1
|
Böttcher J, Alex M, Dänicke S, Gethmann J, Mertens-Scholz K, Janowetz B. Susceptibility, Immunity, and Persistent Infection Drive Endemic Cycles of Coxiellosis on Dairy Farms. Animals (Basel) 2024; 14:1056. [PMID: 38612295 PMCID: PMC11011148 DOI: 10.3390/ani14071056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Coxiella (C.) burnetii, a zoonotic bacterium, is prevalent in dairy farms. Some cows develop a persistent infection and shed C. burnetii into milk and occasionally by amniotic fluid at calving. Serological diagnosis of Q fever in humans is performed by phase (Ph)-specific antibody tests; PhII antibodies usually indicate an acute infection, while the development of a chronic infection is characterised by elevated PhI antibody titres. Phase-specific tests have now been established for diagnosis of coxiellosis in cattle. Additionally, an interferon-γ (IFN-γ) recall assay has been implemented to assess cellular immunity to C. burnetii in cattle. Milk samples from all lactating cows (n = 2718) of 49 Bavarian dairy farms were collected through a convenience sample and analysed for phase-specific antibodies. Antibody profiles were evaluated by age. Based on the seropositivity of first-lactation cows, three distinct herd profiles were observed: an 'acute' state of herd infection was characterised by a PhI-/PhII+ pattern. The detection of PhI antibodies (PhI+/PhII+) characterised the 'chronic' state, and seronegative results defined the 'silent' state of herd infection. If antibodies had not been detected in multiparous cows, the herd was considered as probably free of coxiellosis. The analysed cattle herds were noted to have an 'acute' (n = 12, 24.5%), 'chronic' (n = 18, 36.8%), or 'silent' state of herd infection (n = 16, 32.6%). Only three farms (6.1%) were classified as 'free' of C. burnetii. The detection of these herd states over a time period of 4 years in one farm indicated that the described states occur in a cyclical manner. Frequently, a wave-like profile was seen, i.e., a circumscribed seronegative age group was flanked by seropositive age groups. In seronegative animals, IFN-γ reactivity was demonstrated. Seroconversion after vaccination was observed by day 7 post-vaccination in chronically infected herds, whereas in the case of silent infection, it started by day 14. These data indicated a pre-existing immunity in seronegative animals in chronically infected herds. Additionally, IFN-γ reactivity was detected in seronegative calves (>3 months) and heifers from chronically infected farms compared to a negative farm. An infection prior to 3 months of age resulted in cellular immunity in the absence of detectable antibodies. An infection around calving would explain this. The aforementioned circumscribed seronegative age groups are, therefore, explained by an infection early in life during active shedding at calving. Based on these results, an endemic cycle of coxiellosis is proposed: Susceptible young heifers get infected by persistently infected cows. Subsequently, shedding of C. burnetii at calving results in infection and then in cellular immunity in offspring. When these calves enter the cow herd two years later, a maximum of herd immunity is achieved, shedding ceases, and new susceptible animals are raised. In an acutely infected dairy farm, the PhI+/PhII+ serological pattern prevailed in second-lactation cows. In this study, stored sera collected since birth were analysed retrospectively. From the earliest seroconversion, the peak of seroconversion took about 33 months. These data suggested a slow spread of infection within herds. The classification of dairy cow herds is a promising basis for further analysis of the clinical impact of coxiellosis.
Collapse
Affiliation(s)
- Jens Böttcher
- Bavarian Animal Health Service, Senator-Gerauer-Straße 23, D-85586 Poing, Germany; (M.A.); (B.J.)
| | - Michaela Alex
- Bavarian Animal Health Service, Senator-Gerauer-Straße 23, D-85586 Poing, Germany; (M.A.); (B.J.)
| | - Sven Dänicke
- Institute of Animal Nutrition, Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, Bundesallee 37, D-38116 Braunschweig, Germany;
| | - Jörn Gethmann
- Institute of Epidemiology, Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, Südufer 10, D-17493 Greifswald-Insel Riems, Germany;
| | - Katja Mertens-Scholz
- Institute for Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, Naumburger Straße 96a, D-07743 Jena, Germany;
- Institute for Infectious Diseases and Infection Control and Center for Sepsis Care and Control (CSCC), Jena University Hospital, Am Klinikum 1, D-07745 Jena, Germany
| | - Britta Janowetz
- Bavarian Animal Health Service, Senator-Gerauer-Straße 23, D-85586 Poing, Germany; (M.A.); (B.J.)
