ken-Ichiro Mitsui
- Department of Physiological Chemistry, Faculty of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical University, Toyama, Japan
| | | | | | | | | | | |
Collapse
|
43
|
Repp H, Pamukçi Z, Koschinski A, Domann E, Darji A, Birringer J, Brockmeier D, Chakraborty T, Dreyer F. Listeriolysin of Listeria monocytogenes forms Ca2+-permeable pores leading to intracellular Ca2+ oscillations. Cell Microbiol 2002; 4:483-91. [PMID: 12174083 DOI: 10.1046/j.1462-5822.2002.00207.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Listeriolysin (LLO) is a major virulence factor of Listeria monocytogenes, a Gram-positive bacterium that can cause life-threatening diseases. Various signalling events and cellular effects, including modulation of gene expression, are triggered by LLO through unknown mechanisms. Here, we demonstrate that LLO applied extracellularly at sublytic concentrations causes long-lasting oscillations of the intracellular Ca2+ level of human embryonic kidney cells; resulting from a pulsed influx of extracellular Ca2+ through pores that are formed by LLO in the plasma membrane. Calcium influx does not require the activity of endogenous Ca2+ channels. LLO-formed pores are transient and oscillate between open and closed states. Pore formation and Ca2+ oscillations were also observed after exposure of cells to native Listeria monocytogenes. Our data identify LLO as a tool used by Listeria monocytogenes to manipulate the intracellular Ca2+ level without direct contact of the bacterium with the target cell. As Ca2+ oscillations modulate cellular signalling and gene expression, our findings provide a potential molecular basis for the broad spectrum of Ca2+-dependent cellular responses induced by LLO during Listeria infection.
Collapse
Affiliation(s)
- Holger Repp
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University of Giessen, Frankfurter Str. 107, 35392 Giessen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Almeida-Campos FR, Noronha FSM, Horta MF. The multitalented pore-forming proteins of intracellular pathogens. Microbes Infect 2002; 4:741-50. [PMID: 12067834 DOI: 10.1016/s1286-4579(02)01593-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Being an intracellular pathogen demands being able to invade a host cell, to circumvent the host immune response and to survive in the intracellular environment. Pore-forming proteins are among the innumerable tools used by intracellular microorganisms to achieve these goals. Remarkably, this seems to be a multipurpose group of proteins that can act in several ways. Making channels may signify entering into host cells, inhibiting phagocytosis, escaping phagosomes or promoting pathogen dissemination. In certain cases, pore-forming proteins are double-edged tools and may benefit the host by eliminating infected cells and/or inducing inflammation.
Collapse
Affiliation(s)
- Flávia R Almeida-Campos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | | | | |
Collapse
|
45
|
Stambas J, Pietersz G, McKenzie I, Nagabhushanam V, Cheers C. Oxidised mannan-listeriolysin O conjugates induce Th1/Th2 cytokine responses after intranasal immunisation. Vaccine 2002; 20:1877-86. [PMID: 11906778 DOI: 10.1016/s0264-410x(02)00039-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Clearance of infectious organisms does not always require polarised Th1 or Th2 responses and it may be advantageous for both Th1 and Th2 responses to be elicited for effective protection against an invading pathogen. It was the aim of this study to investigate oxidised mannan as a possible Th1/Th2 adjuvant. Oxidised mannan was conjugated to two candidate antigens and delivered intranasally to mice. Immunisation with the oxidised conjugate resulted in significant antigen specific proliferative responses, IL-2, IFN-gamma and IL-4 production when compared to unconjugated controls.
Collapse
Affiliation(s)
- John Stambas
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Vic. 3052, Australia.
| | | | | | | | | |
Collapse
|
46
|
Kohda C, Kawamura I, Baba H, Nomura T, Ito Y, Kimoto T, Watanabe I, Mitsuyama M. Dissociated linkage of cytokine-inducing activity and cytotoxicity to different domains of listeriolysin O from Listeria monocytogenes. Infect Immun 2002; 70:1334-41. [PMID: 11854218 PMCID: PMC127785 DOI: 10.1128/iai.70.3.1334-1341.2002] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Listeriolysin O (LLO), a cholesterol-binding cytolysin of Listeria monocytogenes, exhibits cytokine-inducing and cytolytic activities. Because the cytolytic activity was abolished by cholesterol treatment but the cytokine-inducing activity was not, these activities appeared to be linked to different domains of the LLO molecule. In this study, we constructed recombinant full-length LLO (rLLO529) and various truncated derivatives and examined their cytolytic, cholesterol-binding, and gamma interferon (IFN-gamma)-inducing activities. rLLO529 exhibited both IFN-gamma-inducing and cytolytic activities. Four truncated rLLOs possessing different C termini, which did not exert either cytolytic or cholesterol-binding activity, stimulated IFN-gamma production in normal spleen cells. However, a truncated rLLO corresponding to domain 4 (rLLO416-529) did not exhibit IFN-gamma-inducing activity, whereas it did bind to immobilized cholesterol. In addition, though the hemolysis induced by rLLO529 was inhibited by rLLO416-529, such inhibition was not detected upon rLLO529-induced IFN-gamma production. These data indicated that domain 4 was responsible for binding of LLO to membrane cholesterol followed by oligomerization and pore formation by the entire LLO molecule. In contrast, the other part of LLO, corresponding to domain 1-3, was essential for IFN-gamma-inducing activity. These findings implied a novel aspect of the function of LLO as a bacterial modulin.
