1
|
Fratzke AP, Szule JA, Butler SM, van Schaik EJ, Samuel JE. Molecular mechanisms of Coxiella burnetii formalin-fixed cellular vaccine reactogenicity. Infect Immun 2024; 92:e0033524. [PMID: 39356158 PMCID: PMC11556133 DOI: 10.1128/iai.00335-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/06/2024] [Indexed: 10/03/2024] Open
Abstract
Local and systemic reactogenic responses to Q-VAX have prevented licensing of this vaccine outside of Australia. These reactogenic responses occur in previously sensitized individuals and have not been well defined at the cellular level, in part because many studies have been done in guinea pigs that have limited molecular tools. We previously characterized a mouse model of reactogenicity where local reaction sites showed an influx of CD8+ and IFNγ-expressing IL17a+ CD4+ T cells consistent with a Th1 delayed-type hypersensitivity. In this study, we determined, using depletion and adoptive transfer experiments, that both anti-Coxiella antibodies and CD4+ T cells were essential for localized reactions at the site of vaccination. Furthermore, IFNγ depletion showed significant histological changes at the local reaction sites demonstrating the essential nature of this cytokine to reactogenicity. In addition to the cells and cytokines required for this response, we determined that whole cell vaccine (WCV) material remained at the site of vaccination for at least 26 weeks post-injection. Transmission electron microscopy (TEM) of these sites demonstrated intact rod-shaped bacteria at 2 weeks post-injection and partially degraded bacteria within macrophages at 26 weeks post-injection. Finally, because small cell variants (SCVs) are an environmentally stable form, we determined that local reactions were more severe when the WCV material was prepared with higher levels of SCVs compared to typical WCV or with higher levels of large cell variant (LCV). These studies support the hypothesis that antigen persistence at the site of injection contributes to this reactogenicity and that anti-Coxiella antibodies, CD4+ T cells, and IFNγ each contribute to this process.
Collapse
Affiliation(s)
- A. P. Fratzke
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, Texas, USA
| | - J. A. Szule
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - S. M. Butler
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, Texas, USA
| | - E. J. van Schaik
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, Texas, USA
| | - J. E. Samuel
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, Texas, USA
| |
Collapse
|
2
|
Marena Guzman R, Voth DE. Embracing multiple infection models to tackle Q fever: A review of in vitro, in vivo, and lung ex vivo models. Cell Immunol 2024; 405-406:104880. [PMID: 39357100 DOI: 10.1016/j.cellimm.2024.104880] [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: 05/08/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Multiple animal and cell culture models are employed to study pathogenesis of Coxiella burnetii, the causative agent of acute and chronic human Q fever. C. burnetii is a lung pathogen that is aerosolized in contaminated products and inhaled by humans to cause acute disease that can disseminate to other organs and establish chronic infection. Cellular models of Q fever include a variety of tissue-derived cell lines from mice and humans such as lung alveolar ex vivo cells. These models have the advantage of being cost-effective and reproducible. Similarly, animal models including mice and guinea pigs are cost-effective, although only immunocompromised SCID mice display a severe disease phenotype in response to Nine Mile I and Nine Mile II isolates of C. burnetii while immunocompetent guinea pigs display human-like symptoms and robust immune responses. Non-human primates such as macaques and marmosets are the closest model of human disease but are costly and largely used for adaptive immune response studies. All animal models are used for vaccine development but many differences exist in the pathogen's ability to establish lung infection when considering infection routes, bacterial isolates, and host genetic background. Similarly, while cellular models are useful for characterization of host-pathogen mechanisms, future developments should include use of a lung infection platform to draw appropriate conclusions. Here, we summarize the current state of the C. burnetii lung pathogenesis field by discussing the contribution of different animal and cell culture models and include suggestions for continuing to move the field forward.
Collapse
Affiliation(s)
- R Marena Guzman
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Daniel E Voth
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| |
Collapse
|
3
|
Fratzke AP, Szule JA, Butler SM, van Schaik EJ, Samuel JE. Molecular Mechanisms of Coxiella burnetii Formalin Fixed Cellular Vaccine Reactogenicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608821. [PMID: 39229146 PMCID: PMC11370449 DOI: 10.1101/2024.08.20.608821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Local and systemic reactogenic responses to Q-VAX® have prevented licensing of this vaccine outside of Australia. These reactogenic responses occur in previously sensitize individuals and have not been well defined at the cellular level, in part because many studies have been done in guinea pigs that have limited molecular tools. We previously characterized a mouse model of reactogenicity where local reactions sites showed an influx of CD8+ and IFNγ-expressing IL17a+ CD4+ T cells consistent with a Th1 delayed-type hypersensitivity. In this study we determined using depletion and adoptive transfer experiments that both anti- Coxiella antibodies and CD4+ T cells were essential for localized reactions at the site of vaccination. Furthermore, IFNγ depletion showed significant histological changes at the local reaction sites demonstrating the essential nature of this cytokine to reactogenicity. In addition to the cells and cytokines required for this response, we determined WCV material remained at the site of vaccination for at least 26 weeks post-injection. Transmission electron microscopy of these sites demonstrated intact rod-shaped bacteria at 2 weeks post-injection and partially degraded bacteria within macrophages at 26 weeks post-injection. Finally, since SCVs are an environmentally stable form, we determined that local reactions were more severe when the WCV material was prepared with higher levels of SCVs compared to typical WCV or with higher levels of LCV. These studies support the hypothesis that antigen persistence at the site of injection contributes to this reactogenicity and that anti- Coxiella antibodies, CD4+ T cells, and IFNγ each contribute to this process.
Collapse
|
4
|
Mertens-Scholz K, Moawad AA, Liebler-Tenorio EM, Helming A, Andrack J, Miethe P, Neubauer H, Pletz MW, Richter IG. Ultraviolet C inactivation of Coxiella burnetii for production of a structurally preserved whole cell vaccine antigen. BMC Microbiol 2024; 24:118. [PMID: 38575865 PMCID: PMC10993581 DOI: 10.1186/s12866-024-03246-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/03/2024] [Indexed: 04/06/2024] Open
Abstract
Q fever, a worldwide-occurring zoonotic disease, can cause economic losses for public and veterinary health systems. Vaccines are not yet available worldwide and currently under development. In this regard, it is important to produce a whole cell antigen, with preserved structural and antigenic properties and free of chemical modifications. Thus, inactivation of Coxiella burnetii with ultraviolet light C (UVC) was evaluated. C. burnetii Nine Mile phase I (NMI) and phase II (NMII) were exposed to decreasing intensities in a time-dependent manner and viability was tested by rescue cultivation in axenic medium or cell culture. Effects on the cell structure were visualized by transmission electron microscopy and antigenicity of UVC-treated NMI was studied by immunization of rabbits. NMI and NMII were inactivated at UVC intensities of 250 µW/cm2 for 5 min or 100 µW/cm2 for 20 min. Reactivation by DNA repair was considered to be unlikely. No morphological changes were observed directly after UVC inactivation by transmission electron microscopy, but severe swelling and membrane degradation of bacteria with increasing severity occurred after 24 and 48 h. Immunization of rabbits resulted in a pronounced antibody response. UVC inactivation of C. burnetii resulted in a structural preserved, safe whole cell antigen and might be useful as antigen for diagnostic purposes or as vaccine candidate.
Collapse
Affiliation(s)
- Katja Mertens-Scholz
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Jena, Germany.
- Institute for Infectious Diseases and Infection Control and Center for Sepsis Care and Control (CSCC), Jena University Hospital, Jena, Germany.
| | - Amira A Moawad
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Jena, Germany
| | | | - Andrea Helming
- Department of In Vitro Diagnostics Development, Research Centre of Medical Technology and Biotechnology, Erfurt, Germany
| | - Jennifer Andrack
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Jena, Germany
| | - Peter Miethe
- Research Centre of Medical Technology and Biotechnology, Bad Langensalza, Germany
| | - Heinrich Neubauer
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Jena, Germany
| | - Mathias W Pletz
- Institute for Infectious Diseases and Infection Control and Center for Sepsis Care and Control (CSCC), Jena University Hospital, Jena, Germany
| | - Ina-Gabriele Richter
- Research Centre of Medical Technology and Biotechnology, Bad Langensalza, Germany
| |
Collapse
|
5
|
Long CM, Beare PA, Cockrell D, Binette P, Tesfamariam M, Richards C, Anderson M, McCormick-Ell J, Brose M, Anderson R, Omsland A, Pearson T, Heinzen RA. Natural reversion promotes LPS elongation in an attenuated Coxiella burnetii strain. Nat Commun 2024; 15:697. [PMID: 38267444 PMCID: PMC10808227 DOI: 10.1038/s41467-023-43972-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/24/2023] [Indexed: 01/26/2024] Open
Abstract
Lipopolysaccharide (LPS) phase variation is a critical aspect of virulence in many Gram-negative bacteria. It is of particular importance to Coxiella burnetii, the biothreat pathogen that causes Q fever, as in vitro propagation of this organism leads to LPS truncation, which is associated with an attenuated and exempted from select agent status (Nine Mile II, NMII). Here, we demonstrate that NMII was recovered from the spleens of infected guinea pigs. Moreover, these strains exhibit a previously unrecognized form of elongated LPS and display increased virulence in comparison with the initial NMII strain. The reversion of a 3-bp mutation in the gene cbu0533 directly leads to LPS elongation. To address potential safety concerns, we introduce a modified NMII strain unable to produce elongated LPS.
Collapse
Affiliation(s)
- Carrie M Long
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT, 59840, USA.
| | - Paul A Beare
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Diane Cockrell
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Picabo Binette
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Mahelat Tesfamariam
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Crystal Richards
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Matthew Anderson
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Jessica McCormick-Ell
- Office of the Director, Office of Research Services, Division of Occupational Health and Safety, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Megan Brose
- Office of the Director, Office of Research Services, Division of Occupational Health and Safety, National Institutes of Health, Hamilton, 59840, USA
| | - Rebecca Anderson
- Office of the Director, Office of Research Services, Division of Occupational Health and Safety, National Institutes of Health, Hamilton, 59840, USA
| | - Anders Omsland
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA
| | - Talima Pearson
- Department of Biological Sciences, Center for Microbial Genetics and Genomics, Northern Arizona University, Flagstaff, AZ, 86011, USA
| | - Robert A Heinzen
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT, 59840, USA
| |
Collapse
|
6
|
Bartoš O, Klimešová B, Volfová K, Chmel M, Dresler J, Pajer P, Kabíčková H, Adamík P, Modrý D, Fučíková AM, Votýpka J. Two novel Bartonella (sub)species isolated from edible dormice ( Glis glis): hints of cultivation stress-induced genomic changes. Front Microbiol 2023; 14:1289671. [PMID: 38033559 PMCID: PMC10684924 DOI: 10.3389/fmicb.2023.1289671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Bartonelloses are neglected emerging infectious diseases caused by facultatively intracellular bacteria transmitted between vertebrate hosts by various arthropod vectors. The highest diversity of Bartonella species has been identified in rodents. Within this study we focused on the edible dormouse (Glis glis), a rodent with unique life-history traits that often enters households and whose possible role in the epidemiology of Bartonella infections had been previously unknown. We identified and cultivated two distinct Bartonella sub(species) significantly diverging from previously described species, which were characterized using growth characteristics, biochemical tests, and various molecular techniques including also proteomics. Two novel (sub)species were described: Bartonella grahamii subsp. shimonis subsp. nov. and Bartonella gliris sp. nov. We sequenced two individual strains per each described (sub)species. During exploratory genomic analyses comparing two genotypes ultimately belonging to the same species, both factually and most importantly even spatiotemporally, we noticed unexpectedly significant structural variation between them. We found that most of the detected structural variants could be explained either by prophage excision or integration. Based on a detailed study of one such event, we argue that prophage deletion represents the most probable explanation of the observed phenomena. Moreover, in one strain of Bartonella grahamii subsp. shimonis subsp. nov. we identified a deletion related to Bartonella Adhesin A, a major pathogenicity factor that modulates bacteria-host interactions. Altogether, our results suggest that even a limited number of passages induced sufficient selective pressure to promote significant changes at the level of the genome.