| |
Collapse
|
2
|
Jan S, Fratzke AP, Felgner J, Hernandez-Davies JE, Liang L, Nakajima R, Jasinskas A, Supnet M, Jain A, Felgner PL, Davies DH, Gregory AE. Multivalent vaccines demonstrate immunogenicity and protect against Coxiella burnetii aerosol challenge. Front Immunol 2023; 14:1192821. [PMID: 37533862 PMCID: PMC10390735 DOI: 10.3389/fimmu.2023.1192821] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/16/2023] [Indexed: 08/04/2023] Open
Abstract
Vaccines are among the most cost-effective public health measures for controlling infectious diseases. Coxiella burnetii is the etiological agent of Q fever, a disease with a wide clinical spectrum that ranges from mild symptoms, such as fever and fatigue, to more severe disease, such as pneumonia and endocarditis. The formalin-inactivated whole-cell vaccine Q-VAX® contains hundreds of antigens and confers lifelong protection in humans, but prior sensitization from infection or vaccination can result in deleterious reactogenic responses to vaccination. Consequently, there is great interest in developing non-reactogenic alternatives based on adjuvanted recombinant proteins. In this study, we aimed to develop a multivalent vaccine that conferred protection with reduced reactogenicity. We hypothesized that a multivalent vaccine consisting of multiple antigens would be more immunogenic and protective than a monovalent vaccine owing to the large number of potential protective antigens in the C. burnetii proteome. To address this, we identified immunogenic T and B cell antigens, and selected proteins were purified to evaluate with a combination adjuvant (IVAX-1), with or without C. burnetii lipopolysaccharide (LPS) in immunogenicity studies in vivo in mice and in a Hartley guinea pig intratracheal aerosol challenge model using C. burnetii strain NMI RSA 493. The data showed that multivalent vaccines are more immunogenic than monovalent vaccines and more closely emulate the protection achieved by Q-VAX. Although six antigens were the most immunogenic, we also discovered that multiplexing beyond four antigens introduces detectable reactogenicity, indicating that there is an upper limit to the number of antigens that can be safely included in a multivalent Q-fever vaccine. C. burnetii LPS also demonstrates efficacy as a vaccine antigen in conferring protection in an otherwise monovalent vaccine formulation, suggesting that its addition in multivalent vaccines, as demonstrated by a quadrivalent formulation, would improve protective responses.
Collapse
Affiliation(s)
- Sharon Jan
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Alycia P. Fratzke
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, United States
- Department of Pathology, Charles River Laboratories, Reno, NV, United States
| | - Jiin Felgner
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Jenny E. Hernandez-Davies
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Li Liang
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Rie Nakajima
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Algimantas Jasinskas
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Medalyn Supnet
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Aarti Jain
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Philip L. Felgner
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - D. Huw Davies
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Anthony E. Gregory
- Vaccine Research & Development Center, Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
3
|
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: 8] [Impact Index Per Article: 4.0] [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
|
4
|
Raju Paul S, Scholzen A, Mukhtar G, Wilkinson S, Hobson P, Dzeng RK, Evans J, Robson J, Cobbold R, Graves S, Poznansky MC, Garritsen A, Sluder AE. Natural Exposure- and Vaccination-Induced Profiles of Ex Vivo Whole Blood Cytokine Responses to Coxiella burnetii. Front Immunol 2022; 13:886698. [PMID: 35812430 PMCID: PMC9259895 DOI: 10.3389/fimmu.2022.886698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Q fever is a zoonotic disease caused by the highly infectious Gram-negative coccobacillus, Coxiella burnetii (C. burnetii). The Q fever vaccine Q-VAX® is characterised by high reactogenicity, requiring individuals to be pre-screened for prior exposure before vaccination. To date it remains unclear whether vaccine side effects in pre-exposed individuals are associated with pre-existing adaptive immune responses to C. burnetii or are also a function of innate responses to Q-VAX®. In the current study, we measured innate and adaptive cytokine responses to C. burnetii and compared these