1
|
Bauer BU, Knittler MR, Andrack J, Berens C, Campe A, Christiansen B, Fasemore AM, Fischer SF, Ganter M, Körner S, Makert GR, Matthiesen S, Mertens-Scholz K, Rinkel S, Runge M, Schulze-Luehrmann J, Ulbert S, Winter F, Frangoulidis D, Lührmann A. Interdisciplinary studies on Coxiella burnetii: From molecular to cellular, to host, to one health research. Int J Med Microbiol 2023; 313:151590. [PMID: 38056089 DOI: 10.1016/j.ijmm.2023.151590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/19/2023] [Accepted: 11/21/2023] [Indexed: 12/08/2023] Open
Abstract
The Q-GAPS (Q fever GermAn interdisciplinary Program for reSearch) consortium was launched in 2017 as a German consortium of more than 20 scientists with exceptional expertise, competence, and substantial knowledge in the field of the Q fever pathogen Coxiella (C.) burnetii. C. burnetii exemplifies as a zoonotic pathogen the challenges of zoonotic disease control and prophylaxis in human, animal, and environmental settings in a One Health approach. An interdisciplinary approach to studying the pathogen is essential to address unresolved questions about the epidemiology, immunology, pathogenesis, surveillance, and control of C. burnetii. In more than five years, Q-GAPS has provided new insights into pathogenicity and interaction with host defense mechanisms. The consortium has also investigated vaccine efficacy and application in animal reservoirs and identified expanded phenotypic and genotypic characteristics of C. burnetii and their epidemiological significance. In addition, conceptual principles for controlling, surveilling, and preventing zoonotic Q fever infections were developed and prepared for specific target groups. All findings have been continuously integrated into a Web-based, interactive, freely accessible knowledge and information platform (www.q-gaps.de), which also contains Q fever guidelines to support public health institutions in controlling and preventing Q fever. In this review, we will summarize our results and show an example of how an interdisciplinary consortium provides knowledge and better tools to control a zoonotic pathogen at the national level.
Collapse
Affiliation(s)
- Benjamin U Bauer
- Clinic for Swine and Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Michael R Knittler
- Friedrich-Loeffler-Institut, Institute of Immunology, Greifswald - Insel Riems, Germany
| | - Jennifer Andrack
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Jena, Germany
| | - Christian Berens
- Friedrich-Loeffler-Institut, Institute of Molecular Pathogenesis, Jena, Germany
| | - Amely Campe
- Department of Biometry, Epidemiology and Information Processing, (IBEI), WHO Collaborating Centre for Research and Training for Health at the Human-Animal-Environment Interface, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Bahne Christiansen
- Friedrich-Loeffler-Institut, Institute of Immunology, Greifswald - Insel Riems, Germany
| | - Akinyemi M Fasemore
- Bundeswehr Institute of Microbiology, Munich, Germany; University of Würzburg, Würzburg, Germany; ZB MED - Information Centre for Life Science, Cologne, Germany
| | - Silke F Fischer
- Landesgesundheitsamt Baden-Württemberg, Ministerium für Soziales, Gesundheit und Integration, Stuttgart, Germany
| | - Martin Ganter
- Clinic for Swine and Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Sophia Körner
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Jena, Germany; Fraunhofer Institute for Cell Therapy and Immunology IZI, 04103 Leipzig, Germany
| | - Gustavo R Makert
- Fraunhofer Institute for Cell Therapy and Immunology IZI, 04103 Leipzig, Germany
| | - Svea Matthiesen
- Friedrich-Loeffler-Institut, Institute of Immunology, Greifswald - Insel Riems, Germany
| | - Katja Mertens-Scholz
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Jena, Germany
| | - Sven Rinkel
- Institut für Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Martin Runge
- Lower Saxony State Office for Consumer Protection and Food Safety (LAVES), Food and Veterinary Institute Braunschweig/Hannover, Hannover, Germany
| | - Jan Schulze-Luehrmann
- Institut für Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Sebastian Ulbert
- Fraunhofer Institute for Cell Therapy and Immunology IZI, 04103 Leipzig, Germany
| | - Fenja Winter
- Department of Biometry, Epidemiology and Information Processing, (IBEI), WHO Collaborating Centre for Research and Training for Health at the Human-Animal-Environment Interface, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Dimitrios Frangoulidis
- Bundeswehr Institute of Microbiology, Munich, Germany; Bundeswehr Medical Service Headquarters VI-2, Medical Intelligence & Information, Munich, Germany
| | - Anja Lührmann
- Institut für Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany.
| |
Collapse
|
2
|
Matthiesen S, Christiansen B, Jahnke R, Zaeck LM, Karger A, Finke S, Franzke K, Knittler MR. TGF-β/IFN-γ Antagonism in Subversion and Self-Defense of Phase II Coxiella burnetii -Infected Dendritic Cells. Infect Immun 2023; 91:e0032322. [PMID: 36688662 PMCID: PMC9933720 DOI: 10.1128/iai.00323-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/20/2022] [Indexed: 01/24/2023] Open
Abstract
Dendritic cells (DCs) belong to the first line of innate defense and come into early contact with invading pathogens, including the zoonotic bacterium Coxiella burnetii, the causative agent of Q fever. However, the pathogen-host cell interactions in C. burnetii-infected DCs, particularly the role of mechanisms of immune subversion beyond virulent phase I lipopolysaccharide (LPS), as well as the contribution of cellular self-defense strategies, are not understood. Using phase II Coxiella-infected DCs, we show that impairment of DC maturation and MHC I downregulation is caused by autocrine release and action of immunosuppressive transforming growth factor-β (TGF-β). Our study demonstrates that IFN-γ reverses TGF-β impairment of maturation/MHC I presentation in infected DCs and activates bacterial elimination, predominantly by inducing iNOS/NO. Induced NO synthesis strongly affects bacterial growth and infectivity. Moreover, our studies hint that Coxiella-infected DCs might be able to protect themselves from mitotoxic NO by switching from oxidative phosphorylation to glycolysis, thus ensuring survival in self-defense against C. burnetii. Our results provide new insights into DC subversion by Coxiella and the IFN-γ-mediated targeting of C. burnetii during early steps in the innate immune response.
Collapse
Affiliation(s)
- Svea Matthiesen
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Bahne Christiansen
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Rico Jahnke
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Luca M. Zaeck
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Axel Karger
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Kati Franzke
- Institute of Infectology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| | - Michael R. Knittler
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Isle of Riems, Germany
| |
Collapse
|
3
|
Tomaiuolo S, Jansen W, Soares Martins S, Devriendt B, Cox E, Mori M. QuilA® adjuvanted Coxevac® sustains Th1-CD8 +-type immunity and increases protection in Coxiella burnetii-challenged goats. NPJ Vaccines 2023; 8:17. [PMID: 36788233 PMCID: PMC9929268 DOI: 10.1038/s41541-023-00607-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 01/24/2023] [Indexed: 02/16/2023] Open
Abstract
Coxevac® is the EMA-approved veterinary vaccine for the protection of cattle and goats against Q fever, a zoonotic bacterial disease due to Coxiella burnetii. Since Coxevac® reduces bacterial shedding and clinical symptoms but does not prevent infection, novel, ready-to-use vaccine formulations are needed to increase its immunogenicity. Here, a goat vaccination-challenge model was used to evaluate the impact of the commercially available saponin-based QuilA® adjuvant on Coxevac® immunity. Upon challenge, the QuilA®-Coxevac® group showed a stronger immune response reflected in a higher magnitude of total IgG and an increase in circulating and splenic CD8+ T-cells compared to the Coxevac® and challenged-control groups. The QuilA®-Coxevac® group was characterized by a targeted Th1-type response (IFNγ, IP10) associated with increased transcripts of CD8+ and NK cells in spleens and γδ T cells in bronchial lymph nodes. Coxevac® vaccinated animals presented an intermediate expression of Th1-related genes, while the challenged-control group showed an immune response characterized by pro-inflammatory (IL1β, TNFα, IL12), Th2 (IL4 and IL13), Th17 (IL17A) and other immunoregulatory cytokines (IL6, IL10). An intriguing role was observed for γδ T cells, which were of TBX21- and SOX4-types in the QuilA®-Coxevac® and challenged control group, respectively. Overall, the addition of QuilA® resulted in a sustained Th1-type activation associated with an increased vaccine-induced bacterial clearance of 33.3% as compared to Coxevac® only. QuilA® could be proposed as a readily-applied veterinary solution to improve Coxevac® efficacy against C. burnetii infection in field settings.