Collapse
Affiliation(s)
- Oldřich Bartoš
- Military Health Institute, Military Medical Agency, Prague, Czechia
| | - Běla Klimešová
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Karolina Volfová
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Martin Chmel
- Military Health Institute, Military Medical Agency, Prague, Czechia
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| | - Jiří Dresler
- Military Health Institute, Military Medical Agency, Prague, Czechia
| | - Petr Pajer
- Military Health Institute, Military Medical Agency, Prague, Czechia
| | - Hana Kabíčková
- Military Health Institute, Military Medical Agency, Prague, Czechia
| | - Peter Adamík
- Department of Zoology, Faculty of Science, Palacký University, Olomouc, Czechia
- Museum of Natural History, Olomouc, Czechia
| | - David Modrý
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czechia
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno, Czechia
- Department of Veterinary Sciences/CINeZ, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| | | | - Jan Votýpka
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czechia
| |
Collapse
|
7
|
Anderson MW, Binette P, Richards C, Beare PA, Heinzen RA, Long CM. A simple method for enrichment of phase I Coxiella burnetii. J Microbiol Methods 2023; 211:106787. [PMID: 37453478 PMCID: PMC10529119 DOI: 10.1016/j.mimet.2023.106787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/16/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Coxiella burnetii is the bacterial causative agent of the zoonosis Q fever. This bacterium undergoes lipopolysaccharide (LPS) phase transition similar to Enterobacteriaciae upon in vitro passage. Full-length, phase I C. burnetii LPS is a critical virulence factor and profoundly impacts vaccine-induced immunogenicity; thus, LPS phase is an important consideration in C. burnetii experimentation and Q fever vaccine design. Typically, phase I LPS-expressing organisms are obtained from the tissues of infected experimental animals. In this process, residual phase II LPS-expressing organisms are thought to be cleared by the host immune system. Here, we propose an efficient and non-animal-based method for the enrichment of C. burnetii phase I LPS-expressing bacteria in vitro. We utilize both Vero cell culture to selectively enrich solutions with phase I and intermediate phase LPS-expressing bacteria. This simple and quick method decreases reliance on experimental animals and is a sustainable solution for Q fever diagnostic and vaccine development hurdles.
Collapse
Affiliation(s)
- Matthew W Anderson
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Picabo Binette
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Crystal Richards
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Paul A Beare
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Robert A Heinzen
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Carrie M Long
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA.
| |
Collapse
|
8
|
Shepherd DC, Kaplan M, Vankadari N, Kim KW, Larson CL, Dutka P, Beare PA, Krzymowski E, Heinzen RA, Jensen GJ, Ghosal D. Morphological remodeling of Coxiella burnetii during its biphasic developmental cycle revealed by cryo-electron tomography. iScience 2023; 26:107210. [PMID: 37485371 PMCID: PMC10362272 DOI: 10.1016/j.isci.2023.107210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/05/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
Coxiella burnetii is an obligate zoonotic bacterium that targets macrophages causing a disease called Q fever. It has a biphasic developmental life cycle where the extracellular and metabolically inactive small cell variant (SCV) transforms inside the host into the vegetative large cell variant (LCV). However, details about the morphological and structural changes of this transition are still lacking. Here, we used cryo-electron tomography to image both SCV and LCV variants grown either under axenic conditions or purified directly from host cells. We show that SCVs are characterized by equidistant stacks of inner membrane that presumably facilitate the transition to LCV, a transition coupled with the expression of the Dot/Icm type IVB secretion system (T4BSS). A class of T4BSS particles were associated with extracellular densities possibly involved in host infection. Also, SCVs contained spherical multilayered membrane structures of different sizes and locations suggesting no connection to sporulation as once assumed.
Collapse
Affiliation(s)
- Doulin C. Shepherd
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Mohammed Kaplan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Naveen Vankadari
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Ki Woo Kim
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- School of Ecology and Environmental System, Kyungpook National University, Sangju, Korea
| | - Charles L. Larson
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Przemysław Dutka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Division od Chemistry and Chemical Engineering, California Institute of Technology, 1200 California Boulevard, Pasadena, CA 91125, USA
| | - Paul A. Beare
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Edward Krzymowski
- Department of Physics and Astronomy, Brigham Young University, Provo, UT 84604, USA
| | - Robert A. Heinzen
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Grant J. Jensen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84604, USA
| | - Debnath Ghosal
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
Osman IO, Caputo A, Pinault L, Mege JL, Levasseur A, Devaux CA. Identification and Characterization of an HtrA Sheddase Produced by Coxiella burnetii. Int J Mol Sci 2023; 24:10904. [PMID: 37446087 PMCID: PMC10342153 DOI: 10.3390/ijms241310904] [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: 05/12/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Having previously shown that soluble E-cadherin (sE-cad) is found in sera of Q fever patients and that infection of BeWo cells by C. burnetii leads to modulation of the E-cad/β-cat pathway, our purpose was to identify which sheddase(s) might catalyze the cleavage of E-cad. Here, we searched for a direct mechanism of cleavage initiated by the bacterium itself, assuming the possible synthesis of a sheddase encoded in the genome of C. burnetii or an indirect mechanism based on the activation of a human sheddase. Using a straightforward bioinformatics approach to scan the complete genomes of four laboratory strains of C. burnetii, we demonstrate that C. burnetii encodes a 451 amino acid sheddase (CbHtrA) belonging to the HtrA family that is differently expressed according to the bacterial virulence. An artificial CbHtrA gene (CoxbHtrA) was expressed, and the CoxbHtrA recombinant protein was found to have sheddase activity. We also found evidence that the C. burnetii infection triggers an over-induction of the human HuHtrA gene expression. Finally, we demonstrate that cleavage of E-cad by CoxbHtrA on macrophages-THP-1 cells leads to an M2 polarization of the target cells and the induction of their secretion of IL-10, which "disarms" the target cells and improves C. burnetii replication. Taken together, these results demonstrate that the genome of C. burnetii encodes a functional HtrA sheddase and establishes a link between the HtrA sheddase-induced cleavage of E-cad, the M2 polarization of the target cells and their secretion of IL-10, and the intracellular replication of C. burnetii.
Collapse
Affiliation(s)
- Ikram Omar Osman
- Microbes Evolution Phylogeny and Infection (MEPHI) Laboratory, Aix-Marseille University, Institut de Recherche Pour le Développement (IRD), Assistance Publique Hôpitaux de Marseille (APHM), Institut Hospitalo-Universitaire (IHU)–Méditerranée Infection, 13005 Marseille, France; (I.O.O.)
| | - Aurelia Caputo
- Microbes Evolution Phylogeny and Infection (MEPHI) Laboratory, Aix-Marseille University, Institut de Recherche Pour le Développement (IRD), Assistance Publique Hôpitaux de Marseille (APHM), Institut Hospitalo-Universitaire (IHU)–Méditerranée Infection, 13005 Marseille, France; (I.O.O.)
| | - Lucile Pinault
- Microbes Evolution Phylogeny and Infection (MEPHI) Laboratory, Aix-Marseille University, Institut de Recherche Pour le Développement (IRD), Assistance Publique Hôpitaux de Marseille (APHM), Institut Hospitalo-Universitaire (IHU)–Méditerranée Infection, 13005 Marseille, France; (I.O.O.)
| | - Jean-Louis Mege
- Microbes Evolution Phylogeny and Infection (MEPHI) Laboratory, Aix-Marseille University, Institut de Recherche Pour le Développement (IRD), Assistance Publique Hôpitaux de Marseille (APHM), Institut Hospitalo-Universitaire (IHU)–Méditerranée Infection, 13005 Marseille, France; (I.O.O.)
- Laboratory of Immunology, Assitance Publique-Hôpitaux de Marseille (APHM), 13005 Marseille, France
| | - Anthony Levasseur
- Microbes Evolution Phylogeny and Infection (MEPHI) Laboratory, Aix-Marseille University, Institut de Recherche Pour le Développement (IRD), Assistance Publique Hôpitaux de Marseille (APHM), Institut Hospitalo-Universitaire (IHU)–Méditerranée Infection, 13005 Marseille, France; (I.O.O.)
| | - Christian A. Devaux
- Microbes Evolution Phylogeny and Infection (MEPHI) Laboratory, Aix-Marseille University, Institut de Recherche Pour le Développement (IRD), Assistance Publique Hôpitaux de Marseille (APHM), Institut Hospitalo-Universitaire (IHU)–Méditerranée Infection, 13005 Marseille, France; (I.O.O.)
- Centre National de la Recherche Scientifique (CNRS), 13009 Marseille, France
| |
Collapse
|
10
|
Omar Osman I, Mezouar S, Brahim-Belhaouari D, Mege JL, Devaux CA. Modulation of the E-cadherin in human cells infected in vitro with Coxiella burnetii. PLoS One 2023; 18:e0285577. [PMID: 37285354 PMCID: PMC10246793 DOI: 10.1371/journal.pone.0285577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/25/2023] [Indexed: 06/09/2023] Open
Abstract
High concentration of soluble E-cadherin (E-cad) was previously found in sera from Q fever patients. Here, BeWo cells which express a high concentration of E-cad were used as an in vitro model to investigate the expression and function of E-cad in response to infection by Coxiella burnetii, the etiological agent of Q fever. Infection of BeWo cells with C. burnetii leads to a decrease in the number of BeWo cells expressing E-cad at their membrane. A shedding of soluble E-cad was associated with the post-infection decrease of membrane-bound E-cad. The modulation of E-cad expression requires bacterial viability and was not found with heat-inactivated C. burnetii. Moreover, the intracytoplasmic cell concentration of β-catenin (β-cat), a ligand of E-cad, was reduced after bacterial infection, suggesting that the bacterium induces modulation of the E-cad/β-cat signaling pathway and CDH1 and CTNNB1 genes transcription. Finally, several genes operating the canonical Wnt-Frizzled/β-cat pathway were overexpressed in cells infected with C. burnetii. This was particularly evident with the highly virulent strain of C. burnetii, Guiana. Our data demonstrate that infection of BeWo cells by live C. burnetii modulates the E-cad/β-cat signaling pathway.
Collapse
Affiliation(s)
- Ikram Omar Osman
- Aix-Marseille Univ, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille Univ, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| | - Djamal Brahim-Belhaouari
- Aix-Marseille Univ, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| | - Jean-Louis Mege
- Aix-Marseille Univ, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| | - Christian Albert Devaux
- Aix-Marseille Univ, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
- CNRS, Marseille, France
| |
Collapse
|
11
|
Asai M, Li Y, Newton SM, Robertson BD, Langford PR. Galleria mellonella-intracellular bacteria pathogen infection models: the ins and outs. FEMS Microbiol Rev 2023; 47:fuad011. [PMID: 36906279 PMCID: PMC10045907 DOI: 10.1093/femsre/fuad011] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/13/2023] Open
Abstract
Galleria mellonella (greater wax moth) larvae are used widely as surrogate infectious disease models, due to ease of use and the presence of an innate immune system functionally similar to that of vertebrates. Here, we review G. mellonella-human intracellular bacteria pathogen infection models from the genera Burkholderia, Coxiella, Francisella, Listeria, and Mycobacterium. For all genera, G. mellonella use has increased understanding of host-bacterial interactive biology, particularly through studies comparing the virulence of closely related species and/or wild-type versus mutant pairs. In many cases, virulence in G. mellonella mirrors that found in mammalian infection models, although it is unclear whether the pathogenic mechanisms are the same. The use of G. mellonella larvae has speeded up in vivo efficacy and toxicity testing of novel antimicrobials to treat infections caused by intracellular bacteria: an area that will expand since the FDA no longer requires animal testing for licensure. Further use of G. mellonella-intracellular bacteria infection models will be driven by advances in G. mellonella genetics, imaging, metabolomics, proteomics, and transcriptomic methodologies, alongside the development and accessibility of reagents to quantify immune markers, all of which will be underpinned by a fully annotated genome.
Collapse
Affiliation(s)
- Masanori Asai
- Section of Paediatric Infectious Disease, Department of Infectious Disease, St Mary’s campus, Imperial College London, London W2 1PG, United Kingdom
| | - Yanwen Li
- Section of Paediatric Infectious Disease, Department of Infectious Disease, St Mary’s campus, Imperial College London, London W2 1PG, United Kingdom
| | - Sandra M Newton
- Section of Paediatric Infectious Disease, Department of Infectious Disease, St Mary’s campus, Imperial College London, London W2 1PG, United Kingdom
| | - Brian D Robertson
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, South Kensington campus, Imperial College London, London SW7 2AZ, United Kingdom
| | - Paul R Langford
- Section of Paediatric Infectious Disease, Department of Infectious Disease, St Mary’s campus, Imperial College London, London W2 1PG, United Kingdom
| |
Collapse
|
12
|
Williams-Macdonald SE, Mitchell M, Frew D, Palarea-Albaladejo J, Ewing D, Golde WT, Longbottom D, Nisbet AJ, Livingstone M, Hamilton CM, Fitzgerald SF, Buus S, Bach E, Dinkla A, Roest HJ, Koets AP, McNeilly TN. Efficacy of Phase I and Phase II Coxiella burnetii Bacterin Vaccines in a Pregnant Ewe Challenge Model. Vaccines (Basel) 2023; 11:vaccines11030511. [PMID: 36992095 DOI: 10.3390/vaccines11030511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
The bacterium Coxiella burnetii can cause the disease Q-fever in a wide range of animal hosts. Ruminants, including sheep, are thought to play a pivotal role in the transmission of C. burnetii to humans; however, the only existing livestock vaccine, namely, Coxevac® (Ceva Animal Health Ltd., Libourne, France), a killed bacterin vaccine based on phase I C. burnetii strain Nine-Mile, is only approved for use in goats and cattle. In this study, a pregnant ewe challenge model was used to determine the protective effects of Coxevac® and an experimental bacterin vaccine based on phase II C. burnetii against C. burnetii challenge. Prior to mating, ewes (n = 20 per group) were vaccinated subcutaneously with either Coxevac®, the phase II vaccine, or were unvaccinated. A subset of pregnant ewes (n = 6) from each group was then challenged 151 days later (~100 days of gestation) with 106 infectious mouse doses of C. burnetii, Nine-Mile strain RSA493. Both vaccines provided protection against C. burnetii challenge as measured by reductions in bacterial shedding in faeces, milk and vaginal mucus, and reduced abnormal pregnancies, compared to unvaccinated controls. This work highlights that the phase I vaccine Coxevac® can protect ewes against C. burnetii infection. Furthermore, the phase II vaccine provided comparable levels of protection and may offer a safer and cost-effective alternative to the currently licensed vaccine.