among individuals with different pre-exposure status. Three groups were included: n=98 Dutch blood bank donors with unknown exposure status, n=95 Dutch village inhabitants with known natural exposure status to C. burnetii during the Dutch Q fever outbreak of 2007-2010, and n=96 Australian students receiving Q-VAX® vaccination in 2021. Whole blood cytokine responses following ex vivo stimulation with heat-killed C. burnetii were assessed for IFNγ, IL-2, IL-6, IL-10, TNFα, IL-1β, IP-10, MIP-1α and IL-8. Serological data were collected for all three cohorts, as well as data on skin test and self-reported vaccine side effects and clinical symptoms during past infection. IFNγ, IP-10 and IL-2 responses were strongly elevated in individuals with prior C. burnetii antigen exposure, whether through infection or vaccination, while IL-1β, IL-6 and TNFα responses were slightly increased in naturally exposed individuals only. High dimensional analysis of the cytokine data identified four clusters of individuals with distinct cytokine response signatures. The cluster with the highest levels of adaptive cytokines and antibodies comprised solely individuals with prior exposure to C. burnetii, while another cluster was characterized by high innate cytokine production and an absence of C. burnetii-induced IP-10 production paired with high baseline IP-10 levels. Prior exposure status was partially associated with these signatures, but could not be clearly assigned to a single cytokine response signature. Overall, Q-VAX® vaccination and natural C. burnetii infection were associated with comparable cytokine response signatures, largely driven by adaptive cytokine responses. Neither individual innate and adaptive cytokine responses nor response signatures were associated retrospectively with clinical symptoms during infection or prospectively with side effects post-vaccination.
Collapse
Affiliation(s)
- Susan Raju Paul
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| | | | - Ghazel Mukhtar
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| | | | - Peter Hobson
- Sullivan Nicolaides Pathology, Brisbane, QLD, Australia
| | - Richard K. Dzeng
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| | | | | | - Rowland Cobbold
- School of Veterinary Science, University of Queensland, Gatton, QLD, Australia
| | - Stephen Graves
- Australian Rickettsial Reference Laboratory, Geelong, VIC, Australia
| | - Mark C. Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
- *Correspondence: Ann E. Sluder, ; Anja Garritsen, ; Mark C. Poznansky,
| | - Anja Garritsen
- InnatOss Laboratories B.V., Oss, Netherlands
- *Correspondence: Ann E. Sluder, ; Anja Garritsen, ; Mark C. Poznansky,
| | - Ann E. Sluder
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
- *Correspondence: Ann E. Sluder, ; Anja Garritsen, ; Mark C. Poznansky,
| |
Collapse
|
5
|
Sireci G, Badami GD, Di Liberto D, Blanda V, Grippi F, Di Paola L, Guercio A, de la Fuente J, Torina A. Recent Advances on the Innate Immune Response to Coxiella burnetii. Front Cell Infect Microbiol 2021; 11:754455. [PMID: 34796128 PMCID: PMC8593175 DOI: 10.3389/fcimb.2021.754455] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Coxiella burnetii is an obligate intracellular Gram-negative bacterium and the causative agent of a worldwide zoonosis known as Q fever. The pathogen invades monocytes and macrophages, replicating within acidic phagolysosomes and evading host defenses through different immune evasion strategies that are mainly associated with the structure of its lipopolysaccharide. The main transmission routes are aerosols and ingestion of fomites from infected animals. The innate immune system provides the first host defense against the microorganism, and it is crucial to direct the infection towards a self-limiting respiratory disease or the chronic form. This review reports the advances in understanding the mechanisms of innate immunity acting during C. burnetii infection and the strategies that pathogen put in place to infect the host cells and to modify the expression of specific host cell genes in order to subvert cellular processes. The mechanisms through which different cell types with different genetic backgrounds are differently susceptible to C. burnetii intracellular growth are discussed. The subsets of cytokines induced following C. burnetii infection as well as the pathogen influence on an inflammasome-mediated response are also described. Finally, we discuss the use of animal experimental systems for studying the innate immune response against C. burnetii and discovering novel methods for prevention and treatment of disease in humans and livestock.