Collapse
Affiliation(s)
- Sara Tomaiuolo
- grid.508031.fBacterial Zoonoses Unit, Veterinary Bacteriology, Infectious Diseases in Animals Scientific Directorate, Sciensano, Brussels, Belgium ,National Reference Centre for Coxiella burnetii and Bartonella, Brussels, Belgium ,grid.5342.00000 0001 2069 7798Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Wiebke Jansen
- grid.508031.fBacterial Zoonoses Unit, Veterinary Bacteriology, Infectious Diseases in Animals Scientific Directorate, Sciensano, Brussels, Belgium ,National Reference Centre for Coxiella burnetii and Bartonella, Brussels, Belgium
| | - Susana Soares Martins
- grid.508031.fBacterial Zoonoses Unit, Veterinary Bacteriology, Infectious Diseases in Animals Scientific Directorate, Sciensano, Brussels, Belgium
| | - Bert Devriendt
- grid.5342.00000 0001 2069 7798Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eric Cox
- grid.5342.00000 0001 2069 7798Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Marcella Mori
- Bacterial Zoonoses Unit, Veterinary Bacteriology, Infectious Diseases in Animals Scientific Directorate, Sciensano, Brussels, Belgium. .,National Reference Centre for Coxiella burnetii and Bartonella, Brussels, Belgium.
| |
Collapse
|
4
|
Coxiella burnetii-Infected NK Cells Release Infectious Bacteria by Degranulation. Infect Immun 2020; 88:IAI.00172-20. [PMID: 32817330 DOI: 10.1128/iai.00172-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/10/2020] [Indexed: 01/24/2023] Open
Abstract
Natural killer (NK) cells are critically involved in the early immune response against various intracellular pathogens, including Coxiella burnetii and Chlamydia psittaci Chlamydia-infected NK cells functionally mature, induce cellular immunity, and protect themselves by killing the bacteria in secreted granules. Here, we report that infected NK cells do not allow intracellular multiday growth of Coxiella, as is usually observed in other host cell types. C. burnetii-infected NK cells display maturation and gamma interferon (IFN-γ) secretion, as well as the release of Coxiella-containing lytic granules. Thus, NK cells possess a potent program to restrain and expel different types of invading bacteria via degranulation. Strikingly, though, in contrast to Chlamydia, expulsed Coxiella organisms largely retain their infectivity and, hence, escape the cell-autonomous self-defense mechanism in NK cells.
Collapse
|
5
|
Ledbetter L, Cherla R, Chambers C, Zhang Y, Zhang G. Eosinophils Affect Antibody Isotype Switching and May Partially Contribute to Early Vaccine-Induced Immunity against Coxiella burnetii. Infect Immun 2019; 87:e00376-19. [PMID: 31427447 PMCID: PMC6803328 DOI: 10.1128/iai.00376-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/09/2019] [Indexed: 01/21/2023] Open
Abstract
Coxiella burnetii is an obligate intracellular Gram-negative bacterium which causes human Q fever. An acidified citrate cysteine medium (ACCM-2) has been developed which mimics the intracellular replicative niche of C. burnetii and allows axenic growth of the bacteria. To determine if C. burnetii cultured in ACCM-2 retains immunogenicity, we compared the protective efficacies of formalin-inactivated C. burnetii Nine Mile phase I (PIV) and phase II (PIIV) vaccines derived from axenic culture 7, 14, and 28 days postvaccination. PIV conferred significant protection against virulent C. burnetii as early as 7 days postvaccination, which suggests that ACCM-2-derived PIV retains immunogenicity and protectivity. We analyzed the cellular immune response in spleens from PIV- and PIIV-vaccinated mice by flow cytometry at 7 and 14 days postvaccination and found significantly more granulocytes in PIV-vaccinated mice than in PIIV-vaccinated mice. Interestingly, we found these infiltrating granulocytes to be SSChigh CD11b+ CD125+ Siglec-F+ (where SSChigh indicates a high side scatter phenotype) eosinophils. There was no change in the number of eosinophils in PIV-vaccinated CD4-deficient mice compared to the level in controls, which suggests that eosinophil accumulation is CD4+ T cell dependent. To evaluate the importance of eosinophils in PIV-mediated protection, we vaccinated and challenged eosinophil-deficient ΔdblGATA mice. ΔdblGATA mice had significantly worse disease than their wild-type counterparts when challenged 7 days postvaccination, while no significant difference was seen at 28 days postvaccination. Nevertheless, ΔdblGATA mice had elevated serum IgM with decreased IgG1 and IgG2a whether mice were challenged at 7 or 28 days postvaccination. These results suggest that eosinophils may play a role in early vaccine protection against C. burnetii and contribute to antibody isotype switching.
Collapse
Affiliation(s)
- Lindsey Ledbetter
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Rama Cherla
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Catherine Chambers
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Yan Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Guoquan Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
6
|
El-Deeb W, Ghoneim I, Fayez M, Elsohaby I, Alhaider A, ElGioushy M. Acute phase proteins, proinflammatory cytokines and oxidative stress biomarkers in sheep, goats and she-camels with Coxiella burnetii infection-induced abortion. Comp Immunol Microbiol Infect Dis 2019; 67:101352. [PMID: 31568899 DOI: 10.1016/j.cimid.2019.101352] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/10/2019] [Accepted: 09/18/2019] [Indexed: 12/01/2022]
Abstract
Acute phase proteins (APPs) and oxidative stress are helpful markers in diagnosis of several infectious diseases. APPs, proinflammatory cytokines and oxidative stress markers were evaluated for their role in the diagnosis of naturally acquired Coxiella burnetii (Q fever) associated with abortion in sheep, goats and she-camels. Blood, aborted materials and vaginal swabs were collected from mixed herds in the Eastern Province of Saudi Arabia. Antioxidant biomarkers showed significant decline in cases of abortion compared to control animals at delivery time. The correlation between disease status and all parameters ranged from moderate to high. The APPs, cytokines and the oxidative stress marker malondialdehyde (MDA) displayed a high degree of distinction between aborted sheep and goat and normal delivered animals (AUC > 0.90). However, only MDA showed a high degree of differentiation (AUC > 0.90) between aborted she-camels and normal delivered controls. In conclusion, results from our study allow us to recommend using APPs, cytokines and oxidative stress markers as an additional tool for diagnosis of naturally occurring C. burnetii infection in sheep, goats and she-camels. However, it does not replace standard procedures for detection of C. burnetii.