Collapse
Affiliation(s)
| | - Mairi Mitchell
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK
| | - David Frew
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK
| | - Javier Palarea-Albaladejo
- Biomathematics and Statistics Scotland, JCMB, The King's Buildings, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK
| | - David Ewing
- Biomathematics and Statistics Scotland, JCMB, The King's Buildings, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK
| | - William T Golde
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK
| | - David Longbottom
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK
| | - Alasdair J Nisbet
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK
| | - Morag Livingstone
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK
| | - Clare M Hamilton
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK
| | - Stephen F Fitzgerald
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK
| | - Søren Buus
- Department of Immunology & Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK 2200 Copenhagen, Denmark
| | - Emil Bach
- Department of Immunology & Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK 2200 Copenhagen, Denmark
| | - Annemieke Dinkla
- Department of Bacteriology, Host-Pathogen Interaction and Diagnostics, Wageningen Bioveterinary Research, Houtribweg 39, 8221 RA Lelystad, The Netherlands
| | - Hendrik-Jan Roest
- Department of Bacteriology, Host-Pathogen Interaction and Diagnostics, Wageningen Bioveterinary Research, Houtribweg 39, 8221 RA Lelystad, The Netherlands
| | - Ad P Koets
- Department of Bacteriology, Host-Pathogen Interaction and Diagnostics, Wageningen Bioveterinary Research, Houtribweg 39, 8221 RA Lelystad, The Netherlands
- Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 7, 3584 CL Utrecht, The Netherlands
| | - Tom N McNeilly
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK
| |
Collapse
|
13
|
Anastácio S, de Sousa SR, Saavedra MJ, da Silva GJ. Role of Goats in the Epidemiology of Coxiella burnetii. BIOLOGY 2022; 11:biology11121703. [PMID: 36552213 PMCID: PMC9774940 DOI: 10.3390/biology11121703] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Since its first description in the late 1930s, Q fever has raised many questions. Coxiella burnetii, the causative agent, is a zoonotic pathogen affecting a wide range of hosts. This airborne organism leads to an obligate, intracellular lifecycle, during which it multiplies in the mononuclear cells of the immune system and in the trophoblasts of the placenta in pregnant females. Although some issues about C. burnetii and its pathogenesis in animals remain unclear, over the years, some experimental studies on Q fever have been conducted in goats given their excretion pattern. Goats play an important role in the epidemiology and economics of C. burnetii infections, also being the focus of several epidemiological studies. Additionally, variants of the agent implicated in human long-term disease have been found circulating in goats. The purpose of this review is to summarize the latest research on C. burnetii infection and the role played by goats in the transmission of the infection to humans.
Collapse
Affiliation(s)
- Sofia Anastácio
- Vasco da Gama Research Centre (CIVG), Department of Veterinary Sciences, Vasco da Gama University School, Avenida José R. Sousa Fernandes 197 Lordemão, 3020-210 Coimbra, Portugal
- Center of Neurosciences and Cell Biology, Health Science Campus, 3000-548 Coimbra, Portugal
- Correspondence:
| | - Sérgio Ramalho de Sousa
- Vasco da Gama Research Centre (CIVG), Department of Veterinary Sciences, Vasco da Gama University School, Avenida José R. Sousa Fernandes 197 Lordemão, 3020-210 Coimbra, Portugal
| | - Maria José Saavedra
- Laboratory Medical Microbiology—Antimicrobials, Biocides and Biofilms Unit, Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal
- Centre for the Research and Technology Agro-Environmental and Biological Sciences and Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, 5000-801 Vila Real, Portugal
| | - Gabriela Jorge da Silva
- Center of Neurosciences and Cell Biology, Health Science Campus, 3000-548 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
14
|
Lau N, Thomas DR, Lee YW, Knodler LA, Newton HJ. Perturbation of ATG16L1 function impairs the biogenesis of Salmonella and Coxiella replication vacuoles. Mol Microbiol 2022; 117:235-251. [PMID: 34874584 PMCID: PMC8844213 DOI: 10.1111/mmi.14858] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 02/03/2023]
Abstract
Anti-bacterial autophagy, known as xenophagy, is a host innate immune response that targets invading pathogens for degradation. Some intracellular bacteria, such as the enteric pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium), utilize effector proteins to interfere with autophagy. One such S. Typhimurium effector, SopF, inhibits recruitment of ATG16L1 to damaged Salmonella-containing vacuoles (SCVs), thereby inhibiting the host xenophagic response. SopF is also required to maintain the integrity of the SCV during the early stages of infection. Here we show disruption of the SopF-ATG16L1 interaction leads to an increased proportion of cytosolic S. Typhimurium. Furthermore, SopF was utilized as a molecular tool to examine the requirement for ATG16L1 in the intracellular lifestyle of Coxiella burnetii, a bacterium that requires a functional autophagy pathway to replicate efficiently and form a single, spacious vacuole called the Coxiella-containing vacuole (CCV). ATG16L1 is required for CCV expansion and fusion but does not influence C. burnetii replication. In contrast, SopF did not affect CCV formation or replication, demonstrating that the contribution of ATG16L1 to CCV biogenesis is via its role in autophagy, not xenophagy. This study highlights the diverse capabilities of bacterial effector proteins to dissect the molecular details of host-pathogen interactions.
Collapse
Affiliation(s)
- Nicole Lau
- The Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - David R Thomas
- The Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Yi Wei Lee
- The Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Leigh A Knodler
- The Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia.,Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Hayley J Newton
- The Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
15
|
Coxiella burnetii Virulent Phase I and avirulent Phase II Variants Differentially Manipulate Autophagy pathway in Neutrophils. Infect Immun 2022; 90:e0053421. [PMID: 35100012 DOI: 10.1128/iai.00534-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Coxiella burnetii is an obligate intracellular gram-negative bacterium that causes Q fever in humans. Virulent C. burnetii Nine Mile Phase I (NMI) strain causes disease in animal models, while avirulent NM phase II (NMII) strain does not. In this study, we found that NMI infection induces severe splenomegaly and bacterial burden in the spleen in BALB/c mice, while NMII infection does not. Compared to NMII-infected mice, a significantly higher number of CD11b+Ly6g+ neutrophils accumulated in the liver, lung and spleen of NMI-infected mice. Thus, neutrophil accumulation correlates with NMI and NMII infection induced inflammatory response. In vitro studies also demonstrated that although NMII exhibited a higher infection rate than NMI in mouse bone-marrow neutrophils (BMNs), NMI-infected BMNs survive longer than NMII-infected BMNs. These results suggest that the differential interactions of NMI and NMII with neutrophils may be related to their ability to cause disease in animals. To understand the molecular mechanism underlying the differential interactions of NMI and NMII with neutrophils, the global transcriptomic gene expressions were compared between NMI- and NMII-infected-BMNs by RNA-seq analysis. Interestingly, several genes involved in autophagy related pathways, particularly the membrane-trafficking and lipid metabolism are upregulated in NMII-infected BMNs but downregulated in NMI-infected BMNs. Immunofluorescence and immunoblot analysis indicate that compared to NMI-infected BMNs, vacuoles in NMII-infected-BMNs exhibit increased autophagic flux along with phosphatidylserine translocation in cell membrane. Similar to neutrophils, NMII activated LC3-mediated autophagy in human macrophage. These findings suggest that NMI and NMII's differential manipulation of autophagy may relate to their pathogenesis.
Collapse
|
16
|
Osbron CA, Goodman AG. To die or not to die: Programmed cell death responses and their interactions with Coxiella burnetii infection. Mol Microbiol 2022; 117:717-736. [PMID: 35020241 PMCID: PMC9018580 DOI: 10.1111/mmi.14878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/04/2022] [Accepted: 01/09/2022] [Indexed: 12/01/2022]
Abstract
Coxiella burnetii is a Gram-negative, obligate intracellular, macrophage-tropic bacterium and the causative agent of the zoonotic disease Q fever. The epidemiology of Q fever is associated with the presence of infected animals; sheep, goats, cattle, and humans primarily become infected by inhalation of contaminated aerosols. In humans, the acute phase of the disease is characterized primarily by influenza-like symptoms, and approximately 3-5% of the infected individuals develop chronic infection. C. burnetii infection activates many types of immune responses, and the bacteria's genome encodes for numerous effector proteins that interact with host immune signaling mechanisms. Here, we will discuss two forms of programmed cell death, apoptosis and pyroptosis. Apoptosis is a form of non-inflammatory cell death that leads to phagocytosis of small membrane-bound bodies. Conversely, pyroptosis results in lytic cell death accompanied by the release of proinflammatory cytokines. Both apoptosis and pyroptosis have been implicated in the clearance of intracellular bacterial pathogens, including C. burnetii. Finally, we will discuss the role of autophagy, the degradation of unwanted cellular components, during C. burnetii infection. Together, the review of these forms of programmed cell death will open new research questions aimed at combating this highly infectious pathogen for which treatment options are limited.
Collapse
Affiliation(s)
- Chelsea A Osbron
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164
| | - Alan G Goodman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164.,Paul G. Allen School of Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164
| |
Collapse
|
17
|
Kumaresan V, Alam S, Zhang Y, Zhang G. The Feasibility of Using Coxiella burnetii Avirulent Nine Mile Phase II Viable Bacteria as a Live Attenuated Vaccine Against Q fever. Front Immunol 2021; 12:754690. [PMID: 34795669 PMCID: PMC8594375 DOI: 10.3389/fimmu.2021.754690] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
This study aimed to explore if viable C. burnetii avirulent Nine Mile phase II (NMII) can elicit protective immunity against virulent NM phase I (NMI) infection. Interestingly, mice immunized with viable NMII elicited significant protection against NMI infection at different time points post-immunization. Viable NMII induced a dose-dependent NMI-specific IgG response in mice, but all doses of NMII-immunized mice conferred a similar level of protection. Comparing different routes of immunization indicated that intranasally immunized mice showed significantly higher levels of protection than other immunization routes. The observation that viable NMII induced a similar level of long-term protection against NMI challenge as the formalin-inactivated NMI vaccine (PIV) suggests that viable NMII bacteria can induce a similar level of long-term protection against virulent NMI challenge as the PIV. Viable NMII also induced significant protection against challenge with virulent Priscilla and Scurry strains, suggesting that viable NMII can elicit broad protection. Immune sera and splenocytes from viable NMII-immunized mice are protective against NMI infection, but immune serum-receiving mice did not control NMI replication. Additionally, viable NMII conferred a comparable level of protection in wild-type, CD4+ T cell-deficient, and CD8+ T cell-deficient mice, and partial protection in B cell-deficient mice. However, NMII-immunized T cell-deficient mice were unable to prevent C. burnetii replication. Thus, both B cells and T cells are required for viable NMII-induced protective immunity but T cells may play a critical role. Collectively, this study demonstrates the feasibility of using avirulent NMII as a live attenuated vaccine against human Q fever.
Collapse
Affiliation(s)
- Venkatesh Kumaresan
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Shawkat Alam
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Yan Zhang
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Guoquan Zhang
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
18
|
Miller HK, Priestley RA, Kersh GJ. Q Fever: A troubling disease and a challenging diagnosis. CLINICAL MICROBIOLOGY NEWSLETTER 2021; 43:109-118. [PMID: 37701818 PMCID: PMC10493821 DOI: 10.1016/j.clinmicnews.2021.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Q fever is a disease caused by the bacterial pathogen Coxiella burnetii. This hardy organism can easily spread long distances in the wind, and only a few infectious aerosolized particles are necessary to cause serious illness. Presentations of Q fever disease can be wide-ranging, allowing it to masquerade as other illnesses and highlight the importance of laboratory testing for diagnosis and treatment. This review summarizes Q fever's epidemiology and clinical presentations and presents classical laboratory diagnostic assays and novel approaches to detecting this troubling disease.