Collapse
Affiliation(s)
- Guido Sireci
- Central Laboratory of Advanced Diagnostic and Biological Research (CLADIBIOR), Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University Hospital "Paolo Giaccone", Università degli studi di Palermo, Palermo, Italy
| | - Giusto Davide Badami
- Central Laboratory of Advanced Diagnostic and Biological Research (CLADIBIOR), Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University Hospital "Paolo Giaccone", Università degli studi di Palermo, Palermo, Italy
| | - Diana Di Liberto
- Central Laboratory of Advanced Diagnostic and Biological Research (CLADIBIOR), Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University Hospital "Paolo Giaccone", Università degli studi di Palermo, Palermo, Italy
| | - Valeria Blanda
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - Francesca Grippi
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - Laura Di Paola
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - Annalisa Guercio
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - José de la Fuente
- SaBio Health and Biotechnology, Instituto de Investigación en Recursos Cinegéticos, IREC -Spanish National Research Council CSIC - University of Castilla-La Mancha UCLM - Regional Government of Castilla-La Mancha JCCM, Ciudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, United States
| | | |
Collapse
|
6
|
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: 2.0] [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
|
7
|
From Coxiella burnetii Infection to Pregnancy Complications: Key Role of the Immune Response of Placental Cells. Pathogens 2021; 10:pathogens10050627. [PMID: 34069587 PMCID: PMC8160966 DOI: 10.3390/pathogens10050627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 01/17/2023] Open
Abstract
The infection of pregnant animals and women by Coxiella burnetii, an intracellular bacterium, compromises both maternal health and foetal development. The placenta is targeted by C. burnetii, as demonstrated by bacteriological and histological evidence. It now appears that placental strains of C. burnetii are highly virulent compared to reference strains and that placental injury involves different types of placental cells. Trophoblasts, the major placental cells, are largely infected by C. burnetii and may represent a replicating niche for the bacteria. The placenta also contains numerous immune cells, including macrophages, dendritic cells, and mast cells. Placental macrophages are infected and activated by C. burnetii in an unusual way of M1 polarisation associated with bacterial elimination. Placental mast cells eliminate bacteria through a mechanism including the release of extracellular actin filaments and antimicrobial peptides. In contrast, C. burnetii impairs the maturation of decidual dendritic cells, favouring bacterial pathogenicity. Our aim is to review C. burnetii infections of human placentas, paying special attention to both the action and function of the different cell types, immune cells, and trophoblasts targeted by C. burnetii in relation to foetal injury.
Collapse
|
8
|
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: 20] [Impact Index Per Article: 6.7] [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
|
9
|
Shanaka KASN, Tharuka MDN, Sellaththurai S, Yang H, Priyathilaka TT, Lee J. Characterization and expression analysis of rockfish (Sebastes schlegelii) myeloid differentiation factor-88 (SsMyD88) and evaluation of its ability to induce inflammatory cytokines through NF-ĸB. FISH & SHELLFISH IMMUNOLOGY 2020; 99:59-72. [PMID: 32006686 DOI: 10.1016/j.fsi.2020.01.060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/07/2020] [Accepted: 01/28/2020] [Indexed: 06/10/2023]
Abstract
Innate immunity is characterized by nonspecific, prompt reactions toward armada of antigens. Animals funnel down a repertoire of immune stimulants to activate non-selective defense mechanisms rapidly. This study was conducted to characterize the rockfish (Sebastes schlegelii) adaptor protein MyD88 (SsMyD88), which interacts with both toll-like receptors and interleukin receptors. The tissue expression of unchallenged SsMyD88 was evaluated by quantitative real time PCR (qPCR). Fish were intraperitoneally injected with immune stimulants including poly I:C, lipopolysaccharides, and Streptococcus iniae. Then, the temporal expression of SsMyD88 was analyzed. Finally, the inflammatory gene expression and downstream promoter activation were analyzed. Strongest expressions were reported in the liver, gills and spleen in unchallenged conditions. All diverse immune stimulants were found to be capable of significantly altering SsMyD88 transcription during the challenge experiment. Evaluation of downstream promoter biases by SsMyD88 found a predominant activation of NF-ĸB transcription factors when compared with the AP-1, revealing significant and substantial upregulation of major inflammatory mediators such as IL-1-β, IL-6, iNOS, COX-2 and TNF-α. Fluorescent detection confirmed an intense production of NO and the predominant differentiation of macrophages into M1 lineage with the overexpression of SsMyD88 in vitro. These results further corroborate the role of SsMyD88 as a mediatory molecule that bridges distinct immune stimulants to induce drastic immune responses in fish.