Collapse
Affiliation(s)
- Wael El-Deeb
- Department of Clinical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Al-Hofuf P.O. 400, Saudi Arabia; Department of Veterinary Medicine, Infectious Diseases and Fish Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| | - Ibrahim Ghoneim
- Department of Clinical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Al-Hofuf P.O. 400, Saudi Arabia; Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Mahmoud Fayez
- Ministry of Agriculture, Al Ahsa Veterinary Diagnostic Laboratory, Saudi Arabia; Veterinary Serum and Vaccine Research Institute, Ministry of Agriculture, Cairo, Egypt
| | - Ibrahim Elsohaby
- Department of Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt; Department of Health Management, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PEI, Canada
| | - Abdulrahman Alhaider
- Department of Clinical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Al-Hofuf P.O. 400, Saudi Arabia
| | - Magdy ElGioushy
- Department of Animal Medicine, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| |
Collapse
|
7
|
Schoffelen T, Textoris J, Bleeker-Rovers CP, Ben Amara A, van der Meer JWM, Netea MG, Mege JL, van Deuren M, van de Vosse E. Intact interferon-γ response against Coxiella burnetii by peripheral blood mononuclear cells in chronic Q fever. Clin Microbiol Infect 2016; 23:209.e9-209.e15. [PMID: 27876593 DOI: 10.1016/j.cmi.2016.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Q fever is caused by Coxiella burnetii, an intracellular bacterium that infects phagocytes. The aim of the present study was to investigate whether the C. burnetii-induced IFN-γ response is defective in chronic Q fever patients. METHODS IFN-γ was measured in supernatants of C. burnetii-stimulated peripheral blood mononuclear cells (PBMCs) of 17 chronic Q fever patients and 17 healthy individuals. To assess IFN-γ responses, expression profiles of IFN-γ-induced genes in C. burnetii-stimulated PBMCs were studied in six patients and four healthy individuals. Neopterin was measured in PBMC supernatants (of eight patients and four healthy individuals) and in sera (of 21 patients and 11 healthy individuals). In a genetic association study, polymorphisms in genes involved in the Th1-cytokine response were analysed in a cohort of 139 chronic Q fever patients and a cohort of 220 control individuals with previous exposition to C. burnetii. RESULTS IFN-γ production by C. burnetii-stimulated PBMCs from chronic Q fever patients was significantly higher than in healthy controls. Many IFN-γ response genes were strongly upregulated in PBMCs of patients. Neopterin levels were significantly higher in PBMC supernatants and sera of patients. The IL12B polymorphisms rs3212227 and rs2853694 were associated with chronic Q fever. CONCLUSIONS IFN-γ production, as well as the response to IFN-γ, is intact in chronic Q fever patients, and even higher than in healthy individuals. Polymorphisms in the IL-12p40 gene are associated with chronic Q fever. Thus, a deficiency in IFN-γ responses does not explain the failure to clear the infection. The genetic data suggest, however, that the IL-12/IFN-γ pathway does play a role.
Collapse
Affiliation(s)
- T Schoffelen
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - J Textoris
- URMITE, CNRS UMR 7278, IRD 198, INSERM 1095, Aix-Marseille University, Marseille, France
| | - C P Bleeker-Rovers
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A Ben Amara
- URMITE, CNRS UMR 7278, IRD 198, INSERM 1095, Aix-Marseille University, Marseille, France
| | - J W M van der Meer
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J-L Mege
- URMITE, CNRS UMR 7278, IRD 198, INSERM 1095, Aix-Marseille University, Marseille, France
| | - M van Deuren
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - E van de Vosse
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
8
|
Murine Alveolar Macrophages Are Highly Susceptible to Replication of Coxiella burnetii Phase II In Vitro. Infect Immun 2016; 84:2439-48. [PMID: 27297388 DOI: 10.1128/iai.00411-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/07/2016] [Indexed: 12/18/2022] Open
Abstract
Coxiella burnetii is a Gram-negative bacterium that causes Q fever in humans. Q fever is an atypical pneumonia transmitted through inhalation of contaminated aerosols. In mammalian lungs, C. burnetii infects and replicates in several cell types, including alveolar macrophages (AMs). The innate immunity and signaling pathways operating during infection are still poorly understood, in part because of the lack of relevant host cell models for infection in vitro In the study described here, we investigated and characterized the infection of primary murine AMs by C. burnetii phase II in vitro Our data reveal that AMs show a pronounced M2 polarization and are highly permissive to C. burnetii multiplication in vitro Murine AMs present an increased susceptibility to infection in comparison to primary bone marrow-derived macrophages. AMs support more than 2 logs of bacterial replication during 12 days of infection in culture, similar to highly susceptible host cells, such as Vero and THP-1 cells. As a proof of principle that AMs are useful for investigation of C. burnetii replication, we performed experiments with AMs from Nos2(-/-) or Ifng(-/-) mice. In the absence of gamma interferon and nitric oxide synthase 2 (NOS2), AMs were significantly more permissive than wild-type cells. In contrast, AMs from Il4(-/-) mice were more restrictive to C. burnetii replication, supporting the importance of M2 polarization for the permissiveness of AMs to C. burnetii replication. Collectively, our data account for understanding the high susceptibility of alveolar macrophages to bacterial replication and support the use of AMs as a relevant model of C. burnetii growth in primary macrophages.
Collapse
|
9
|
Xiong X, Qi Y, Jiao J, Gong W, Duan C, Wen B. Exploratory study on Th1 epitope-induced protective immunity against Coxiella burnetii infection. PLoS One 2014; 9:e87206. [PMID: 24498044 PMCID: PMC3907486 DOI: 10.1371/journal.pone.0087206] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 12/21/2013] [Indexed: 11/18/2022] Open
Abstract
Coxiella burnetii is a Gram-negative bacterium that causes Q fever in humans. In the present study, 131 candidate peptides were selected from the major immunodominant proteins (MIPs) of C. burnetii due to their high-affinity binding capacity for the MHC class II molecule H2 I-Ab based on bioinformatic analyses. Twenty-two of the candidate peptides with distinct MIP epitopes were well recognized by the IFN-γ recall responses of CD4+ T cells from mice immunized with parental proteins in an ELISPOT assay. In addition, 7 of the 22 peptides could efficiently induce CD4+ T cells from mice immunized with C. burnetii to rapidly proliferate and significantly increase IFN-γ production. Significantly higher levels of IL-2, IL-12p70, IFN-γ, and TNF-α were also detected in serum from mice immunized with a pool of the 7 peptides. Immunization with the pool of 7 peptides, but not the individual peptides, conferred a significant protection against C. burnetii infection in mice, suggesting that these Th1 peptides could work together to efficiently activate CD4+ T cells to produce the Th1-type immune response against C. burnetii infection. These observations could contribute to the rational design of molecular vaccines for Q fever.
Collapse
Affiliation(s)
- Xiaolu Xiong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- * E-mail: (XX); (BW)
| | - Yong Qi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jun Jiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Wenping Gong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Changsong Duan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Bohai Wen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- * E-mail: (XX); (BW)
| |
Collapse
|
10
|
van Schaik EJ, Chen C, Mertens K, Weber MM, Samuel JE. Molecular pathogenesis of the obligate intracellular bacterium Coxiella burnetii. Nat Rev Microbiol 2013; 11:561-73. [PMID: 23797173 DOI: 10.1038/nrmicro3049] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The agent of Q fever, Coxiella burnetii, is an obligate intracellular bacterium that causes acute and chronic infections. The study of C. burnetii pathogenesis has benefited from two recent fundamental advances: improved genetic tools and the ability to grow the bacterium in extracellular media. In this Review, we describe how these recent advances have improved our understanding of C. burnetii invasion and host cell modulation, including the formation of replication-permissive Coxiella-containing vacuoles. Furthermore, we describe the Dot/Icm (defect in organelle trafficking/intracellular multiplication) system, which is used by C. burnetii to secrete a range of effector proteins into the host cell, and we discuss the role of these effectors in remodelling the host cell.
Collapse
Affiliation(s)
- Erin J van Schaik
- Department of Microbial and Molecular Pathogenesis, Texas A&M Health Science Center College of Medicine, Bryan, Texas 77807-3260, USA
| | | | | | | | | |
Collapse
|
11
|
Abstract
Coxiella burnetii is an obligate intracellular bacterium that causes a worldwide zoonotic disease, Q fever. Since C. burnetii infection could develop into severe chronic disease in humans, vaccination is the logical approach to prevent individuals at risk of natural and deliberate exposure. Although formalin-inactivated C. burnetii phase I vaccine (PIV) is effective in protecting vaccinated host against the infection in humans, widespread use of this vaccine is limited by its high incidence of adverse reactions, especially in individuals with prior immunity to the agent. Creation of a safe and effective vaccine to prevent Q fever remains an important goal for public health and international biosecurity. It is critical to clearly understand the mechanisms that involved in development of protective immunity against C. burnetii infection and to identify the key protective antigens for developing a safe and effective new generation vaccine against Q fever. This chapter describes new information related to the characterization of acquired immunity to C. burnetii vaccination and infection that will provide a fundamental understanding of the development of protective immunity against Q fever.