Collapse
Affiliation(s)
- Chikara Kohda
- Department of Microbiology, Kyoto University Graduate School of Medicine, Kyoto 606-8501,USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Nomura T, Kawamura I, Tsuchiya K, Kohda C, Baba H, Ito Y, Kimoto T, Watanabe I, Mitsuyama M. Essential role of interleukin-12 (IL-12) and IL-18 for gamma interferon production induced by listeriolysin O in mouse spleen cells. Infect Immun 2002; 70:1049-55. [PMID: 11854182 PMCID: PMC127750 DOI: 10.1128/iai.70.3.1049-1055.2002] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mechanism of gamma interferon (IFN-gamma) production induced by listeriolysin O (LLO), a cytolytic virulence factor of Listeria monocytogenes, was analyzed with special reference to the involvement of macrophage-derived cytokines in spleen cells of mice. LLO purified from the culture supernatant of L. monocytogenes was capable of inducing a high level of IFN-gamma when its cytolytic activity was blocked by cholesterol treatment. The IFN-gamma-inducing ability of LLO was not dependent on possibly contaminating lipopolysaccharide. Depletion of CD11b(+) cells resulted in a profound decrease in IFN-gamma production in response to LLO stimulation. Negative selection also suggested the contribution of DX5(+) cells in IFN-gamma production. Reverse transcription-PCR revealed that expression of interleukin-12 (IL-12) p35 and p40 was induced by LLO but that the IL-18 mRNA level in the CD11b(+) fraction of spleen cells was unchanged. There was no change in the expression of the IFN-gamma-inducing cytokine genes in the CD11b(-) fraction. Neutralization of IL-12 and IL-18 in culture abolished the IFN-gamma production almost completely. Spleen cells from IL-12- or IL-18-deficient mice never produced IFN-gamma after stimulation with LLO. These results clearly indicated that LLO, a well-known virulence factor of L. monocytogenes, is capable of inducing IFN-gamma from NK cells through induction of IL-12 and IL-18 from macrophages. LLO appeared to play essential roles, not only as a bacterial virulence factor but also as a bacterial modulin in the immune response of the host.
Collapse
Affiliation(s)
- Takamasa Nomura
- Department of Microbiology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Baba H, Kawamura I, Kohda C, Nomura T, Ito Y, Kimoto T, Watanabe I, Ichiyama S, Mitsuyama M. Induction of gamma interferon and nitric oxide by truncated pneumolysin that lacks pore-forming activity. Infect Immun 2002; 70:107-13. [PMID: 11748170 PMCID: PMC127632 DOI: 10.1128/iai.70.1.107-113.2002] [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] [Received: 07/09/2001] [Revised: 09/21/2001] [Accepted: 10/11/2001] [Indexed: 11/20/2022] Open
Abstract
Pneumolysin (PLY), an important virulence factor of Streptococcus pneumoniae, is known to exert various effects on the host immune cells, including cytokine induction, in addition to its known cytolytic activity as a member of the thiol-activated cytolysins. It is of interest to determine whether cytolytic activity is involved in triggering the cytokine production. In this study, we constructed full-length recombinant PLY and noncytolytic truncated PLYs with C-terminal deletions to examine the response of spleen cells to these PLY preparations. When cytolytic activity was blocked by treatment with cholesterol, full-length PLY was capable of inducing gamma interferon (IFN-gamma) production. Truncated PLYs that originally exhibited no cytolytic activity were also active in IFN-gamma induction. Therefore, the IFN-gamma-inducing ability of PLY appeared to be independent of the cytolytic activity. Furthermore, IFN-gamma-inducing preparations were also capable of inducing nitric oxide synthase expression and nitric oxide (NO) production, and the addition of neutralizing antibody to IFN-gamma abolished the NO production. These results clearly demonstrated that PLY is capable of inducing IFN-gamma production in spleen cells by a mechanism different from pore formation and that the induced IFN-gamma stimulates NO production. These findings were discussed with reference to the contribution of PLY to the virulence of S. pneumoniae in vivo.