Collapse
Affiliation(s)
- K A S N Shanaka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - M D Neranjan Tharuka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Sarithaa Sellaththurai
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Hyerim Yang
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Thanthrige Thiunuwan Priyathilaka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea.
| |
Collapse
|
10
|
Robison A, Snyder DT, Christensen K, Kimmel E, Hajjar AM, Jutila MA, Hedges JF. Expression of human TLR4/myeloid differentiation factor 2 directs an early innate immune response associated with modest increases in bacterial burden during Coxiella burnetii infection. Innate Immun 2019; 25:401-411. [PMID: 31180798 PMCID: PMC6900644 DOI: 10.1177/1753425919855420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/24/2019] [Accepted: 05/13/2019] [Indexed: 01/25/2023] Open
Abstract
Human TLR4 (hTLR4) and mouse TLR4 molecules respond differently to hypo-acylated LPS. The LPS of Coxiella burnetii is hypo-acylated and heavily glycosylated and causes a minimal response by human cells. Thus, we hypothesized that mice expressing hTLR4 molecules would be more susceptible to C. burnetii infection. Our results show that transgenic mice expressing hTLR4 and the human myeloid differentiation factor 2 (MD-2) adaptor protein (hTLR4/MD-2) respond similarly to wild type mice with respect to overall disease course. However, differences in bacterial burdens in tissues were noted, and lung pathology was increased in hTLR4/MD2 compared to wild type mice. Surprisingly, bone marrow chimera experiments indicated that hTLR4/MD-2 expression on non-hematopoietic cells, rather than the target cells for C. burnetii infection, accounted for increased bacterial burden. Early during infection, cytokines involved in myeloid cell recruitment were detected in the plasma of hTLR4/MD2 mice but not wild type mice. This restricted cytokine response was accompanied by neutrophil recruitment to the lung in hTLR4/MD2 mice. These data suggest that hTLR4/MD-2 alters early responses during C. burnetii infection. These early responses are precursors to later increased bacterial burdens and exacerbated pathology in the lung. Our data suggest an unexpected role for hTLR4/MD-2 in non-hematopoietic cells during C. burnetii infection.
Collapse
Affiliation(s)
- Amanda Robison
- Department of Microbiology and Immunology, Montana State University, USA
| | - Deann T Snyder
- Department of Microbiology and Immunology, Montana State University, USA
| | - Kelly Christensen
- Department of Microbiology and Immunology, Montana State University, USA
| | - Emily Kimmel
- Department of Microbiology and Immunology, Montana State University, USA
| | - Adeline M Hajjar
- Department of Comparative Medicine, School of Medicine, University of Washington, USA
| | - Mark A Jutila
- Department of Microbiology and Immunology, Montana State University, USA
| | - Jodi F Hedges
- Department of Microbiology and Immunology, Montana State University, USA
| |
Collapse
|
11
|
Lee WY. "Hairiness" is a Facsimile of Reorganized Cytoskeletons: A Cytopathic Effect of Coxiella burnetii. Yonsei Med J 2019; 60:890-897. [PMID: 31538423 PMCID: PMC6753337 DOI: 10.3349/ymj.2019.60.10.890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 01/14/2023] Open
Abstract
In 1993, I reported that Coxiella burnetii transforms human B cells into hairy cells (cbHCs), the first hairy cell reported outside of hairy cell leukemia (HCL). Over last few decades, advances in molecular biology have provided evidence supporting that C. burnetii induces hairiness and inhibits the apoptosis of host cells. The present review summarizes new information in support of cbHC. C. burnetii was shown to induce reorganization of the cytoskeleton and to inhibit apoptosis in host cells. Peritoneal B1a cells were found to be permissive for virulent C. burnetii Nine Mile phase I (NMI) strains in mice. C. burnetii severely impaired E-cad expression in circulating cells of Q fever patients. B-cell non-Hodgkin lymphoma was linked to C. burnetii. Mutation of BRAF V600E was pronounced in HCL, but "hairiness" was not linked to the mutation. Risk factors shared among coxiellosis and HCL in humans and animals were reported in patients with Q-fever. Accordingly, I propose that C. burnetii induces reorganization of the cytoskeleton and inhibits apoptosis as cytopathic effects that are not target cell specific. The observed hairiness in cbHC appears to be a fixed image of dynamic nature, and hairy cells in HCL are distinct among lymphoid cells in circulation. As the cytoskeleton plays key roles in maintaining cell structural integrity in health and disease, the pathophysiology of similar cytopathic effects should be addressed in other diseases, such as myopathies, B-cell dyscrasias, and autoimmune syndromes.