Collapse
|
12
|
Shannon JG, Heinzen RA. Adaptive immunity to the obligate intracellular pathogen Coxiella burnetii. Immunol Res 2009; 43:138-48. [PMID: 18813881 DOI: 10.1007/s12026-008-8059-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Coxiella burnetii is an obligate intracellular bacterial pathogen that causes the zoonosis Q fever. While an effective whole-cell vaccine (WCV) against Q fever exists, the vaccine has limitations in being highly reactogenic in sensitized individuals. Thus, a safe and effective vaccine based on recombinant protein antigen (Ag) is desirable. To achieve this goal, a better understanding of the host response to primary infection and the precise mechanisms involved in protective immunity to C. burnetii are needed. This review summarizes our current understanding of adaptive immunity to C. burnetii with a focus on recent developments in the field.
Collapse
Affiliation(s)
- Jeffrey G Shannon
- Coxiella Pathogenesis Section, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, NIAID, NIH, 903 S 4th Street, Hamilton, MT 59840, USA
| | | |
Collapse
|
13
|
Ochoa-Repáraz J, Sentissi J, Trunkle T, Riccardi C, Pascual DW. Attenuated Coxiella burnetii phase II causes a febrile response in gamma interferon knockout and Toll-like receptor 2 knockout mice and protects against reinfection. Infect Immun 2007; 75:5845-58. [PMID: 17893129 PMCID: PMC2168348 DOI: 10.1128/iai.00901-07] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Revised: 08/28/2007] [Accepted: 09/11/2007] [Indexed: 01/09/2023] Open
Abstract
Coxiella burnetii is a highly infectious obligate intracellular bacterium. The phase I form is responsible for Q fever, a febrile illness with flu-like symptoms that often goes undiagnosed. The attenuated C. burnetii phase II (having a truncated "O" chain of its lipopolysaccharide) does not cause disease in immunocompetent animals; however, phase II organisms remain infectious, and we questioned whether disease could be produced in immunodeficient mice. To study C. burnetii phase II infections, febrile responses in gamma interferon knockout (IFN-gamma(-/-)), BALB/c, Toll-like receptor 2 knockout (TLR2(-/-)), and C57BL/6 mice were measured using the Nine Mile phase II (NMII) strain of C. burnetii. Immunocompetent mice showed minimal febrile responses, unlike those obtained with IFN-gamma(-/-) and TLR2(-/-) mice, which showed elevated rectal temperatures that were sustained for approximately 15 days with transient increases in splenic weights. Reinfection of IFN-gamma(-/-) and TLR2(-/-) mice with C. burnetii NMII 30 days after primary infection protected mice as evident by reduced febrile responses and a lack of splenic inflammation. Although minimal detection of Coxiella in TLR2(-/-) mouse spleens was observed, greater colonization was evident in the IFN-gamma(-/-) mice. Cytokine analysis was performed on infected peritoneal macrophages isolated from these mice, and immunocompetent macrophages showed robust tumor necrosis factor alpha, IFN-gamma, and granulocyte-macrophage colony-stimulating factor (GM-CSF) but no interleukin-12 (IL-12) responses. IFN-gamma(-/-) macrophages produced elevated levels of IL-6, IL-10, and IL-12, while TLR2(-/-) macrophages produced GM-CSF, IL-12, and minimal IL-10. To distinguish immunity conferred by innate or adaptive systems, adoptive transfer studies were performed and showed that immune lymphocytes obtained from immunocompetent mice protected against a subsequent challenge with NMII, indicating that adaptive immunity mediates the observed protection. Thus, our data show that NMII is capable of eliciting disease in immunocompromised mice, which may help in evaluation of vaccine candidates as well as the study of host-pathogen interactions.
Collapse
Affiliation(s)
- Javier Ochoa-Repáraz
- Veterinary Molecular Biology, Montana State University, P.O. Box 173610, Bozeman, MT 59717-3610, USA
| | | | | | | | | |
Collapse
|
14
|
Waag DM. Coxiella burnetii: host and bacterial responses to infection. Vaccine 2007; 25:7288-95. [PMID: 17825460 DOI: 10.1016/j.vaccine.2007.08.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Revised: 07/23/2007] [Accepted: 08/02/2007] [Indexed: 10/22/2022]
Abstract
Designation as a Category B biothreat agent has propelled Coxiella burnetii from a relatively obscure, underappreciated, "niche" microorganism on the periphery of bacteriology, to one of possibly great consequence if actually used in acts of bioterrorism. Advances in the study of this microorganism proceeded slowly, primarily because of the difficulty in studying this obligate intracellular pathogen that must be manipulated under biosafety level-3 conditions. The dogged determination of past and current C. burnetii researchers and the application of modern immunological and molecular techniques have more clearly defined the host and bacterial response to infection. This review is intended to provide a basic introduction to C. burnetii and Q fever, while emphasizing immunomodulatory properties, both positive and negative, of Q fever vaccines and C. burnetii infections.
Collapse
Affiliation(s)
- David M Waag
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA.
| |
Collapse
|
15
|
Voth DE, Howe D, Heinzen RA. Coxiella burnetii inhibits apoptosis in human THP-1 cells and monkey primary alveolar macrophages. Infect Immun 2007; 75:4263-71. [PMID: 17606599 PMCID: PMC1951190 DOI: 10.1128/iai.00594-07] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Coxiella burnetii, the cause of human Q fever, is an aerosol-borne, obligate intracellular bacterium that targets host alveolar mononuclear phagocytic cells during infection. In all cell types examined, C. burnetii establishes a replicative niche in a lysosome-like parasitophorous vacuole where it carries out a lengthy infectious cycle with minimal cytopathic effects. The persistent and mild nature of C. burnetii infection in vitro suggests that the pathogen modulates apoptosis to sustain the host cell. In the current study, we examined the ability of C. burnetii to inhibit apoptotic cell death during infection of human THP-1 monocyte-derived macrophages and primary monkey alveolar macrophages. C. burnetii-infected cells demonstrated significant protection from death relative to uninfected cells following treatment with staurosporine, a potent inducer of intrinsic apoptosis. This protection correlated with reduced cleavage of caspase-9, caspase-3, and poly(ADP-ribose) polymerase (PARP), all proteolytic events that occur during apoptosis. Reduced PARP cleavage was also observed in cells treated with tumor necrosis factor alpha to induce extrinsic apoptosis. Apoptosis inhibition was a C. burnetii-driven process as infected cells treated with rifampin or chloramphenicol, inhibitors of bacterial RNA and protein synthesis, respectively, showed significantly reduced protection against staurosporine-induced apoptosis. C. burnetii infection affected the expression of multiple apoptosis-related genes and resulted in increased synthesis of the antiapoptotic proteins A1/Bfl-1 and c-IAP2. Collectively, these data suggest that C. burnetii modulates apoptotic pathways to inhibit host cell death, thus providing a stable, intracellular niche for the course of the pathogen's infectious cycle.
Collapse
Affiliation(s)
- Daniel E Voth
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, 903 S. 4th Street, Hamilton, MT 59840, USA
| | | | | |
Collapse
|
16
|
Brennan RE, Russell K, Zhang G, Samuel JE. Both inducible nitric oxide synthase and NADPH oxidase contribute to the control of virulent phase I Coxiella burnetii infections. Infect Immun 2004; 72:6666-75. [PMID: 15501800 PMCID: PMC523001 DOI: 10.1128/iai.72.11.6666-6675.2004] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Host control of Coxiella burnetii infections is believed to be mediated primarily by activated monocytes/macrophages. The activation of macrophages by cytokines leads to the production of reactive oxygen intermediates (ROI) and reactive nitrogen intermediates (RNI) that have potent antimicrobial activities. The contributions of ROI and RNI to the inhibition of C. burnetii replication were examined in vitro by the use of murine macrophage-like cell lines and primary mouse macrophages. A gamma interferon (IFN-gamma) treatment of infected cell lines and primary macrophages resulted in an increased production of nitric oxide (NO) and hydrogen peroxide (H2O2) and a significant inhibition of C. burnetii replication. The inhibition of replication was reversed in the murine cell line J774.16 upon the addition of either the inducible nitric oxide synthase (iNOS) inhibitor NG-monomethyl-L-arginine (NGMMLA) or the H2O2 scavenger catalase. IFN-gamma-treated primary macrophages from iNOS-/- and p47phox-/- mice significantly inhibited replication but were less efficient at controlling infection than IFN-gamma-treated wild-type macrophages. To investigate the contributions of ROI and RNI to resistance to infection, we performed in vivo studies, using C57BL/6 wild-type mice and knockout mice lacking iNOS or p47phox. Both iNOS-/- and p47phox-/- mice were attenuated in the ability to control C. burnetii infection compared to wild-type mice. Together, these results strongly support a role for both RNI and ROI in the host control of C. burnetii infection.