Collapse
Affiliation(s)
- Hisashi Baba
- Department of Microbiology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Vázquez-Boland JA, Kuhn M, Berche P, Chakraborty T, Domínguez-Bernal G, Goebel W, González-Zorn B, Wehland J, Kreft J. Listeria pathogenesis and molecular virulence determinants. Clin Microbiol Rev 2001; 14:584-640. [PMID: 11432815 PMCID: PMC88991 DOI: 10.1128/cmr.14.3.584-640.2001] [Citation(s) in RCA: 1519] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The gram-positive bacterium Listeria monocytogenes is the causative agent of listeriosis, a highly fatal opportunistic foodborne infection. Pregnant women, neonates, the elderly, and debilitated or immunocompromised patients in general are predominantly affected, although the disease can also develop in normal individuals. Clinical manifestations of invasive listeriosis are usually severe and include abortion, sepsis, and meningoencephalitis. Listeriosis can also manifest as a febrile gastroenteritis syndrome. In addition to humans, L. monocytogenes affects many vertebrate species, including birds. Listeria ivanovii, a second pathogenic species of the genus, is specific for ruminants. Our current view of the pathophysiology of listeriosis derives largely from studies with the mouse infection model. Pathogenic listeriae enter the host primarily through the intestine. The liver is thought to be their first target organ after intestinal translocation. In the liver, listeriae actively multiply until the infection is controlled by a cell-mediated immune response. This initial, subclinical step of listeriosis is thought to be common due to the frequent presence of pathogenic L. monocytogenes in food. In normal individuals, the continual exposure to listerial antigens probably contributes to the maintenance of anti-Listeria memory T cells. However, in debilitated and immunocompromised patients, the unrestricted proliferation of listeriae in the liver may result in prolonged low-level bacteremia, leading to invasion of the preferred secondary target organs (the brain and the gravid uterus) and to overt clinical disease. L. monocytogenes and L. ivanovii are facultative intracellular parasites able to survive in macrophages and to invade a variety of normally nonphagocytic cells, such as epithelial cells, hepatocytes, and endothelial cells. In all these cell types, pathogenic listeriae go through an intracellular life cycle involving early escape from the phagocytic vacuole, rapid intracytoplasmic multiplication, bacterially induced actin-based motility, and direct spread to neighboring cells, in which they reinitiate the cycle. In this way, listeriae disseminate in host tissues sheltered from the humoral arm of the immune system. Over the last 15 years, a number of virulence factors involved in key steps of this intracellular life cycle have been identified. This review describes in detail the molecular determinants of Listeria virulence and their mechanism of action and summarizes the current knowledge on the pathophysiology of listeriosis and the cell biology and host cell responses to Listeria infection. This article provides an updated perspective of the development of our understanding of Listeria pathogenesis from the first molecular genetic analyses of virulence mechanisms reported in 1985 until the start of the genomic era of Listeria research.
Collapse
Affiliation(s)
- J A Vázquez-Boland
- Grupo de Patogénesis Molecular Bacteriana, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
We previously reported that treatment of mice with a neutralizing mAb against listeriolysin O (LLO), the pore-forming toxin of Listeria monocytogenes, provided resistance to this intracellular bacterium. We evaluated whether anti-LLO mAb would affect Listeria handling by macrophages, essential cells in Listeria resistance. Macrophages infected in the presence of anti-LLO mAb showed a marked reduction in intracellular Listeria growth, with a concomitant block in LLO-dependent Listeria passage from phagosome to cytosol. Anti-LLO mAb did not opsonize Listeria but, rather, acted within macrophages to neutralize LLO. Importantly, anti-LLO mAb effects on Listeria growth were independent of Fcgamma receptor expression, IFNgamma signaling, and production of nitric oxide and superoxide. These results identify a novel mechanism for antibody control of bacteria within macrophages.
Collapse
MESH Headings
- Animals
- Antibodies, Bacterial/immunology
- Antibodies, Bacterial/pharmacology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigens, Bacterial/immunology
- Bacterial Proteins/immunology
- Bacterial Toxins
- Cells, Cultured
- Cytosol/immunology
- Female
- Heat-Shock Proteins/immunology
- Hemolysin Proteins
- Humans
- Interferon-gamma/immunology
- Intracellular Fluid/immunology
- Listeria monocytogenes/growth & development
- Listeria monocytogenes/immunology
- Macrophages, Peritoneal/cytology
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/microbiology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neutralization Tests
- Signal Transduction/immunology
Collapse
Affiliation(s)
- B T Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8118, St. Louis, MO 63110, USA
| | | |
Collapse
|