Collapse
Affiliation(s)
- Won Young Lee
- Emeritus Professor, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
12
|
Mezouar S, Diarra I, Roudier J, Desnues B, Mege JL. Tumor Necrosis Factor-Alpha Antagonist Interferes With the Formation of Granulomatous Multinucleated Giant Cells: New Insights Into Mycobacterium tuberculosis Infection. Front Immunol 2019; 10:1947. [PMID: 31475008 PMCID: PMC6702871 DOI: 10.3389/fimmu.2019.01947] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/01/2019] [Indexed: 12/21/2022] Open
Abstract
More than half of tuberculosis cases in the world are due to resuscitation of dormant Mycobacterium tuberculosis (Mtb) sequestered into cell-derived structures called granulomas. It is fairly admitted that cytokines and more particularly Tumor Necrosis Factor (TNF)-α is critical in the control of Mtb infections and that anti-TNF-α drugs constitute one of the main risk factors for reactivation of latent Mtb infection. The aim of this study was to evaluate the role of etanercept, a dimeric fusion protein consisting of the extracellular ligand-binding portion of the human p75 TNF receptor linked to the Fc portion of human IgG1, in an in vitro model of human tuberculous granuloma. We showed that etanercept slightly delayed the formation of granuloma and reduced the generation of multinuclear giant cells (MGCs). In addition, etanercept exacerbated the expression of M1 polarization genes but also induced interleukin (IL)-10 release. In addition, our results indicated that etanercept inhibited cell fusion in an IL-10-dependent manner. Moreover, adalimumab, a human monoclonal anti-TNF-α IgG1 inhibited MGC formation in granuloma, without altering IL-10 secretion and induced macrophage apoptosis. Taken together, our data provides new insights into the role of TNF-α blockers in MGCs formation and the impact of such immunomodulatory drugs on tuberculous granuloma maturation.
Collapse
Affiliation(s)
- Soraya Mezouar
- Aix-Marseille Université, IRD, APHM, MEPHI, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| | - Issa Diarra
- Aix-Marseille Université, IRD, APHM, MEPHI, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| | - Jean Roudier
- Department of Rheumatology, Institut du Mouvement et de l'appareil Locomoteur, APHM, Marseille, France
| | - Benoit Desnues
- Aix-Marseille Université, IRD, APHM, MEPHI, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| | - Jean-Louis Mege
- Aix-Marseille Université, IRD, APHM, MEPHI, Marseille, France.,IHU-Méditerranée Infection, Marseille, France.,APHM, IHU Méditerranée Infection, UF Immunologie, Marseille, France
| |
Collapse
|
13
|
Chen C, van Schaik EJ, Gregory AE, Vigil A, Felgner PL, Hendrix LR, Faris R, Samuel JE. Chemokine Receptor 7 Is Essential for Coxiella burnetii Whole-Cell Vaccine-Induced Cellular Immunity but Dispensable for Vaccine-Mediated Protective Immunity. J Infect Dis 2019; 220:624-634. [PMID: 30938819 PMCID: PMC6639598 DOI: 10.1093/infdis/jiz146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 03/27/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Protective immunity against Coxiella burnetii infection is conferred by vaccination with virulent (PI-WCV), but not avirulent (PII-WCV) whole-cell inactivated bacterium. The only well-characterized antigenic difference between virulent and avirulent C. burnetii is they have smooth and rough lipopolysaccharide (LPS), respectively. METHODS Mice were vaccinated with PI-WCV and PII-WCV. Humoral and cellular responses were evaluated using protein chip microarrays and ELISpots, respectively. Dendritic cell (DC) maturation after stimulation with PI-WVC and PII-WVC was evaluated using flow cytometry. Vaccine-challenge studies were performed to validate the importance of the receptor CCR7. RESULTS Other than specific antibody response to PI-LPS, similar antibody profiles were observed but IgG titers were significantly higher after vaccination with PI-WCV. Furthermore, higher frequency of antigen-specific CD4+ T cells was detected in mice immunized with PI-WCV. PI-WCV-stimulated DCs displayed significantly higher levels of CCR7 and migratory ability to secondary lymphoid organs. Challenge-protection studies in wild-type and CCR7-deficient mice confirmed that CCR7 is critical for PI-WCV-induced cellular immunity. CONCLUSIONS PI-WVC stimulates protective immunity to C. burnetii in mice through stimulation of migratory behavior in DCs for protective cellular immunity. Additionally, the humoral immune response to LPS is an important component of protective immunity.