Collapse
Affiliation(s)
- Halie K. Miller
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | |
Collapse
|
19
|
Kovacs-Simon A, Metters G, Norville I, Hemsley C, Titball RW. Coxiella burnetii replicates in Galleria mellonella hemocytes and transcriptome mapping reveals in vivo regulated genes. Virulence 2021; 11:1268-1278. [PMID: 32970966 PMCID: PMC7549970 DOI: 10.1080/21505594.2020.1819111] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Larvae of the greater wax moth (Galleria mellonella) are susceptible to infection with C. burnetii, an obligate intracellular bacterial pathogen. We show that bacteria are found in hemocytes after infection, and occupy vacuoles which are morphologically similar to Coxiella-containing vacuoles seen in infected mammalian phagocytes. We characterized the infection by transcriptome profiling of bacteria isolated from the hemocytes of infected larvae and identified 46 highly upregulated genes. The encoded proteins are predicted to be involved in translation, LPS biosynthesis, biotin synthesis, scavenging of reactive oxygen species, and included a T4SS effector and 30 hypothetical proteins. Some of these genes had previously been shown to be upregulated in buffalo green monkey (BGM) cells or in mice, whilst others appear to be regulated in a host-specific manner. Altogether, our results demonstrate the value of the G. mellonella model to study intracellular growth and identify potential virulence factors of C. burnetii.
Collapse
Affiliation(s)
- Andrea Kovacs-Simon
- College of Life and Environmental Sciences - Biosciences, University of Exeter , Exeter, UK
| | - Georgie Metters
- College of Life and Environmental Sciences - Biosciences, University of Exeter , Exeter, UK
| | - Isobel Norville
- CBR Division, Defence Science and Technology Laboratory , Porton Down,Salisbury, UK
| | - Claudia Hemsley
- College of Life and Environmental Sciences - Biosciences, University of Exeter , Exeter, UK
| | - Richard W Titball
- College of Life and Environmental Sciences - Biosciences, University of Exeter , Exeter, UK
| |
Collapse
|
20
|
Delaney MA, Hartigh AD, Carpentier SJ, Birkland TP, Knowles DP, Cookson BT, Frevert CW. Avoidance of the NLRP3 Inflammasome by the Stealth Pathogen, Coxiella burnetii. Vet Pathol 2021; 58:624-642. [PMID: 33357072 DOI: 10.1177/0300985820981369] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Coxiella burnetii, a highly adapted obligate intracellular bacterial pathogen and the cause of the zoonosis Q fever, is a reemerging public health threat. C. burnetii employs a Type IV secretion system (T4SS) to establish and maintain its intracellular niche and modulate host immune responses including the inhibition of apoptosis. Interactions between C. burnetii and caspase-1-mediated inflammasomes are not fully elucidated. This study confirms that C. burnetii does not activate caspase-1 during infection of mouse macrophages in vitro. C. burnetii-infected cells did not develop NLRP3 and ASC foci indicating its ability to avoid cytosolic detection. C. burnetii is unable to inhibit the pyroptosis and IL-1β secretion that is induced by potent inflammasome stimuli but rather enhances these caspase-1-mediated effects. We found that C. burnetii upregulates pro-IL-1β and robustly primes NLRP3 inflammasomes via TLR2 and MyD88 signaling. As for wildtype C. burnetii, T4SS-deficient mutants primed and potentiated NLRP3 inflammasomes. An in vivo model of pulmonary infection in C57BL/6 mice was developed. Mice deficient in NLRP3 or caspase-1 were like wildtype mice in the development and resolution of splenomegaly due to red pulp hyperplasia, and histologic lesions and macrophage kinetics, but had slightly higher pulmonary bacterial burdens at the greatest measured time point. Together these findings indicate that C. burnetii primes but avoids cytosolic detection by NLRP3 inflammasomes, which are not required for the clinical resistance of C57BL/6 mice. Determining mechanisms employed by C. burnetii to avoid cytosolic detection via NLRP3 inflammasomes will be beneficial to the development of preventative and interventional therapies for Q fever.
Collapse
Affiliation(s)
- Martha A Delaney
- Departments of Comparative Medicine and Pathology, and the Comparative Pathology Program, 7284University of Washington, Seattle, WA
- Current address: Martha A. Delaney, Zoological Pathology Program, University of Illinois, Brookfield, IL, USA
| | - Andreas den Hartigh
- Departments of Microbiology and Lab Medicine, 7284University of Washington, Seattle, WA
| | - Samuel J Carpentier
- Departments of Microbiology and Lab Medicine, 7284University of Washington, Seattle, WA
| | - Timothy P Birkland
- Departments of Comparative Medicine and Pathology, and the Comparative Pathology Program, 7284University of Washington, Seattle, WA
| | - Donald P Knowles
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, WA
- Department of Veterinary Microbiology and Pathology, 6760Washington State University, Pullman, WA
| | - Brad T Cookson
- Departments of Microbiology and Lab Medicine, 7284University of Washington, Seattle, WA
| | - Charles W Frevert
- Departments of Comparative Medicine and Pathology, and the Comparative Pathology Program, 7284University of Washington, Seattle, WA
| |
Collapse
|
21
|
Burette M, Bonazzi M. From neglected to dissected: How technological advances are leading the way to the study of Coxiella burnetii pathogenesis. Cell Microbiol 2021; 22:e13180. [PMID: 32185905 DOI: 10.1111/cmi.13180] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/08/2020] [Accepted: 01/19/2020] [Indexed: 11/26/2022]
Abstract
Coxiella burnetii is an obligate intracellular bacterial pathogen responsible for severe worldwide outbreaks of the zoonosis Q fever. The remarkable resistance to environmental stress, extremely low infectious dose and ease of dissemination, contributed to the classification of C. burnetii as a class B biothreat. Unique among intracellular pathogens, C. burnetii escapes immune surveillance and replicates within large autophagolysosome-like compartments called Coxiella-containing vacuoles (CCVs). The biogenesis of these compartments depends on the subversion of several host signalling pathways. For years, the obligate intracellular nature of C. burnetii imposed significant experimental obstacles to the study of its pathogenic traits. With the development of an axenic culture medium in 2009, C. burnetii became genetically tractable, thus allowing the implementation of mutagenesis tools and screening approaches to identify its virulence determinants and investigate its complex interaction with host cells. Here, we review the key advances that have contributed to our knowledge of C. burnetii pathogenesis, leading to the rise of this once-neglected pathogen to an exceptional organism to study the intravacuolar lifestyle.
Collapse
Affiliation(s)
- Melanie Burette
- IRIM, UMR 9004 CNRS, Université de Montpellier, Montpellier, France
| | - Matteo Bonazzi
- IRIM, UMR 9004 CNRS, Université de Montpellier, Montpellier, France
| |
Collapse
|
22
|
Long CM, Beare PA, Cockrell DC, Fintzi J, Tesfamariam M, Shaia CI, Heinzen RA. Contributions of lipopolysaccharide and the type IVB secretion system to Coxiella burnetii vaccine efficacy and reactogenicity. NPJ Vaccines 2021; 6:38. [PMID: 33741986 PMCID: PMC7979919 DOI: 10.1038/s41541-021-00296-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 02/12/2021] [Indexed: 12/15/2022] Open
Abstract
Coxiella burnetii is the bacterial causative agent of the zoonosis Q fever. The current human Q fever vaccine, Q-VAX®, is a fixed, whole cell vaccine (WCV) licensed solely for use in Australia. C. burnetii WCV administration is associated with a dermal hypersensitivity reaction in people with pre-existing immunity to C. burnetii, limiting wider use. Consequently, a less reactogenic vaccine is needed. Here, we investigated contributions of the C. burnetii Dot/Icm type IVB secretion system (T4BSS) and lipopolysaccharide (LPS) in protection and reactogenicity of fixed WCVs. A 32.5 kb region containing 23 dot/icm genes was deleted in the virulent Nine Mile phase I (NMI) strain and the resulting mutant was evaluated in guinea pig models of C. burnetii infection, vaccination-challenge, and post-vaccination hypersensitivity. The NMI ∆dot/icm strain was avirulent, protective as a WCV against a robust C. burnetii challenge, and displayed potentially altered reactogenicity compared to NMI. Nine Mile phase II (NMII) strains of C. burnetii that produce rough LPS, were similarly tested. NMI was significantly more protective than NMII as a WCV; however, both vaccines exhibited similar reactogenicity. Collectively, our results indicate that, like phase I LPS, the T4BSS is required for full virulence by C. burnetii. Conversely, unlike phase I LPS, the T4BSS is not required for vaccine-induced protection. LPS length does not appear to contribute to reactogenicity while the T4BSS may contribute to this response. NMI ∆dot/icm represents an avirulent phase I strain with full vaccine efficacy, illustrating the potential of genetically modified C. burnetii as improved WCVs.
Collapse
Affiliation(s)
- Carrie M Long
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| | - Paul A Beare
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Diane C Cockrell
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jonathan Fintzi
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Mahelat Tesfamariam
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Carl I Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Robert A Heinzen
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
23
|
Kuba M, Neha N, Newton P, Lee YW, Bennett-Wood V, Hachani A, De Souza DP, Nijagal B, Dayalan S, Tull D, McConville MJ, Sansom FM, Newton HJ. EirA Is a Novel Protein Essential for Intracellular Replication of Coxiella burnetii. Infect Immun 2020; 88:e00913-19. [PMID: 32205404 PMCID: PMC7240097 DOI: 10.1128/iai.00913-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/18/2020] [Indexed: 02/06/2023] Open
Abstract
The zoonotic bacterial pathogen Coxiella burnetii is the causative agent of Q fever, a febrile illness which can cause a serious chronic infection. C. burnetii is a unique intracellular bacterium which replicates within host lysosome-derived vacuoles. The ability of C. burnetii to replicate within this normally hostile compartment is dependent on the activity of the Dot/Icm type 4B secretion system. In a previous study, a transposon mutagenesis screen suggested that the disruption of the gene encoding the novel protein CBU2072 rendered C. burnetii incapable of intracellular replication. This protein, subsequently named EirA (essential for intracellular replication A), is indispensable for intracellular replication and virulence, as demonstrated by infection of human cell lines and in vivo infection of Galleria mellonella The putative N-terminal signal peptide is essential for protein function but is not required for localization of EirA to the bacterial inner membrane compartment and axenic culture supernatant. In the absence of EirA, C. burnetii remains viable but nonreplicative within the host phagolysosome, as coinfection with C. burnetii expressing native EirA rescues the replicative defect in the mutant strain. In addition, while the bacterial ultrastructure appears to be intact, there is an altered metabolic profile shift in the absence of EirA, suggesting that EirA may impact overall metabolism. Most strikingly, in the absence of EirA, Dot/Icm effector translocation was inhibited even when EirA-deficient C. burnetii replicated in the wild type (WT)-supported Coxiella containing vacuoles. EirA may therefore have a novel role in the control of Dot/Icm activity and represent an important new therapeutic target.
Collapse
Affiliation(s)
- Miku Kuba
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Nitika Neha
- Metabolomics Australia, The Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Patrice Newton
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Yi Wei Lee
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Vicki Bennett-Wood
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - David P De Souza
- Metabolomics Australia, The Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Brunda Nijagal
- Metabolomics Australia, The Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Saravanan Dayalan
- Metabolomics Australia, The Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Dedreia Tull
- Metabolomics Australia, The Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Malcolm J McConville
- Metabolomics Australia, The Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, The Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Fiona M Sansom
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Hayley J Newton
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
The Effect of pH on Antibiotic Efficacy against Coxiella burnetii in Axenic Media. Sci Rep 2019; 9:18132. [PMID: 31792307 PMCID: PMC6889355 DOI: 10.1038/s41598-019-54556-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/11/2019] [Indexed: 01/06/2023] Open
Abstract
Coxiella burnetii, the etiologic agent of Q fever, replicates in an intracellular phagolysosome with pH between 4 and 5. The impact of this low pH environment on antimicrobial treatment is not well understood. An in vitro system for testing antibiotic susceptibility of C. burnetii in axenic media was set up to evaluate the impact of pH on C. burnetii growth and survival in the presence and absence of antimicrobial agents. The data show that C. burnetii does not grow in axenic media at pH 6.0 or higher, but the organisms remain viable. At pH of 4.75, 5.25, and 5.75 moxifloxacin, doxycycline, and rifampin are effective at preventing growth of C. burnetii in axenic media, with moxifloxacin and doxycycline being bacteriostatic and rifampin having bactericidal activity. The efficacy of doxycycline and moxifloxacin improved at higher pH, whereas rifampin activity was pH independent. Hydroxychloroquine is thought to inhibit growth of C. burnetii in vivo by raising the pH of typically acidic intracellular compartments. It had no direct bactericidal or bacteriostatic activity on C. burnetii in axenic media, suggesting that raising pH of acidic intracellular compartments is its primary mechanism of action in vivo. The data suggest that doxycycline and hydroxychloroquine are primarily independent bacteriostatic agents.