Collapse
Affiliation(s)
- Robert E Brennan
- Department of Medical Microbiology and Immunology, Texas A&M University System Health Science Center, College Station 77843-1114, USA
| | | | | | | |
Collapse
|
17
|
Abstract
Q fever is a widespread zoonosis caused by the Gram-negative bacterium Coxiella burnetii. Aborting domestic ruminants are the main sources of human infection but the reservoir of infection is extremely wide. In humans, Q fever may occur as acute pneumonia, hepatitis or flu-like illness or may take a severe chronic form, characterized by endocarditis, chronic hepatitis and chronic fatigue syndrome. In animals, the main clinical manifestation is late abortion. Infection with C. burnetii can be diagnosed using cultural, serological and genetic methods but because the organism is potentially dangerous and requires specialized skills only specialist laboratories are capable of undertaking diagnostic tests. This paper provides a brief overview of the epidemiology and pathogenesis of Q fever (coxiellosis).
Collapse
Affiliation(s)
- Zerai Woldehiwet
- Department of Veterinary Pathology, University of Liverpool, Veterinary Teaching Hospital, Leahurst, Neston, Wirral CH64 7TE, UK.
| |
Collapse
|
18
|
Abstract
The agent of acute and chronic Q fever, Coxiella burnetii, occupies a unique niche among intracellular pathogens. The mechanisms the organism employs to cause disease are unclear but involve persistence in a parasitophorous vacuole and the subsequent host response. Studies designed to model molecular mechanisms of pathogenesis have relied upon indirect evidence for testing the role of virulence factors since methods for generation of defined mutations have not been developed. Evidence suggests replication involving a developmental lifecycle is critical for intra- and extracellular survival but this cycle is incompletely defined. It has been proposed that survival in the phagolysosomal-like parasitophorous vacuole requires specific iron uptake systems, secretion of enzymes to detoxify the compartment (catalase and SOD), and down-regulation of an oxidative burst (acid phosphatase). Studies to test these potential virulence mechanisms can be accelerated with the recent development of the complete genome sequence for the prototype acute disease isolate, Nine Mile. Proteins differentially expressed during the developmental cycle can more readily be identified with MALDI-TOF description of proteomic profiles. Genes encoding secreted Cu/Zn SOD, catalase, and acid phosphatase are predicted and can be tested for function and expression. An iron regulon is predicted based upon Fur-regulated open reading frames. The specific role the iron-regulated genes play in iron acquisition can be tested. Confirmation of the iron regulon and others can be tested using microarrays based upon the genomic ORF predictions. These are examples of how we are rapidly changing the experimental approaches used to investigate C. burnetii to improve our understanding of the biology of this unusual and highly adapted organism.
Collapse
Affiliation(s)
- J E Samuel
- Department of Medical Microbiology and Immunology, Texas A&M University System Health Science Center, College Station, Texas 77843-1114, USA.
| | | | | |
Collapse
|
19
|
Howe D, Barrows LF, Lindstrom NM, Heinzen RA. Nitric oxide inhibits Coxiella burnetii replication and parasitophorous vacuole maturation. Infect Immun 2002; 70:5140-7. [PMID: 12183564 PMCID: PMC128226 DOI: 10.1128/iai.70.9.5140-5147.2002] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nitric oxide is a recognized cytotoxic effector against facultative and obligate intracellular bacteria. This study examined the effect of nitric oxide produced by inducible nitric oxide synthase (iNOS) up-regulated in response to cytokine stimulation, or by a synthetic nitric oxide donor, on replication of obligately intracellular Coxiella burnetii in murine L-929 cells. Immunoblotting and nitrite assays revealed that C. burnetii infection of L-929 cells augments expression of iNOS up-regulated in response to gamma interferon (IFN-gamma) and tumor necrosis factor alpha (TNF-alpha). Infection in the absence of cytokine stimulation did not result in demonstrable up-regulation of iNOS expression or in increased nitrite production. Nitrite production by cytokine-treated cells was significantly inhibited by the iNOS inhibitor S-methylisothiourea (SMT). Treatment of infected cells with IFN-gamma and TNF-alpha or the synthetic nitric oxide donor 2,2'-(hydroxynitrosohydrazino)bis-ethanamine (DETA/NONOate) had a bacteriostatic effect on C. burnetii replication. Inhibition of replication was reversed upon addition of SMT to the culture medium of cytokine-treated cells. Microscopic analysis of infected cells revealed that nitric oxide (either cytokine induced or donor derived) inhibited formation of the mature (large) parasitophorous vacuole that is characteristic of C. burnetii infection of host cells. Instead, exposure of infected cells to nitric oxide resulted in the formation of multiple small, acidic vacuoles usually containing one C. burnetii cell. Removal of nitrosative stress resulted in the coalescence of small vacuoles to form a large vacuole harboring multiple C. burnetii cells. These experiments demonstrate that nitric oxide reversibly inhibits replication of C. burnetii and formation of the parasitophorous vacuole.
Collapse
Affiliation(s)
- Dale Howe
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming 82071-3944, USA
| | | | | | | |
Collapse
|
20
|
Abstract
The lungs are a port of entry and primary infectious focus of Coxiella burnetii, the obligate intracellular contagium of the worldwide zoonosis Q fever. The infectious process and immune response are characterised by studies in cell culture and animal systems. Following endocytosis, replication exclusively occurs in the phagolysosome. Several potential virulence factors are described.
Collapse
Affiliation(s)
- L Norlander
- Defence Research Establishment, Division of NBC Defence, Umeå, Sweden
| |
Collapse
|
21
|
Abstract
Listeria monocytogenes is an enteroinvasive bacterial pathogen of man and animals. Listeriae have been shown capable of infecting the host by translocating from the intestinal lumen through Peyer's Patches (PP), however, results of experiments now indicate that these facultative intracellular parasites may also translocate through PP-independent routes. With regards to this, on occasion we observed that listeriae were absent from the PP of mice inoculated intragastrically with L. monocytogenes, but were present in the mesenteric lymph nodes of these same mice. These observations suggested that PP were not necessary for listerial translocation from the intestinal lumen. Two experimental approaches were used to determine whether luminal listeriae could indeed infect the host through PP-independent routes. First, since it is known that: 1) following the intragastric inoculation of L. monocytogenes, listeriae rapidly transit the length of the gastrointestinal tract and reside in the colonic lumen for up to a week, 2) the colon lacks PP, and 3) the descending colon and rectum are drained exclusively by the caudal lymph node (CLN), it was determined whether colonic listeriae could access the CLN. Inoculation of listeriae into the rectum of mice resulted in the infection of the CLN which indicated that PP were not required for listerial translocation. Second, since germfree SCID mice lack PP, it was determined whether listeriae could translocate from the intestinal lumen and infect these immunoincompetent mice. Shortly after the intragastric inoculation of L. monocytogenes into germfree SCID mice, listeriae were found in the mesenteries, livers and spleens. These results also indicate that PP are not required for listerial translocation from the intestinal lumen. One possible route of translocation from the intestinal lumen might occur by listeriae entering enterocytes. Results were obtained showing that listeriae were capable of entering cultured mouse small intestine enterocytes. Internalized listeriae were observed to multiply and spread intracellularly between enterocytes.