Collapse
Affiliation(s)
- Chen Chen
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College of Medicine, Bryan
| | - Erin J van Schaik
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College of Medicine, Bryan
| | - Anthony E Gregory
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College of Medicine, Bryan
| | - Adam Vigil
- Department of Medicine, Division of Infectious Diseases, University of California, Irvine
| | - Phillip L Felgner
- Department of Medicine, Division of Infectious Diseases, University of California, Irvine
| | - Laura R Hendrix
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College of Medicine, Bryan
| | - Robert Faris
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College of Medicine, Bryan
| | - James E Samuel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College of Medicine, Bryan
| |
Collapse
|
14
|
Abstract
Mast cells (MCs) are found in tissues that are in close contact with external environment, such as skin, lungs, or intestinal mucosa but also in the placenta during pregnancy. If their role in mediating allergic conditions is established, several studies now highlight their importance during infection with extracellular pathogens. This study showed a new and effective antimicrobial mechanism of MCs against Coxiella burnetii, an intracellular bacterium whose infection during pregnancy is associated with abortion, preterm labor, and stillbirth. The data reveal that in response to C. burnetii, MCs release extracellular actin filaments that contain antimicrobial agents and are capable to trap and kill bacteria. We show that this mechanism is dependent on the cooperation of two membrane receptors, CD36 and Toll-like receptor 4, and may occur in the placenta during pregnancy by using ex vivo placental MCs. Overall, this study reports an unexpected role for MCs during infection with intracellular bacteria and suggests that MC response to C. burnetii infection is a protective defense mechanism during pregnancy. Mast cells (MCs) are critical mediators of inflammation; however, their microbicidal activity against invading pathogens remains largely unknown. Here, we describe a nonpreviously reported antibacterial mechanism used by MCs against Coxiella burnetii, the agent of Q fever. We show that C. burnetii interaction with MCs does not result in bacterial uptake but rather induces the formation of extracellular actin filaments named cytonemes. MC cytonemes express cathelicidin and neutrophil elastase and mediate the capture and destruction of entrapped bacteria. We provide evidence that MC cytoneme formation and microbicidal activity are dependent on the cooperation of the scavenger receptor CD36 and Toll-like receptor 4. Taken together, our results suggest that MCs use an extracellular sophisticated mechanism of defense to eliminate intracellular pathogens, such as C. burnetii, before their entry into host cells.
Collapse
|
15
|
Karaś MA, Turska-Szewczuk A, Janczarek M, Szuster-Ciesielska A. Glycoconjugates of Gram-negative bacteria and parasitic protozoa - are they similar in orchestrating the innate immune response? Innate Immun 2019; 25:73-96. [PMID: 30782045 PMCID: PMC6830889 DOI: 10.1177/1753425918821168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/03/2018] [Indexed: 02/06/2023] Open
Abstract
Innate immunity is an evolutionarily ancient form of host defense that serves to limit infection. The invading microorganisms are detected by the innate immune system through germline-encoded PRRs. Different classes of PRRs, including TLRs and cytoplasmic receptors, recognize distinct microbial components known collectively as PAMPs. Ligation of PAMPs with receptors triggers intracellular signaling cascades, activating defense mechanisms. Despite the fact that Gram-negative bacteria and parasitic protozoa are phylogenetically distant organisms, they express glycoconjugates, namely bacterial LPS and protozoan GPI-anchored glycolipids, which share many structural and functional similarities. By activating/deactivating MAPK signaling and NF-κB, these ligands trigger general pro-/anti-inflammatory responses depending on the related patterns. They also use conservative strategies to subvert cell-autonomous defense systems of specialized immune cells. Signals triggered by Gram-negative bacteria and parasitic protozoa can interfere with host homeostasis and, depending on the type of microorganism, lead to hypersensitivity or silencing of the immune response. Activation of professional immune cells, through a ligand which triggers the opposite effect (antagonist versus agonist) appears to be a promising solution to restoring the immune balance.