Collapse
|
25
|
Dresler J, Klimentova J, Pajer P, Salovska B, Fucikova AM, Chmel M, Schmoock G, Neubauer H, Mertens-Scholz K. Quantitative Proteome Profiling of Coxiella burnetii Reveals Major Metabolic and Stress Differences Under Axenic and Cell Culture Cultivation. Front Microbiol 2019; 10:2022. [PMID: 31620097 PMCID: PMC6759588 DOI: 10.3389/fmicb.2019.02022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/19/2019] [Indexed: 12/14/2022] Open
Abstract
Coxiella burnetii is the causative agent of the zoonotic disease Q fever. To date, the lipopolysaccharide (LPS) is the only defined and characterized virulence determinant of C. burnetii. In this study, proteome profiles of C. burnetii Nine Mile phase I (RSA 493, NMI) and its isogenic Nine Mile phase II (RSA 439 NMII) isolate with a deep rough LPS were compared on L-929 mouse fibroblasts and in complex (ACCM-2), and defined (ACCM-D) media. Whole proteome extracts were analyzed using a label-free quantification approach. Between 659 and 1,046 C. burnetii proteins of the 2,132 annotated coding sequences (CDS) were identified in any particular experiment. Proteome profiles clustered according to the cultivation conditions used, indicating different regulation patterns. NMI proteome profiles compared to NMII in ACCM-D indicate transition from an exponential to a stationary phase. The levels of regulatory proteins such as RpoS, CsrA2, UspA1, and UspA2 were increased. Comparison of the oxidative stress response of NMI and NMII indicated that ACCM-2 represents a high oxidative stress environment. Expression of peroxidases, superoxide dismutases, as well as thioredoxins was increased for NMI. In contrast, in ACCM-D, only osmoregulation seems to be necessary. Proteome profiles of NMII do not differ and indicate that both axenic media represent similar oxidative stress environments. Deep rough LPS causes changes of the outer membrane stability and fluidity. This might be one reason for the observed differences. Proteins associated with the T4SS and Sec translocon as well as several effector proteins were detectable under all three conditions. Interestingly, none of these putatively secreted proteins are upregulated in ACCM-2 compared to ACCM-D, and L-929 mouse fibroblasts. Curiously, a higher similarity of proteomic patterns (overlapping up- and downregulated proteins) of ACCM-D and bacteria grown in cell culture was observed. Particularly, the proteins involved in a better adaptation or homeostasis in response to the harsh environment of the parasitophorous vacuole were demonstrated for NMI. This semi-quantitative proteomic analysis of C. burnetii compared axenically grown bacteria to those propagated in cell culture.
Collapse
Affiliation(s)
| | - Jana Klimentova
- Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czechia
| | - Petr Pajer
- Military Health Institute, Prague, Czechia
| | - Barbora Salovska
- Department of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | | | - Martin Chmel
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| | - Gernot Schmoock
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Jena, Germany
| | - Heinrich Neubauer
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Jena, Germany
| | - Katja Mertens-Scholz
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Jena, Germany
| |
Collapse
|
26
|
Kohl L, Hayek I, Daniel C, Schulze-Lührmann J, Bodendorfer B, Lührmann A, Lang R. MyD88 Is Required for Efficient Control of Coxiella burnetii Infection and Dissemination. Front Immunol 2019; 10:165. [PMID: 30800124 PMCID: PMC6376249 DOI: 10.3389/fimmu.2019.00165] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 01/18/2019] [Indexed: 12/28/2022] Open
Abstract
The intracellular pathogen Coxiella (C.) burnetii causes Q fever, a usually self-limiting respiratory infection that becomes chronic and severe in some patients. Innate immune recognition of C. burnetii and its role in the decision between resolution and chronicity is not understood well. However, TLR2 is important for the response to C. burnetii in mice, and genetic polymorphisms in Myd88 have been associated with chronic Q fever in humans. Here, we have employed MyD88-deficient mice in infection models with the attenuated C. burnetii Nine Mile phase II strain (NMII). Myd88−/− macrophages failed to restrict the growth of NMII in vitro, and to upregulate production of the cytokines TNF, IL-6, and IL-10. Following intraperitoneal infection, NMII bacterial burden was significantly higher on day 5 and 20 in organs of Myd88−/− mice. After infection via the natural route by intratracheal injection, a higher bacterial load in the lung and increased dissemination of NMII to other organs was observed in MyD88-deficient mice. While wild-type mice essentially cleared NMII on day 27 after intratracheal infection, it was still readily detectable on day 42 in multiple organs in the absence of MyD88. Despite the elevated bacterial load, Myd88−/− mice had less granulomatous inflammation and expressed significantly lower levels of chemoattractants, inflammatory cytokines, and of several IFNγ-induced genes relevant for control of intracellular pathogens. Together, our results show that MyD88-dependent signaling is essential for early control of C. burnetii replication and to prevent systemic spreading. The continued presence of NMII in the organs of Myd88−/− mice constitutes a new mouse model to study determinants of chronicity and resolution in Q fever.
Collapse
Affiliation(s)
- Lisa Kohl
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Inaya Hayek
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jan Schulze-Lührmann
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Barbara Bodendorfer
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anja Lührmann
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
27
|
Crabill E, Schofield WB, Newton HJ, Goodman AL, Roy CR. Dot/Icm-Translocated Proteins Important for Biogenesis of the Coxiella burnetii-Containing Vacuole Identified by Screening of an Effector Mutant Sublibrary. Infect Immun 2018; 86:e00758-17. [PMID: 29339460 PMCID: PMC5865027 DOI: 10.1128/iai.00758-17] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/05/2018] [Indexed: 01/25/2023] Open
Abstract
Coxiella burnetii is an intracellular pathogen that replicates in a lysosome-derived vacuole. A determinant necessary for C. burnetii virulence is the Dot/Icm type IVB secretion system (T4SS). The Dot/Icm system delivers more than 100 proteins, called type IV effectors (T4Es), across the vacuolar membrane into the host cell cytosol. Several T4Es have been shown to be important for vacuolar biogenesis. Here, transposon (Tn) insertion sequencing technology (INSeq) was used to identify C. burnetii Nine Mile phase II mutants in an arrayed library, which facilitated the identification and clonal isolation of mutants deficient in 70 different T4E proteins. These effector mutants were screened in HeLa cells for deficiencies in Coxiella-containing vacuole (CCV) biogenesis. This screen identified and validated seven new T4Es that were important for vacuole biogenesis. Loss-of-function mutations in cbu0414 (coxH1), cbu0513, cbu0978 (cem3), cbu1387 (cem6), cbu1524 (caeA), cbu1752, or cbu2028 resulted in a small-vacuole phenotype. These seven mutant strains produced small CCVs in all cells tested, which included macrophage-like cells. The cbu2028::Tn mutant, though unable to develop large CCVs, had intracellular replication rates similar to the rate of the parental strain of C. burnetii, whereas the other six effector mutants defective in CCV biogenesis displayed significant reductions in intracellular replication. Vacuoles created by the cbu0513::Tn mutant did not accumulate lipidated microtubule-associated protein 1A/1B light chain 3 (LC3-II), suggesting a failure in fusion of the CCV with autophagosomes. These seven T4E proteins add to the growing repertoire of C. burnetii factors that contribute to CCV biogenesis.
Collapse
Affiliation(s)
- Emerson Crabill
- Department of Microbial Pathogenesis, Yale University School of Medicine, Boyer Center for Molecular Medicine, New Haven, Connecticut, USA
| | - Whitman B Schofield
- Department of Microbial Pathogenesis, Yale University School of Medicine, Boyer Center for Molecular Medicine, New Haven, Connecticut, USA
- Microbial Sciences Institute, Yale University, West Haven, Connecticut, USA
| | - Hayley J Newton
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Andrew L Goodman
- Department of Microbial Pathogenesis, Yale University School of Medicine, Boyer Center for Molecular Medicine, New Haven, Connecticut, USA
- Microbial Sciences Institute, Yale University, West Haven, Connecticut, USA
| | - Craig R Roy
- Department of Microbial Pathogenesis, Yale University School of Medicine, Boyer Center for Molecular Medicine, New Haven, Connecticut, USA
| |
Collapse
|
28
|
Beare PA, Jeffrey BM, Long CM, Martens CM, Heinzen RA. Genetic mechanisms of Coxiella burnetii lipopolysaccharide phase variation. PLoS Pathog 2018; 14:e1006922. [PMID: 29481553 PMCID: PMC5843353 DOI: 10.1371/journal.ppat.1006922] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/08/2018] [Accepted: 02/05/2018] [Indexed: 12/30/2022] Open
Abstract
Coxiella burnetii is an intracellular pathogen that causes human Q fever, a disease that normally presents as a severe flu-like illness. Due to high infectivity and disease severity, the pathogen is considered a risk group 3 organism. Full-length lipopolysaccharide (LPS) is required for full virulence and disease by C. burnetii and is the only virulence factor currently defined by infection of an immunocompetent animal. Transition of virulent phase I bacteria with smooth LPS, to avirulent phase II bacteria with rough LPS, occurs during in vitro passage. Semi-rough intermediate forms are also observed. Here, the genetic basis of LPS phase conversion was investigated to obtain a more complete understanding of C. burnetii pathogenesis. Whole genome sequencing of strains producing intermediate and/or phase II LPS identified several common mutations in predicted LPS biosynthesis genes. After passage in broth culture for 30 weeks, phase I strains from different genomic groups exhibited similar phase transition kinetics and elevation of mutations in LPS biosynthesis genes. Targeted mutagenesis and genetic complementation using a new C. burnetii nutritional selection system based on lysine auxotrophy confirmed that six of the mutated genes were necessary for production of phase I LPS. Disruption of two of these genes in a C. burnetii phase I strain resulted in production of phase II LPS, suggesting inhibition of the encoded enzymes could represent a new therapeutic strategy for treatment of Q fever. Additionally, targeted mutagenesis of genes encoding LPS biosynthesis enzymes can now be used to construct new phase II strains from different genomic groups for use in pathogen-host studies at a risk group 2 level.
Collapse
Affiliation(s)
- Paul A. Beare
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Brendan M. Jeffrey
- Bioinformatics and Computational Biosciences Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Carrie M. Long
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Craig M. Martens
- Research Technologies Section, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Robert A. Heinzen
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
- * E-mail:
| |
Collapse
|
29
|
Pechstein J, Schulze-Luehrmann J, Lührmann A. Coxiella burnetii as a useful tool to investigate bacteria-friendly host cell compartments. Int J Med Microbiol 2017; 308:77-83. [PMID: 28935173 DOI: 10.1016/j.ijmm.2017.09.010] [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] [Received: 06/29/2017] [Revised: 08/21/2017] [Accepted: 09/11/2017] [Indexed: 10/25/2022] Open
Abstract
Coxiella burnetii is an obligate intracellular and airborne pathogen which can cause the zoonotic disease Q fever. After inhalation of contaminated aerosols alveolar macrophages are taking up C. burnetii into a phagosome. This phagosome matures to a very large vacuole called the C. burnetii-containing vacuole (CCV). Host endogenous and bacterial driven processes lead to the biogenesis of this unusual compartment, which resembles partially a phagolysosome. However, there are several important differences to the classical phagolysosome, which are crucial for the ability of C. burnetii to replicate intracellularly and depend on a functional type IV secretion system (T4SS). The T4SS delivers effector proteins into the host cell cytoplasm to redirect intracellular processes, leading to the establishment of a microenvironment allowing bacterial replication. This article summarizes the current knowledge of the microenvironment permissive for C. burnetii replication.