Collapse
Affiliation(s)
- E A Havell
- Department of Microbiology, Pathology and Parasitology, North Carolina State University, Raleigh, USA.
| | | | | |
Collapse
|
22
|
Abstract
Q fever is a zoonosis with a worldwide distribution with the exception of New Zealand. The disease is caused by Coxiella burnetii, a strictly intracellular, gram-negative bacterium. Many species of mammals, birds, and ticks are reservoirs of C. burnetii in nature. C. burnetii infection is most often latent in animals, with persistent shedding of bacteria into the environment. However, in females intermittent high-level shedding occurs at the time of parturition, with millions of bacteria being released per gram of placenta. Humans are usually infected by contaminated aerosols from domestic animals, particularly after contact with parturient females and their birth products. Although often asymptomatic, Q fever may manifest in humans as an acute disease (mainly as a self-limited febrile illness, pneumonia, or hepatitis) or as a chronic disease (mainly endocarditis), especially in patients with previous valvulopathy and to a lesser extent in immunocompromised hosts and in pregnant women. Specific diagnosis of Q fever remains based upon serology. Immunoglobulin M (IgM) and IgG antiphase II antibodies are detected 2 to 3 weeks after infection with C. burnetii, whereas the presence of IgG antiphase I C. burnetii antibodies at titers of >/=1:800 by microimmunofluorescence is indicative of chronic Q fever. The tetracyclines are still considered the mainstay of antibiotic therapy of acute Q fever, whereas antibiotic combinations administered over prolonged periods are necessary to prevent relapses in Q fever endocarditis patients. Although the protective role of Q fever vaccination with whole-cell extracts has been established, the population which should be primarily vaccinated remains to be clearly identified. Vaccination should probably be considered in the population at high risk for Q fever endocarditis.
Collapse
Affiliation(s)
- M Maurin
- Unité des Rickettsies, CNRS UPRES A 6020, Université de la Méditerranée, Faculté de Médecine, 13385 Marseilles Cedex 5, France
| | | |
Collapse
|
23
|
Lem L, Riethof DA, Scidmore-Carlson M, Griffiths GM, Hackstadt T, Brodsky FM. Enhanced Interaction of HLA-DM with HLA-DR in Enlarged Vacuoles of Hereditary and Infectious Lysosomal Diseases. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.1.523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Following biosynthesis, class II MHC molecules are transported through a lysosome-like compartment, where they acquire antigenic peptides for presentation to T cells at the cell surface. This compartment is characterized by the presence of HLA-DM, which catalyzes the peptide loading process. Here we report that the morphology and function of the class II loading compartment is affected in diseases with a phenotypic change in lysosome morphology. Swollen lysosomes are observed in cells from patients with the hereditary immunodeficiency Chediak-Higashi syndrome and in cells infected with Coxiella burnetii, the rickettsial organism that causes Q fever. In both disease states, we observed that HLA-DR and HLA-DM accumulate in enlarged intracellular compartments, which label with the lysosomal marker LAMP-1. The distribution of class I MHC molecules was not affected, localizing disease effects to the endocytic pathway. Thus, cellular mechanisms controlling lysosome biogenesis also affect formation of the class II loading compartment. Analysis of cell surface class II molecules revealed that their steady-state levels were not reduced on diseased cells. However, in both disease states, enhanced interaction between HLA-DR and HLA-DM was detected. In the Chediak-Higashi syndrome cells, this correlated with more efficient removal of the CLIP peptide. These findings suggest a mechanism for perturbation of Ag presentation by class II molecules and consequent immune deficiencies in both diseases.
Collapse
Affiliation(s)
- Lawrence Lem
- *The G. W. Hooper Foundation, Department of Microbiology and Immunology and
| | - David A. Riethof
- †Departments of Biopharmaceutical Sciences and Pharmaceutical Chemistry, University of California, San Francisco, CA 94143
| | - Marci Scidmore-Carlson
- ‡Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840; and
| | | | - Ted Hackstadt
- ‡Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840; and
| | - Frances M. Brodsky
- *The G. W. Hooper Foundation, Department of Microbiology and Immunology and
- †Departments of Biopharmaceutical Sciences and Pharmaceutical Chemistry, University of California, San Francisco, CA 94143
| |
Collapse
|
24
|
Abstract
The Q fever agent, Coxiella burnetii, thrives in the acidic environment of the phagolysosome of the host cell. How this obligate intracellular agent manages to survive within this hostile milieu is unknown; however, several of its enzymes may eliminate or prevent the formation of toxic oxygen metabolites by the host cell. Also implicated as virulence factors are its surface lipopolysaccharide and plasmids.
Collapse
Affiliation(s)
- O G Baca
- Dept of Biology, University of New Mexico, Albuquerque 87060
| | | | | |
Collapse
|
25
|
Izzo AA, Marmion BP. Variation in interferon-gamma responses to Coxiella burnetii antigens with lymphocytes from vaccinated or naturally infected subjects. Clin Exp Immunol 1993; 94:507-15. [PMID: 8252811 PMCID: PMC1534438 DOI: 10.1111/j.1365-2249.1993.tb08226.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Previous work in our laboratory has shown that lymphocytes from persons vaccinated with a formalin-inactivated Phase I Q fever vaccine (Q-Vax CSL Ltd) show a mitogenic response to Coxiella burnetii antigens. The mitogenic response is the sum of that from various subsets of CD4+, T helper cells, CD8+ T cells and probably B cells. It does not distinguish between T helper cell responses leading to formation of interferon-gamma (IFN-gamma)--a cytokine responsible for clearing intracellular infection with C. burnetii organisms--and responses of other T cell subsets which may produce disease-enhancing cytokines. The present study analyses (i) the capacity of Q-Vax to induce T cell sensitization which leads to IFN-gamma responses on antigen stimulation, and (ii) the immunomodulatory, (down-regulatory) effects of the Phase I lipopolysaccharide (LPS) of the organism, which interacts with monocyte/macrophages to limit IL-2 production and production of IFN-gamma by sensitized T lymphocytes.
Collapse
Affiliation(s)
- A A Izzo
- Department of Pathology, University of Adelaide, South Australia
| | | |
Collapse
|
26
|
Abstract
The author has reviewed different aspects of the role of immunocompetence in the development of Q fever. Coxiella burnetii lives within the phagolysosomes of infected cells. In animals, the immunosuppression caused by either cortisone or X-irradiation reactivates Q fever. In humans, cases of Q fever are reported in immunocompromised hosts suffering from leukemia, cancer, and human immunodeficiency virus infection (AIDS). Similar data are reported with strict or facultative intracellular parasites living within the phagolysosome. Sporadic publications reported the appearance of auto-antibodies during Q fever which may change the clinical picture of the disease. The pathological findings of hepatitis diagnosed during acute Q fever and those with C. burnetii chronic endocarditis are quite different and may reflect a different immunological response to C. burnetii. These facts emphasize the importance of host factors in the clinical expression and outcome of Q fever.
Collapse
Affiliation(s)
- D Raoult
- Centre National de Reference des Rickettsies, Chu La Timone, Marseille, France
| |
Collapse
|
27
|
Waag DM, Kende M, Damrow TA, Wood OL, Williams JC. Injection of inactivated phase I Coxiella burnetii increases non-specific resistance to infection and stimulates lymphokine production in mice. Ann N Y Acad Sci 1990; 590:203-14. [PMID: 1696075 DOI: 10.1111/j.1749-6632.1990.tb42221.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The encounter of phase I C. burnetii with the host results in seemingly disparate consequences. On the one hand, in vitro lymphocyte responses to mitogens and homologous recall antigen are suppressed. On the other, host resistance to a variety of infectious agents and to a tumor is increased. An explanation for this augmented immune response surely involves the ability of C. burnetii to stimulate cytokines, such as interferon and TNF, which enhance host immune function.