Collapse
Affiliation(s)
- Julian Pechstein
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| | - Jan Schulze-Luehrmann
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| | - Anja Lührmann
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany.
| |
Collapse
|
30
|
Kuley R, Kuijt E, Smits MA, Roest HIJ, Smith HE, Bossers A. Genome Plasticity and Polymorphisms in Critical Genes Correlate with Increased Virulence of Dutch Outbreak-Related Coxiella burnetii Strains. Front Microbiol 2017; 8:1526. [PMID: 28848533 PMCID: PMC5554327 DOI: 10.3389/fmicb.2017.01526] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/28/2017] [Indexed: 12/20/2022] Open
Abstract
Coxiella burnetii is an obligate intracellular bacterium and the etiological agent of Q fever. During 2007-2010 the largest Q fever outbreak ever reported occurred in The Netherlands. It is anticipated that strains from this outbreak demonstrated an increased zoonotic potential as more than 40,000 individuals were assumed to be infected. The acquisition of novel genetic factors by these C. burnetii outbreak strains, such as virulence-related genes, has frequently been proposed and discussed, but is not proved yet. In the present study, the whole genome sequence of several Dutch strains (CbNL01 and CbNL12 genotypes), a few additionally selected strains from different geographical locations and publicly available genome sequences were used for a comparative bioinformatics approach. The study focuses on the identification of specific genetic differences in the outbreak related CbNL01 strains compared to other C. burnetii strains. In this approach we investigated the phylogenetic relationship and genomic aspects of virulence and host-specificity. Phylogenetic clustering of whole genome sequences showed a genotype-specific clustering that correlated with the clustering observed using Multiple Locus Variable-number Tandem Repeat Analysis (MLVA). Ortholog analysis on predicted genes and single nucleotide polymorphism (SNP) analysis of complete genome sequences demonstrated the presence of genotype-specific gene contents and SNP variations in C. burnetii strains. It also demonstrated that the currently used MLVA genotyping methods are highly discriminatory for the investigated outbreak strains. In the fully reconstructed genome sequence of the Dutch outbreak NL3262 strain of the CbNL01 genotype, a relatively large number of transposon-linked genes were identified as compared to the other published complete genome sequences of C. burnetii. Additionally, large numbers of SNPs in its membrane proteins and predicted virulence-associated genes were identified in all Dutch outbreak strains compared to the NM reference strain and other strains of the CbNL12 genotype. The presence of large numbers of transposable elements and mutated genes, thereof most likely resulted in high level of genome rearrangements and genotype-specific pathogenicity of outbreak strains. Thus, the epidemic potential of Dutch outbreak strains could be linked to increased genome plasticity and mutations in critical genes involved in virulence and the evasion of the host immune system.
Collapse
Affiliation(s)
- Runa Kuley
- Department of Infection Biology, Wageningen Bioveterinary ResearchLelystad, Netherlands
- Host Microbe Interactomics, Wageningen University and Research CentreWageningen, Netherlands
| | - Eric Kuijt
- Department of Infection Biology, Wageningen Bioveterinary ResearchLelystad, Netherlands
| | - Mari A. Smits
- Department of Infection Biology, Wageningen Bioveterinary ResearchLelystad, Netherlands
- Host Microbe Interactomics, Wageningen University and Research CentreWageningen, Netherlands
| | - Hendrik I. J. Roest
- Department of Bacteriology and Epidemiology, Wageningen Bioveterinary ResearchLelystad, Netherlands
| | - Hilde E. Smith
- Department of Infection Biology, Wageningen Bioveterinary ResearchLelystad, Netherlands
| | - Alex Bossers
- Department of Infection Biology, Wageningen Bioveterinary ResearchLelystad, Netherlands
| |
Collapse
|
31
|
Host and Bacterial Factors Control Susceptibility of Drosophila melanogaster to Coxiella burnetii Infection. Infect Immun 2017; 85:IAI.00218-17. [PMID: 28438980 DOI: 10.1128/iai.00218-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 04/17/2017] [Indexed: 12/12/2022] Open
Abstract
Coxiella burnetii is the causative agent of Q fever, a zoonotic disease that threatens both human and animal health. Due to the paucity of experimental animal models, little is known about how host factors interface with bacterial components and affect pathogenesis. Here, we used Drosophila melanogaster, in conjunction with the biosafety level 2 (BSL2) Nine Mile phase II (NMII) clone 4 strain of C. burnetii, as a model to investigate host and bacterial components implicated in infection. We demonstrate that adult Drosophila flies are susceptible to C. burnetii NMII infection and that this bacterial strain, which activates the immune deficiency (IMD) pathway, is able to replicate and cause mortality in the animals. We show that in the absence of Eiger, the only known tumor necrosis factor (TNF) superfamily homolog in Drosophila, Coxiella-infected flies exhibit reduced mortality from infection. We also demonstrate that the Coxiella type 4 secretion system (T4SS) is critical for the formation of the Coxiella-containing vacuole and establishment of infection in Drosophila Altogether, our data reveal that the Drosophila TNF homolog Eiger and the Coxiella T4SS are implicated in the pathogenesis of C. burnetii in flies. The Drosophila/NMII model mimics relevant aspects of the infection in mammals, such as a critical role of host TNF and the bacterial T4SS in pathogenesis. Our work also demonstrates the usefulness of this BSL2 model to investigate both host and Coxiella components implicated in infection.
Collapse
|
32
|
Whole-Genome Sequence of Coxiella burnetii Nine Mile RSA439 (Phase II, Clone 4), a Laboratory Workhorse Strain. GENOME ANNOUNCEMENTS 2017; 5:5/23/e00471-17. [PMID: 28596399 PMCID: PMC5465618 DOI: 10.1128/genomea.00471-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Here, we report the whole-genome sequence of Coxiella burnetii Nine Mile RSA439 (phase II, clone 4), a laboratory strain used extensively to investigate the biology of this intracellular bacterial pathogen. The genome consists of a 1.97-Mb chromosome and a 37.32-kb plasmid.
Collapse
|
33
|
van Schaik EJ, Case ED, Martinez E, Bonazzi M, Samuel JE. The SCID Mouse Model for Identifying Virulence Determinants in Coxiella burnetii. Front Cell Infect Microbiol 2017; 7:25. [PMID: 28217558 PMCID: PMC5289997 DOI: 10.3389/fcimb.2017.00025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/19/2017] [Indexed: 01/09/2023] Open
Abstract
Coxiella burnetii is an intracellular, zoonotic pathogen that is the causative agent of Q fever. Infection most frequently occurs after inhalation of contaminated aerosols, which can lead to acute, self-limiting febrile illness or more serve chronic infections such as hepatitis or endocarditis. Macrophages are the principal target cells during infection where C. burnetii resides and replicates within a unique phagolysosome-like compartment, the Coxiella-containing vacuole (CCV). The first virulence determinant described as necessary for infection was full-length lipopolysaccarride (LPS); spontaneous rough mutants (phase II) arise after passage in immuno-incompetent hosts. Phase II C. burnetii are attenuated in immuno-competent animals, but are fully capable of infecting a variety of host cells in vitro. A clonal strain of the Nine Mile isolate (RSA439, clone 4), has a 26 KDa chromosomal deletion that includes LPS biosynthetic genes and is uniquely approved for use in BL2/ABL2 conditions. With the advances of axenic media and genetic tools for C. burnetii research, the characterization of novel virulence determinants is ongoing and almost exclusively performed using this attenuated clone. A major problem with predicting essential virulence loci with RSA439 is that, although some cell-autonomous phenotypes can be assessed in tissue culture, no animal model for assessing pathogenesis has been defined. Here we describe the use of SCID mice for predicting virulence factors of C. burnetii, in either independent or competitive infections. We propose that this model allows for the identification of mutations that are competent for intracellular replication in vitro, but attenuated for growth in vivo and predict essential innate immune responses modulated by the pathogen during infection as a central pathogenic strategy.
Collapse
Affiliation(s)
- Erin J. van Schaik
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M UniversityBryan, TX, USA
| | - Elizabeth D. Case
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M UniversityBryan, TX, USA
| | - Eric Martinez
- Centre National de la Recherche Scientifique, Formation de Recherche en Évolution 3689, Centre d'études d'agents Pathogènes et Biotechnologies Pour la Santé, Université MontpellierMontpellier, France
| | - Matteo Bonazzi
- Centre National de la Recherche Scientifique, Formation de Recherche en Évolution 3689, Centre d'études d'agents Pathogènes et Biotechnologies Pour la Santé, Université MontpellierMontpellier, France
| | - James E. Samuel
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M UniversityBryan, TX, USA
| |
Collapse
|
34
|
Newton P, Latomanski EA, Newton HJ. Applying Fluorescence Resonance Energy Transfer (FRET) to Examine Effector Translocation Efficiency by Coxiella burnetii during siRNA Silencing. J Vis Exp 2016. [PMID: 27501079 DOI: 10.3791/54210] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Coxiella burnetii, the causative agent of Q fever, is an intracellular pathogen that relies on a Type IV Dot/Icm Secretion System to establish a replicative niche. A cohort of effectors are translocated through this system into the host cell to manipulate host processes and allow the establishment of a unique lysosome-derived vacuole for replication. The method presented here involves the combination of two well-established techniques: specific gene silencing using siRNA and measurement of effector translocation using a FRET-based substrate that relies on β-lactamase activity. Applying these two approaches, we can begin to understand the role of host factors in bacterial secretion system function and effector translocation. In this study we examined the role of Rab5A and Rab7A, both important regulators of the endocytic trafficking pathway. We demonstrate that silencing the expression of either protein results in a decrease in effector translocation efficiency. These methods can be easily modified to examine other intracellular and extracellular pathogens that also utilize secretion systems. In this way, a global picture of host factors involved in bacterial effector translocation may be revealed.
Collapse
Affiliation(s)
- Patrice Newton
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne
| | - Eleanor A Latomanski
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne
| | - Hayley J Newton
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne;
| |
Collapse
|
35
|
Primary Role for Toll-Like Receptor-Driven Tumor Necrosis Factor Rather than Cytosolic Immune Detection in Restricting Coxiella burnetii Phase II Replication within Mouse Macrophages. Infect Immun 2016; 84:998-1015. [PMID: 26787725 DOI: 10.1128/iai.01536-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 12/13/2022] Open
Abstract
Coxiella burnetii replicates within permissive host cells by employing a Dot/Icm type IV secretion system (T4SS) to translocate effector proteins that direct the formation of a parasitophorous vacuole. C57BL/6 mouse macrophages restrict the intracellular replication of the C. burnetii. Nine Mile phase II (NMII) strain. However, eliminating Toll-like receptor 2 (TLR2) permits bacterial replication, indicating that the restriction of bacterial replication is immune mediated. Here, we examined whether additional innate immune pathways are employed by C57BL/6 macrophages to sense and restrict NMII replication. In addition to the known role of TLR2 in detecting and restricting NMII infection, we found that TLR4 also contributes to cytokine responses but is not required to restrict bacterial replication. Furthermore, the TLR signaling adaptors MyD88 and Trif are required for cytokine responses and restricting bacterial replication. The C. burnetii NMII T4SS translocates bacterial products into C57BL/6 macrophages. However, there was little evidence of cytosolic immune sensing of NMII, as there was a lack of inflammasome activation, T4SS-dependent cytokine responses, and robust type I interferon (IFN) production, and these pathways were not required to restrict bacterial replication. Instead, endogenous tumor necrosis factor (TNF) produced upon TLR sensing of C. burnetii NMII was required to control bacterial replication. Therefore, our findings indicate a primary role for TNF produced upon immune detection of C. burnetii NMII by TLRs, rather than cytosolic PRRs, in enabling C57BL/6 macrophages to restrict bacterial replication.
Collapse
|
36
|
Schulze-Luehrmann J, Eckart RA, Ölke M, Saftig P, Liebler-Tenorio E, Lührmann A. LAMP proteins account for the maturation delay during the establishment of the Coxiella burnetii-containing vacuole. Cell Microbiol 2016; 18:181-94. [PMID: 26249821 DOI: 10.1111/cmi.12494] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 07/21/2015] [Accepted: 07/21/2015] [Indexed: 12/28/2022]
Abstract
The obligate intracellular pathogen Coxiella burnetii replicates in a large phagolysosomal-like vacuole. Currently, both host and bacterial factors required for creating this replicative parasitophorous C. burnetii-containing vacuole (PV) are poorly defined. Here, we assessed the contributions of the most abundant proteins of the lysosomal membrane, LAMP-1 and LAMP-2, to the establishment and maintenance of the PV. Whereas these proteins were not critical for uptake of C. burnetii, they influenced the intracellular replication of C. burnetii. In LAMP-1/2 double-deficient fibroblasts as well as in LAMP-1/2 knock-down cells, C. burnetii establishes a significantly smaller, yet faster maturing vacuole, which harboured more bacteria. The accelerated maturation of PVs in LAMP double-deficient fibroblasts, which was partially or fully reversed by ectopic expression of LAMP-1 or LAMP-2, respectively, was characterized by an increased fusion rate with endosomes, lysosomes and bead-containing phagosomes, but not by different fusion kinetics with autophagy vesicles. These findings establish that LAMP proteins are critical for the maturation delay of PVs. Unexpectedly, neither the creation of the spacious vacuole nor the delay in maturation was found to be prerequisites for the intracellular replication of C. burnetii.