Collapse
Affiliation(s)
- D M Waag
- United States Army Medical Research Institute of Infectious Diseases, Department of Intracellular Pathogens, Fort Detrick, Frederick, Maryland 21701-5011
| | | | | | | | | |
Collapse
|
28
|
Russo M, Starobinas N, Ribeiro-Dos-Santos R, Minoprio P, Eisen H, Hontebeyrie-Joskowicz M. Susceptible mice present higher macrophage activation than resistant mice during infections with myotropic strains of Trypanosoma cruzi. Parasite Immunol 1989; 11:385-95. [PMID: 2528710 DOI: 10.1111/j.1365-3024.1989.tb00675.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The kinetics of macrophage activation were compared among inbred strains of mice (C3H, BALB, B6 and B10.A) that are known to differ in their relative resistance to infections with the myotropic strains (Colombian and CL) of Trypanosoma cruzi. The parameters utilized to measure macrophage activation were rapid spreading on glass surfaces, hydrogen peroxide release and tumour necrosis factor/cachectin production. Macrophages obtained from C3H (susceptible), BALB (intermediate) and B6 or B10.A (resistant) mice infected with both strains of T. cruzi began to spread rapidly at the onset of parasitaemia. Surprisingly, the amount of hydrogen peroxide released by peritoneal cells obtained from the more susceptible mouse strain (C3H) was significantly higher than in the other mouse strains. Also, only in the serum of C3H mice was tumour necrosis factor/cachectin detected. These results suggest that resistance against infections with myotropic strains of T. cruzi does not correlate with enhanced macrophage activation. It is also shown that the acquired macrophage activation is largely dependent on T-lymphocytes bearing the phenotypic marker CD4 (helper/inducer), since all parameters of macrophage activation were significantly inhibited in athymic mice or in C3H mice treated in vivo with monoclonal antibody anti-CD4+ T-cells.
Collapse
Affiliation(s)
- M Russo
- Department of Immunology, University of São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
29
|
Kogut MH, Lange C. Recombinant interferon-gamma inhibits cell invasion by Eimeria tenella. JOURNAL OF INTERFERON RESEARCH 1989; 9:67-77. [PMID: 2497199 DOI: 10.1089/jir.1989.9.67] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Treatment of primary chicken kidney (CK) cultures with supernatants from concanavalin A-treated Eimeria tenella-immune avian splenic T cells significantly inhibited the invasion of the cells by sporozoites of E. tenella. Biochemical evaluation of this T-cell factor is consistent with the idea that this anti-Eimeria activity in the avian lymphokines is due to interferon-gamma (IFN-gamma). We then studied the effect of recombinant bovine and human IFN-gamma on the invasion of sporozoites of E. tenella in cultures of bovine and human cell lines, respectively. The initial infection of either the Madin-Darby bovine kidney cell line (MDBK) or the human larynx epidermoid carcinoma cell line (HEp-2) by the obligate intracellular protozoan parasite E. tenella was inhibited following a 24-h pretreatment with recombinant bovine or human IFN-gamma, respectively. The IFN-gamma-mediated inhibition was dose-dependent for both cell lines. Incubation of sporozoites of E. tenella with IFN-gamma alone before infection had no detectable inhibitory effect on cell invasion of either cell line by the parasite. In addition, the establishment of the antiinfective cellular state by IFN-gamma required at least a 16- to 24-h preincubation for significant inhibitory effects. These results are suggestive of a direct effect of the IFN-gamma on the host cells and not on the parasite themselves. Taken together these studies provide evidence for a role for IFN-gamma as a nonspecific mediator of host protection against coccidiosis.
Collapse
Affiliation(s)
- M H Kogut
- Department of Animal Sciences, Cook College, Rutgers, University of New Jersey, New Brunswick 08903
| | | |
Collapse
|
30
|
Russo M, Starobinas N, Minoprio P, Coutinho A, Hontebeyrie-Joskowicz M. Parasitic load increases and myocardial inflammation decreases in Trypanosoma cruzi-infected mice after inactivation of helper T cells. ANNALES DE L'INSTITUT PASTEUR. IMMUNOLOGY 1988; 139:225-36. [PMID: 2901844 DOI: 10.1016/0769-2625(88)90136-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In order to characterize the role played by CD4+ T lymphocytes in the immunopathology of acute Trypanosoma cruzi infection, we compared the numbers of blood and tissue parasites and the heart inflammatory reaction in normal and anti-CD4 antibody-treated C3H mice. Treatment of mice with anti-CD4 mAb during acute infection markedly inhibited T-helper-cell-dependent activities, as measured by peritoneal macrophage activation and immunoglobulin secretion by splenic B lymphocytes. After in vivo inactivation of helper T cells, the number of blood and tissue parasites significantly increased, while the inflammatory cellular infiltrates of heart muscles diminished. Our results indicate that CD4+ T lymphocytes play a dual role in the immunopathology of acute experimental Chagas' disease.
Collapse
Affiliation(s)
- M Russo
- Departamento de Immunologia, Universidade de Sao Paulo, Brasil
| | | | | | | | | |
Collapse
|
31
|
Takikawa O, Kuroiwa T, Yamazaki F, Kido R. Mechanism of interferon-gamma action. Characterization of indoleamine 2,3-dioxygenase in cultured human cells induced by interferon-gamma and evaluation of the enzyme-mediated tryptophan degradation in its anticellular activity. J Biol Chem 1988. [DOI: 10.1016/s0021-9258(19)77982-4] [Citation(s) in RCA: 362] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
32
|
Abstract
Alveolar macrophages from BALB/c mice readily phagocytized endospores (2 to 5 micron) and arthroconidia of Coccidioides immitis in vitro. Within 24 to 30 h at 37 degrees C, the phagocytized endospores started developing into spherules, and the arthroconidia formed germ tubes and hyphae. However, these processes did not occur if the macrophages were incubated with murine recombinant gamma interferon (rIFN-gamma) during infection with C. immitis. Treatment with rIFN-gamma activated the fungicidal capabilities of the alveolar macrophages, as evidenced by the 50% reduction in the CFU which could be recovered from macrophages infected in the presence of gamma interferon compared with alveolar macrophages infected without gamma interferon (P less than 0.05). Similar results were seen with peritoneal macrophages incubated with rIFN-gamma and infected with C. immitis. As little as 10 U of rIFN-gamma per ml reduced by half the number of C. immitis CFU which could be recovered from the phagocytes 8 h after infection with arthroconidia, although interferon alone did not affect the viability of the fungi.
Collapse
Affiliation(s)
- L Beaman
- Medical Microbiology and Immunology, University of California School of Medicine, Davis 95616
| |
Collapse
|
33
|
Reyes L, Chinchilla M. Growth inhibition of Trypanosoma cruzi in cultured murine myocardial cells mediated by a specifically induced lymphokine. Infect Immun 1987; 55:1513-6. [PMID: 3106224 PMCID: PMC260544 DOI: 10.1128/iai.55.6.1513-1516.1987] [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] [Indexed: 01/04/2023] Open
Abstract
An in vitro model for studying the protection of mouse myocardial cells against Trypanosoma cruzi was developed. It was demonstrated that immune mouse lymphocytes stimulated with a specific antigen elicited a mediator which protected myocardial cells from newborn mice against T. cruzi infection. The substance responsible for this activity was sensitive to acid (pH 2) treatment.
Collapse
|
34
|
Maheshwari RK, Czarniecki CW, Dutta GP, Puri SK, Dhawan BN, Friedman RM. Recombinant human gamma interferon inhibits simian malaria. Infect Immun 1986; 53:628-30. [PMID: 3091507 PMCID: PMC260838 DOI: 10.1128/iai.53.3.628-630.1986] [Citation(s) in RCA: 64] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Prophylactic treatment with 0.1 mg of human gamma interferon per kg (body weight) per day completely suppressed experimental infection with Plasmodium cynomolgi B sporozoites in rhesus monkeys. Treatment with lower doses partially suppressed this infection. Prophylactic treatment with human gamma interferon, however, had no protective effect against trophozoite-induced infection, suggesting that the interferon effect was limited to the exoerythrocytic stage of parasitic development.
Collapse
|
35
|
Byrne GI, Lehmann LK, Landry GJ. Induction of tryptophan catabolism is the mechanism for gamma-interferon-mediated inhibition of intracellular Chlamydia psittaci replication in T24 cells. Infect Immun 1986; 53:347-51. [PMID: 3089936 PMCID: PMC260881 DOI: 10.1128/iai.53.2.347-351.1986] [Citation(s) in RCA: 295] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Human uroepithelial (T24) cells were incubated for 24 h in the presence of various concentrations of human recombinant gamma interferon (Hu-rIFN-gamma) and then infected with the 6BC strain of Chlamydia psittaci. This resulted in a reduction of intracellular chlamydial inclusion development in proportion to the concentration of Hu-rIFN-gamma present when Giemsa-stained cells were examined by light microscopy 24 h after infection. When tryptophan was added to Hu-rIFN-gamma-treated cells just after infection, reversal of the Hu-rIFN-gamma-mediated inhibition occurred in proportion to the concentration of tryptophan added. Addition of either isoleucine or lysine did not result in reversal of the antichlamydial state. Transport of L-[3H]tryptophan into acid-soluble intracellular pools was found to be greatly enhanced in Hu-rIFN-gamma-treated T24 cells compared with the rates measured for untreated cells. Transport of [3H]leucine was not increased in treated cells. Cells treated with Hu-rIFN-gamma also degraded L-[3H]tryptophan to catabolites that cochromatographed with N-formylkynurenine and kynurenine as measured by high-performance liquid chromatography. We conclude that Hu-rIFN-gamma-mediated inhibition of intracellular C. psittaci replication in T24 cells occurs by depletion of the essential amino acid tryptophan, most likely via the induction of indoleamine-2,3-dioxygenase, the initial enzyme of tryptophan catabolism.
Collapse
|
36
|
Turco J, Winkler HH. Gamma-interferon-induced inhibition of the growth of Rickettsia prowazekii in fibroblasts cannot be explained by the degradation of tryptophan or other amino acids. Infect Immun 1986; 53:38-46. [PMID: 3087883 PMCID: PMC260072 DOI: 10.1128/iai.53.1.38-46.1986] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We examined the role of amino acid deprivation in gamma-interferon-induced (IFN-gamma) suppression of the growth of Rickettsia prowazekii in mouse L929 cells and human fibroblasts by measuring the amino acid pools in untreated and IFN-gamma-treated cells. In recombinant IFN-gamma-treated cultures of human fibroblasts, tryptophan was undetectable in both the intracellular pool and the extracellular medium. In contrast, tryptophan was not depleted from the intracellular pool or the extracellular medium of L929 cells treated with recombinant IFN-gamma or crude mouse lymphokines. None of the other amino acids measured was severely depleted in IFN-gamma-treated L929 cells and human fibroblasts. Extracts prepared from IFN-gamma-treated human fibroblasts exhibited indoleamine 2,3-dioxygenase activity, converting tryptophan into products that cochromatographed with N-formylkynurenine and kynurenine; however, extracts prepared from untreated human fibroblasts, untreated L929 cells, recombinant IFN-gamma-treated L929 cells, and mouse lymphokine-treated L929 cells did not degrade tryptophan. Human HeLa cells resembled the human fibroblasts in that they degraded tryptophan after IFN-gamma treatment. Similarly, mouse 3T3-A31 cells and mouse embryo fibroblasts resembled mouse L929 cells in that they did not degrade tryptophan. Supplementation of the extracellular medium with additional tryptophan reconstituted the tryptophan pool in mock-infected and R. prowazekii-infected, X-irradiated, IFN-gamma-treated human fibroblasts to values greater than those observed in untreated control cultures. However, reconstitution of the tryptophan pool did not relieve IFN-gamma-induced inhibition of rickettsial growth. Addition of kynurenine or N-formylkynurenine to rickettsia-infected human fibroblasts at concentrations four times the usual tryptophan concentration did not inhibit growth of R. prowazekii. We conclude that neither tryptophan depletion nor depletion of the other amino acids studied explains the inhibitory effect of IFN-gamma on rickettsial growth in mouse L929 cells. In IFN-gamma-treated human fibroblasts, either tryptophan depletion is not involved in the inhibition of rickettsial growth or tryptophan depletion and some other mechanism(s) together contribute to the inhibition of rickettsial growth.
Collapse
|
37
|
Ferreira A, Schofield L, Enea V, Schellekens H, van der Meide P, Collins WE, Nussenzweig RS, Nussenzweig V. Inhibition of development of exoerythrocytic forms of malaria parasites by gamma-interferon. Science 1986; 232:881-4. [PMID: 3085218 DOI: 10.1126/science.3085218] [Citation(s) in RCA: 250] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A specific DNA probe was used to study the effect of recombinant rat, mouse, and human gamma-interferon (gamma-IFN) on the course of sporozoite-induced malaria infections. In mice and rats infected with sporozoites of Plasmodium berghei, mouse and rat gamma-IFN's strongly inhibited the development of the exoerythrocytic forms in the liver liver cells of the hosts, but not the development of the erythrocytic stages. The degree of inhibition of the exoerythrocytic forms was proportional to the dose of gamma-IFN administered, but was independent of the number of sporozoites used for challenge. A 30 percent reduction in the development of exoerythrocytic forms in rat liver was achieved when 150 units (about 15 nanograms of protein) of rat gamma-IFN were injected a few hours before sporozoite challenge; the reduction was 90 percent or more with higher doses of gamma-IFN. The effect was less pronounced if the gamma-IFN was administered 18 hours before or a few hours after challenge. Human gamma-IFN also diminished the parasitemia in chimpanzees infected with sporozoites of the human malaria parasite Plasmodium vivax. The target of gamma-IFN activity may be the infected hepatocytes themselves, as shown by in vitro experiments in which small doses of the human lymphokine inhibited the development of exoerythrocytic forms of Plasmodium berghei in a human hepatoma cell line. These results suggest that immunologically induced interferon may be involved in controlling malaria infection under natural conditions.
Collapse
|
38
|
Speer CA, Reduker DW, Burgess DE, Whitmire WM, Splitter GA. Lymphokine-induced inhibition of growth of Eimeria bovis and Eimeria papillata (Apicomplexa) in cultured bovine monocytes. Infect Immun 1985; 50:566-71. [PMID: 3876996 PMCID: PMC261993 DOI: 10.1128/iai.50.2.566-571.1985] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Sporozoites of Eimeria bovis penetrated and developed normally to first-generation meronts in bovine monocytes (BM) and Madin-Darby bovine kidney (MDBK) cells that had been pretreated with culture medium (CM) or supernatant (NS) from nonstimulated bovine T cells. At 240 h after sporozoite inoculation (ASI), the mean percent development (meronts/[sporozoites + meronts]) in CM- and NS-pretreated BM was 52 and 28%, respectively; values for MDBK cells were 36 and 35%, respectively. Pretreatment of BM and MDBK cells with supernatant (ConAS) from concanavalin A-stimulated bovine T cells had no effect on the ability of sporozoites to penetrate cells; however, at 240 h ASI, only 1% of the sporozoites in ConAS-pretreated BM cultures had developed to meronts. In contrast, ConAS had no adverse effect on the ability of E. bovis sporozoites to develop to first-generation meronts in MDBK cells. At 240 h ASI, E. bovis meronts in ConAS-pretreated BM were abnormal in appearance and retarded in development, whereas sporozoites appeared structurally normal by light microscopy. Pretreatment of BM with ConAS had no effect on the ability of sporozoites of Eimeria papillata (Apicomplexa) to penetrate cells. Sporozoites of E. papillata did not develop to meronts in ConAS-pretreated BM and, in contrast to E. bovis, most sporozoites were destroyed intracellularly.
Collapse
|
39
|
|
40
|
Baron S, Weigent D, Stanton GJ, Peterson J. The protective role of endogenous interferon in viral, bacterial, and protozoal infections. Antiviral Res 1985; Suppl 1:173-83. [PMID: 3936412 DOI: 10.1016/s0166-3542(85)80026-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|