Collapse
Affiliation(s)
- Jan Schulze-Luehrmann
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Germany
| | - Rita A Eckart
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Germany
| | - Martha Ölke
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Germany
| | - Paul Saftig
- Biochemical Institute, Christian Albrechts University of Kiel, Kiel, Germany
| | | | - Anja Lührmann
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Germany
| |
Collapse
|
37
|
Kuley R, Bossers-deVries R, Smith HE, Smits MA, Roest HIJ, Bossers A. Major differential gene regulation in Coxiella burnetii between in vivo and in vitro cultivation models. BMC Genomics 2015; 16:953. [PMID: 26572556 PMCID: PMC4647677 DOI: 10.1186/s12864-015-2143-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/23/2015] [Indexed: 11/29/2022] Open
Abstract
Background Coxiella burnetii is the causative agent of the zoonotic disease Q fever. As it is an intracellular pathogen, infection by C. burnetii requires adaptation to its eukaryotic host and intracellular environment. The recently developed cell-free medium also allows the bacteria to propagate without host cells, maintaining its infection potential. The adaptation to different hosts or extracellular environments has been assumed to involve genome-wide modulation of C. burnetii gene expression. However, little is currently known about these adaptation events which are critical for understanding the intracellular survival of C. burnetii. Results We studied C. burnetii genome–wide transcriptional patterns in vivo (mice spleen) and in cell and cell-free in vitro culture models to examine its metabolic pathways and virulence associated gene expression patterns that are required to colonize and persist in different environments. Within each model, the gene expression profiles of the Dutch C. burnetii outbreak strain (602) and NM reference strains were largely similar. In contrast, modulation of gene-expression was strongly influenced by the cultivation method, indicating adaptation of the bacterium to available components. Genome–wide expression profiles of C. burnetii from in vitro cell culture were more similar to those seen for in vivo conditions, while gene expression profiles of cell-free culture were more distant to in vivo. Under in vivo conditions, significant alterations of genes involved in metabolism and virulence were identified. We observed that C. burnetii under in vivo conditions predominantly uses glucose as a carbon source (mostly for biosynthetic processes) and fatty acids for energy generation. C. burnetii experienced nutrient limitation and anaerobiosis as major stressors, while phosphate limitation was identified as an important signal for intracellular growth inside eukaryotic host cells. Finally, the in vivo environment significantly induced expression of several virulence genes, including those implicated in LPS synthesis, colonization, host component modulation and DNA repair mechanisms. Conclusion Our study shows that C. burnetii, with its relative small genome, requires only a subset of core gene functions to survive under in vitro conditions, but requires the induction of full repertoire of genes for successful pathogenesis and thriving in harsh environments in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2143-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Runa Kuley
- Department of Infection Biology, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands.,Host Microbe Interactomics, Wageningen University, Wageningen, The Netherlands
| | - Ruth Bossers-deVries
- Department of Infection Biology, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands
| | - Hilde E Smith
- Department of Infection Biology, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands
| | - Mari A Smits
- Department of Infection Biology, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands.,Host Microbe Interactomics, Wageningen University, Wageningen, The Netherlands
| | - Hendrik I J Roest
- Department of Bacteriology and TSEs, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands
| | - Alex Bossers
- Department of Infection Biology, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands.
| |
Collapse
|
38
|
Gottlieb Y, Lalzar I, Klasson L. Distinctive Genome Reduction Rates Revealed by Genomic Analyses of Two Coxiella-Like Endosymbionts in Ticks. Genome Biol Evol 2015; 7:1779-96. [PMID: 26025560 PMCID: PMC4494066 DOI: 10.1093/gbe/evv108] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Genome reduction is a hallmark of symbiotic genomes, and the rate and patterns of gene loss associated with this process have been investigated in several different symbiotic systems. However, in long-term host-associated coevolving symbiont clades, the genome size differences between strains are normally quite small and hence patterns of large-scale genome reduction can only be inferred from distant relatives. Here we present the complete genome of a Coxiella-like symbiont from Rhipicephalus turanicus ticks (CRt), and compare it with other genomes from the genus Coxiella in order to investigate the process of genome reduction in a genus consisting of intracellular host-associated bacteria with variable genome sizes. The 1.7-Mb CRt genome is larger than the genomes of most obligate mutualists but has a very low protein-coding content (48.5%) and an extremely high number of identifiable pseudogenes, indicating that it is currently undergoing genome reduction. Analysis of encoded functions suggests that CRt is an obligate tick mutualist, as indicated by the possible provisioning of the tick with biotin (B7), riboflavin (B2) and other cofactors, and by the loss of most genes involved in host cell interactions, such as secretion systems. Comparative analyses between CRt and the 2.5 times smaller genome of Coxiella from the lone star tick Amblyomma americanum (CLEAA) show that many of the same gene functions are lost and suggest that the large size difference might be due to a higher rate of genome evolution in CLEAA generated by the loss of the mismatch repair genes mutSL. Finally, sequence polymorphisms in the CRt population sampled from field collected ticks reveal up to one distinct strain variant per tick, and analyses of mutational patterns within the population suggest that selection might be acting on synonymous sites. The CRt genome is an extreme example of a symbiont genome caught in the act of genome reduction, and the comparison between CLEAA and CRt indicates that losses of particular genes early on in this process can potentially greatly influence the speed of this process.
Collapse
Affiliation(s)
- Yuval Gottlieb
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Itai Lalzar
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Lisa Klasson
- Molecular Evolution, Department of Cell and Molecular Biology, Uppsala University, Sweden
| |
Collapse
|
39
|
Moffatt JH, Newton P, Newton HJ. Coxiella burnetii: turning hostility into a home. Cell Microbiol 2015; 17:621-31. [PMID: 25728389 DOI: 10.1111/cmi.12432] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/08/2015] [Accepted: 02/24/2015] [Indexed: 01/20/2023]
Abstract
Coxiella burnetii, the causative agent of the human disease Q fever, is a unique intracellular bacterial pathogen. Coxiella replicates to high numbers within a pathogen-derived lysosome-like vacuole, thriving within a low pH, highly proteolytic and oxidative environment. In 2009, researchers developed means to axenically culture Coxiella paving the way for the development of tools to genetically manipulate the organism. These advances have revolutionized our capacity to examine the pathogenesis of Coxiella. In recent years, targeted and random mutant strains have been used to demonstrate that the Dot/Icm type IV secretion system is essential for intracellular replication of Coxiella. Current research is focused towards understanding the unique cohort of over 130 effector proteins that are translocated into the host cell. Mutagenesis screens have been employed to identify effectors that play important roles for the biogenesis of the Coxiella-containing vacuole and intracellular replication of Coxiella. A surprisingly high number of effector mutants demonstrate significant intracellular growth defects, and future studies on the molecular function of these effectors will provide great insight into the pathogenesis of Coxiella. Already, this expanse of new data implicates many eukaryotic processes that are targeted by the arsenal of Coxiella effectors including autophagy, apoptosis and vesicular trafficking.
Collapse
Affiliation(s)
- Jennifer H Moffatt
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Vic., Australia
| | | | | |
Collapse
|
40
|
Kuley R, Smith HE, Frangoulidis D, Smits MA, Jan Roest HI, Bossers A. Cell-free propagation of Coxiella burnetii does not affect its relative virulence. PLoS One 2015; 10:e0121661. [PMID: 25793981 PMCID: PMC4368683 DOI: 10.1371/journal.pone.0121661] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/02/2015] [Indexed: 12/02/2022] Open
Abstract
Q fever is caused by the obligate intracellular bacterium Coxiella burnetii. In vitro growth of the bacterium is usually limited to viable eukaryotic host cells imposing experimental constraints for molecular studies, such as the identification and characterisation of major virulence factors. Studies of pathogenicity may benefit from the recent development of an extracellular growth medium for C. burnetii. However, it is crucial to investigate the consistency of the virulence phenotype of strains propagated by the two fundamentally different culturing systems. In the present study, we assessed the viability of C. burnetii and the lipopolysaccaride (LPS) encoding region of the bacteria in both culture systems as indirect but key parameters to the infection potential of C. burnetii. Propidium monoazide (PMA) treatment-based real-time PCR was used for enumeration of viable C. burnetii which were validated by fluorescent infectious focus forming unit counting assays. Furthermore, RNA isolated from C. burnetiipropagated in both the culture systems was examined for LPS-related gene expression. All thus far known LPS-related genes were found to be expressed in early passages in both culturing systems indicating the presence of predominantly the phase I form of C. burnetii. Finally, we used immune-competent mice to provide direct evidence, that the relative virulence of different C. burnetii strains is essentially the same for both axenic and cell-based methods of propagation.
Collapse
Affiliation(s)
- Runa Kuley
- Department of Infection Biology, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands
- Host Microbe Interactomics, Wageningen University, Wageningen, The Netherlands
| | - Hilde E. Smith
- Department of Infection Biology, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands
| | | | - Mari A. Smits
- Department of Infection Biology, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands
- Host Microbe Interactomics, Wageningen University, Wageningen, The Netherlands
| | - Hendrik I. Jan Roest
- Department of Bacteriology and TSEs, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands
| | - Alex Bossers
- Department of Infection Biology, Central Veterinary Institute part of Wageningen UR, Lelystad, The Netherlands
- * E-mail:
| |
Collapse
|
41
|
A screen of Coxiella burnetii mutants reveals important roles for Dot/Icm effectors and host autophagy in vacuole biogenesis. PLoS Pathog 2014; 10:e1004286. [PMID: 25080348 PMCID: PMC4117601 DOI: 10.1371/journal.ppat.1004286] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 06/17/2014] [Indexed: 12/18/2022] Open
Abstract
Coxiella burnetii is an intracellular pathogen that replicates in a lysosome-derived vacuole. The molecular mechanisms used by this bacterium to create a pathogen-occupied vacuole remain largely unknown. Here, we conducted a visual screen on an arrayed library of C. burnetii NMII transposon insertion mutants to identify genes required for biogenesis of a mature Coxiella-containing vacuole (CCV). Mutants defective in Dot/Icm secretion system function or the PmrAB regulatory system were incapable of intracellular replication. Several mutants with intracellular growth defects were found to have insertions in genes encoding effector proteins translocated into host cells by the Dot/Icm system. These included mutants deficient in the effector proteins Cig57, CoxCC8 and Cbu1754. Mutants that had transposon insertions in genes important in central metabolism or encoding tRNA modification enzymes were identified based on the appearance filamentous bacteria intracellularly. Lastly, mutants that displayed a multi-vacuolar phenotype were identified. All of these mutants had a transposon insertion in the gene encoding the effector protein Cig2. Whereas vacuoles containing wild type C. burnetii displayed robust accumulation of the autophagosome protein LC3, the vacuoles formed by the cig2 mutant did not contain detectible amounts of LC3. Furthermore, interfering with host autophagy during infection by wild type C. burnetii resulted in a multi-vacuolar phenotype similar to that displayed by the cig2 mutant. Thus, a functional Cig2 protein is important for interactions between the CCV and host autophagosomes and this drives a process that enhances the fusogenic properties of this pathogen-occupied organelle. Coxiella burnetii is the causative agent of the human disease Q fever. This bacterium uses the Dot/Icm type IV secretion system to deliver effectors into the cytosol of host cells. The Dot/Icm system is required for intracellular replication of C. burnetii. To determine the contribution of individual proteins to the establishment of a vacuole that supports C. burnetii replication, we conducted a visual screen on a library of C. burnetii transposon insertion mutants and identified genes required for distinct stages of intracellular replication. This approach was validated through the identification of intracellular replication mutants that included insertions in most of the dot and icm genes, and through the identification of individual effector proteins delivered into host cell by the Dot/Icm system that participate in creating a vacuole that supports intracellular replication of C. burnetii. Complementation studies showed convincingly that the effector Cig57 was critical for intracellular replication. The effector protein Cig2 was found to play a unique role in promoting homotypic fusion of C. burnetii vacuoles. Disrupting host autophagy phenocopied the defect displayed by the cig2 mutant. Thus, our visual screen has successfully identified effectors required for intracellular replication of C. burnetii and indicates that Dot/Icm-dependent subversion of host autophagy promotes homotypic fusion of CCVs.
Collapse
|
42
|
Galleria mellonella as an alternative model of Coxiella burnetii infection. Microbiology (Reading) 2014; 160:1175-1181. [DOI: 10.1099/mic.0.077230-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Coxiella burnetii is a Gram-negative intracellular bacterium and is the causative agent of the zoonotic disease Q fever. Several rodent and non-human primate models of virulent phase I C. burnetii [Nine Mile (NM)I] have been developed, and have been used to determine the efficacy of antibiotics and vaccine candidates. However, there are several advantages to using insect models to study host–microbe interactions, such as reduced animal use, lowered cost and ease of manipulation in high containment. In addition, many laboratories use the avirulent phase II C. burnetii clone (NMII) to study cellular interactions and identify novel virulence determinants using genetic manipulation. We report that larvae of the greater wax moth, Galleria mellonella, were susceptible to infection with both C. burnetii NMI and NMII. Following subcutaneous infection, we report that intracellular bacteria were present within haemocytes and that larval death occurred in a dose-dependent manner. Additionally, we have used the model to characterize the role of the type 4 secretion system in C. burnetii NMII and to determine antibiotic efficacy in a non-mammalian model of disease.
Collapse
|
43
|
Roest HIJ, Bossers A, van Zijderveld FG, Rebel JML. Clinical microbiology of Coxiella burnetii and relevant aspects for the diagnosis and control of the zoonotic disease Q fever. Vet Q 2013; 33:148-60. [PMID: 24161079 DOI: 10.1080/01652176.2013.843809] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Coxiella burnetii is the causative agent of the zoonotic disease Q fever. Since its first recognition as a disease in the 1930s, the knowledge about the agent and the disease itself has increased. This review summarizes the current knowledge on C. burnetii and Q fever, its pathogenesis, diagnosis and control. C. burnetii is a bacterium which naturally replicates inside human or animal host cells. The clinical presentation of Q fever varies per host species. C. burnetii infection in animals is mainly asymptomatic except for pregnant ruminants in which abortions and stillbirth can occur. In humans, the disease is also mainly asymptomatic, but clinical presentations include acute and chronic Q fever and the post-Q fever fatigue syndrome. Knowledge of the pathogenesis of Q fever in animals and excretion of C. burnetii in infected animals is crucial in understanding the transmission routes and risks of human infection. Our studies indicated that infected pregnant animals only excrete C. burnetii during and after parturition, independent of abortion, and that C. burnetii phase specific serology can be a useful tool in the early detection of infection. Domestic ruminants are the main reservoir for human Q fever, which has a major public health impact when outbreaks occur. In outbreaks, epidemiological source identification can only be refined by genotypic analysis of the strains involved. To control outbreaks and Q fever in domestic ruminants, vaccination with a phase 1 vaccine is effective. Future challenges are to identify factors for virulence, host susceptibility and protection.
Collapse
Affiliation(s)
- Hendrik I J Roest
- a Department of Bacteriology and TSEs, Central Veterinary Institute , Wageningen University and Research Centre , Lelystad , the Netherlands
| | | | | | | |
Collapse
|
44
|
Islam A, Lockhart M, Stenos J, Graves S. The attenuated nine mile phase II clone 4/RSA439 strain of Coxiella burnetii is highly virulent for severe combined immunodeficient (SCID) mice. Am J Trop Med Hyg 2013; 89:800-3. [PMID: 23958905 DOI: 10.4269/ajtmh.12-0653] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The Nine Mile phase II clone 4 (NMIIC4) strain of Coxiella burnetii is an attenuated phase II strain that has lost the genes for virulence determinant type 1 lipopolysaccharide. These bacteria were very virulent for severe combined immunodeficient (SCID) mice. The lethal dose 50 (LD50) was ~10 bacteria. Infected SCID mice died between Day 28 and Day 53 post-infection. At termination of the experiment (Day 60) only 5 of 24 mice had survived. The degree of splenomegaly was directly related to the bacterial load in the SCID mice spleens. The NMIIC4 was avirulent in immunocompetent wild mice and bacterial DNA copies in splenic tissue were extremely low. The SCID mice that were inoculated with high doses of heat inactivated NMIIC4 C. burnetii were all alive at Day 60 and without splenomegaly. It appears that the phase I lipopolysaccharide present in virulent Nine Mile phase I but not in attenuated NMIIC4 is not the only virulence factor for C. burnetii.
Collapse
Affiliation(s)
- Aminul Islam
- Department of Microbiology, Pathology North-Hunter, NSW Health Pathology, John Hunter Hospital, Locked Bag 1, HRMC, Australia; Australian Rickettsial Reference Laboratory, Barwon Health, Geelong, Australia
| | | | | | | |
Collapse
|
45
|
Hendrix LR, Chen C. Antigenic analysis for vaccines and diagnostics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 984:299-328. [PMID: 22711639 DOI: 10.1007/978-94-007-4315-1_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Coxiella burnetii infection is frequently unrecognized or misdiagnosed, as symptoms generally mimic an influenza-like illness. However, the disease (Q fever) may result in chronic infection, usually manifesting as potentially fatal endocarditis. The development of a chronic fatigue-like sequela may also occur. Infected ruminants are the major reservoir for infection in humans, primarily through exposure to birth products or aerosols that transmit the bacterium over wide regions. A vaccine against C. burnetii infection has been in use in Australia for abattoir and agricultural workers for many years. The possibility of adverse reactions in those with previous exposure to the agent has prevented its use elsewhere. Subunit vaccines, utilizing chemical extraction of components thought to cause adverse reactions, are in development, but none are yet licensed. Others have sought to combine immunogenic peptides with or without selected lipopolysaccharide components to produce a vaccine without the possibility of adverse reactions. Selected immunogenic proteins have been shown to induce both humoral and cellular immune responses. Although current diagnosis of infection relies on serological testing, the presentation of specific antibody occurs 7-15 days following the onset of symptoms, delaying treatment that may result in prolonged morbidity. PCR detection of DNA to specific C. burnetii antigens in the blood is possible early in infection, but PCR may become negative when PII IgG antibodies appear. PCR is useful for early diagnosis when Q fever is suspected, as in large epidemics, and shortens the delay in the identification of Q fever endocarditis. Others have combined PCR with ELISA or other methods to increase the ability to detect infection at any stage. The search for new diagnostic reagents and vaccines has utilized new methods for discovery of immunoreactive proteins. DNA analysis of the heterogeneity of C. burnetii isolates has led to a greater understanding of the diversity of isolates and a means to determine whether there is a correlation between strain and disease severity. 2-D SDS PAGE of immunogenic proteins reactive with human or animal infection sera and mass spectrometric analysis of specific secreted or outer membrane proteins have identified candidate antigens. Microarrays have allowed the analysis of peptide libraries of open reading frames to evaluate the immunogenicity of complete genomes.
Collapse
Affiliation(s)
- Laura R Hendrix
- Department of Microbial and Molecular Pathogenesis, College of Medicine, Texas A&M Health Science Center, 3107 Medical Research and Education Building, Bryan, TX 77807-3260, USA.
| | | |
Collapse
|
46
|
Flores-Ramirez G, Janecek S, Miernyk JA, Skultety L. In silico biosynthesis of virenose, a methylated deoxy-sugar unique to Coxiella burnetii lipopolysaccharide. Proteome Sci 2012; 10:67. [PMID: 23150954 PMCID: PMC3539893 DOI: 10.1186/1477-5956-10-67] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 11/05/2012] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED BACKGROUND Coxiella burnetii is Gram-negative bacterium responsible for the zoonosis Q-fever. While it has an obligate intracellular growth habit, it is able to persist for extended periods outside of a host cell and can resist environmental conditions that would be lethal to most prokaryotes. It is these extracellular bacteria that are the infectious stage encountered by eukaryotic hosts. The intracellular form has evolved to grow and replicate within acidified parasitophorous vacuoles. The outer coat of C. burnetii comprises a complex lipopolysaccharide (LPS) component that includes the unique methylated-6-deoxyhexose, virenose. Although potentially important as a biomarker for C. burnetii, the pathway for its biosynthesis remains obscure. RESULTS The 6-deoxyhexoses constitute a large family integral to the LPS of many eubacteria. It is believed that precursors of the methylated-deoxyhexoses traverse common early biosynthetic steps as nucleotide-monosaccharides. As a prelude to a full biosynthetic characterization, we present herein the results from bioinformatics-based, proteomics-supported predictions of the pathway for virenose synthesis. Alternative possibilities are considered which include both GDP-mannose and TDP-glucose as precursors. CONCLUSION We propose that biosynthesis of the unique C. burnetii biomarker, virenose, involves an early pathway similar to that of other C-3'-methylated deoxysugars which then diverges depending upon the nucleotide-carrier involved. The alternatives yield either the D- or L-enantiomers of virenose. Both pathways require five enzymatic steps, beginning with either glucose-6-phosphate or mannose-6-phosphate. Our in silico results comprise a model for virenose biosynthesis that can be directly tested. Definition of this pathway should facilitate the development of therapeutic agents useful for treatment of Q fever, as well as allowing improvements in the methods for diagnosing this highly infectious disease.
Collapse
Affiliation(s)
- Gabriela Flores-Ramirez
- Department of Rickettsiology, Institute of Virology, Slovak Academy of Sciences, Dubravska cesta, 9, Bratislava, 845 05, Slovakia
| | - Stefan Janecek
- Laboratory of Protein Evolution, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, 845 51, Slovakia
| | - Ján A Miernyk
- USDA, Agricultural Research Service, Plant Genetics Research Unit, Columbia, MO, 65211, USA
- Interdisciplinary Plant Group, University of Missouri, Columbia, MO, 65211, USA
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Ludovit Skultety
- Department of Rickettsiology, Institute of Virology, Slovak Academy of Sciences, Dubravska cesta, 9, Bratislava, 845 05, Slovakia
- Centre for Molecular Medicine, Slovak Academy of Sciences, Bratislava, 831 01, Slovakia
| |
Collapse
|
47
|
Ihnatko R, Shaw E, Toman R. Proteome of Coxiella burnetii. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 984:105-30. [DOI: 10.1007/978-94-007-4315-1_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
48
|
van Schaik EJ, Samuel JE. Phylogenetic diversity, virulence and comparative genomics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 984:13-38. [PMID: 22711625 DOI: 10.1007/978-94-007-4315-1_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Coxiella burnetii, the causative agent of Q fever, has remained a public health concern since the identification of this organism in 1935 by E. H. Derrick in Australia and at the Rocky Mountain Laboratory in the USA by H.R. Cox and G. Davis. Human Q fever has been described in most countries where C. burnetii is ubiquitous in the environment except in New Zealand where no cases have been described. Most human infections are acquired through inhalation of contaminated aerosols that can lead to acute self-limiting febrile illness or more severe chronic cases of hepatitis or endocarditis. It is estimated that the actual incidence of human infection is under-reported as a result of imprecise tools for differential diagnosis. An intracellular lifestyle, low infectious dose, and ease of transmission have resulted in the classification of C. burnetii as a category B bio-warfare agent. The recent outbreaks in Europe are a reminder that there is much to learn about this unique intracellular pathogen, especially with the speculation of a hyper-virulent strain contributing to an outbreak in the Netherlands where over 4,000 human cases were reported. A new era in C. burnetii research has begun with the recent description of an axenic media making this an exciting time to study this bacterial pathogen.
Collapse
Affiliation(s)
- Erin J van Schaik
- Department of Microbial and Molecular Pathogenesis, College of Medicine, Texas A&M Health Science Center, 3112 Medical Research and Education Building, Bryan, TX, 77807-3260, USA
| | | |
Collapse
|
49
|
Narasaki CT, Toman R. Lipopolysaccharide of Coxiella burnetii. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 984:65-90. [PMID: 22711627 DOI: 10.1007/978-94-007-4315-1_4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A lipopolysaccharide (LPS) is considered to be one of the major determinants of virulence expression and infection of virulent Coxiella burnetii. The LPSs from virulent phase I (LPS I) and from avirulent phase II (LPS II) bacteria were investigated for their chemical composition, structure and biological properties. LPS II is of rough (R) type in contrast to LPS I, which is phenotypically smooth (S) and contains a noticeable amount of two sugars virenose (Vir) and dihydrohydroxystreptose (Strep), which have not been found in other LPSs and can be considered as unique biomarkers of the bacterium. Both sugars were suggested to be located mostly in terminal positions of the O-specific chain of LPS I (O-PS I) and to be involved in the immunobiology of Q fever. There is a need to establish a more detailed chemical structure of LPS I in connection with prospective, deeper studies on mechanisms of pathogenesis and immunity of Q fever, its early and reliable diagnosis, and effective prophylaxis against the disease. This will also help to better understanding of host-pathogen interactions and contribute to improved modulation of pathological reactions which in turn are prerequisite for research and development of vaccines of new type. A fundamental understanding of C. burnetii LPS biosynthesis is still lacking. The intracellular nature of the bacterium, lack of genetic tools and its status as a selected agent have made elucidating basic physiological mechanisms challenging. The GDP-β-D-Vir biosynthetic pathway proposed most recently is an important initial step in this endeavour. The current advanced technologies providing the genetic tools necessary to screen C. burnetii mutants and propagate isogenic mutants might speed the discovery process.
Collapse
Affiliation(s)
- Craig T Narasaki
- Center Department of Microbial and Molecular Pathogenesis, Texas A&M University Health Science, College Station, TX 77843, USA
| | | |
Collapse
|
50
|
Delsing CE, Warris A, Bleeker-Rovers CP. Q Fever: Still More Queries than Answers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 719:133-43. [DOI: 10.1007/978-1-4614-0204-6_12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|