Collapse
Affiliation(s)
- Magdalena A Karaś
- Department of Genetics and Microbiology, Maria Curie–Skłodowska
University, Lublin, Poland
| | - Anna Turska-Szewczuk
- Department of Genetics and Microbiology, Maria Curie–Skłodowska
University, Lublin, Poland
| | - Monika Janczarek
- Department of Genetics and Microbiology, Maria Curie–Skłodowska
University, Lublin, Poland
| | | |
Collapse
|
16
|
Rodríguez Y, Rojas M, Gershwin ME, Anaya JM. Tick-borne diseases and autoimmunity: A comprehensive review. J Autoimmun 2018; 88:21-42. [DOI: 10.1016/j.jaut.2017.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022]
|
17
|
Ali I, Manzoor Z, Koo JE, Kim JE, Byeon SH, Yoo ES, Kang HK, Hyun JW, Lee NH, Koh YS. 3-Hydroxy-4,7-megastigmadien-9-one, isolated from Ulva pertusa, attenuates TLR9-mediated inflammatory response by down-regulating mitogen-activated protein kinase and NF-κB pathways. PHARMACEUTICAL BIOLOGY 2017; 55:435-440. [PMID: 27937044 PMCID: PMC6130523 DOI: 10.1080/13880209.2016.1246574] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
CONTEXT Seaweeds are rich in bioactive compounds in the form of vitamins, phycobilins, polyphenols, carotenoids, phycocyanins and polysaccharides; many of these are known to have advantageous applications in human health. 3-Hydroxy-4,7-megastigmadien-9-one (comp) was isolated from Ulva pertusa (U. pertusa) Kjellman (Ulvaceae), which is a familiar edible green seaweed. OBJECTIVE This study evaluates the anti-inflammatory activity of comp in CpG DNA-stimulated bone marrow-derived dendritic cells (BMDCs). MATERIALS AND METHODS For evaluating the effect of comp on cytokines production, BMDCs were treated with doses of comp (0, 0.5, 1, 2, 5, 10, 25 and 50 μM) for 1 h before stimulation with CpG DNA (1 μM). Cytokine production was measured by ELISA. Western blotting was conducted for evaluating effect of comp (50 μM) on MAPKs and NF-κB pathways. Luciferase reporter gene assay was conducted for effect of comp (0, 5, 10 and 25 μM) on transcriptional activity of AP-1 and NF-κB. RESULTS Comp exhibited strong inhibition of interleukin (IL)-12 p40, IL-6 and TNF-α cytokine production with IC50 values of 6.02 ± 0.35, 27.14 ± 0.73, and 7.56 ± 0.21 μM, respectively. It blocked MAPKs and NF-κB pathways by inhibiting the phosphorylation of ERK1/2, JNK1/2, p38 and IκBα. In addition, it strongly inhibited the transcriptional activity of AP-1 and NF-κB with IC50 values of 8.74 ± 0.31 and 12.08 ± 0.24 μM, respectively. DISCUSSION AND CONCLUSION Taken together, these data suggest that comp has a significant anti-inflammatory property and warrants further studies concerning the potential of comp for medicinal use.
Collapse
Affiliation(s)
- Irshad Ali
- School of Medicine and Brain Korea 21 PLUS Program, Jeju National University, Jeju, South Korea
- Institute of Medical Science, Jeju National University, Jeju, South Korea
| | - Zahid Manzoor
- School of Medicine and Brain Korea 21 PLUS Program, Jeju National University, Jeju, South Korea
- Institute of Medical Science, Jeju National University, Jeju, South Korea
| | - Jung-Eun Koo
- School of Medicine and Brain Korea 21 PLUS Program, Jeju National University, Jeju, South Korea
- Institute of Medical Science, Jeju National University, Jeju, South Korea
| | - Jung-Eun Kim
- Department of Chemistry and Cosmetics, College of Natural Sciences, Jeju National University, Jeju, South Korea
| | - Sang-Hee Byeon
- Department of Chemistry and Cosmetics, College of Natural Sciences, Jeju National University, Jeju, South Korea
| | - Eun-Sook Yoo
- School of Medicine and Brain Korea 21 PLUS Program, Jeju National University, Jeju, South Korea
- Institute of Medical Science, Jeju National University, Jeju, South Korea
| | - Hee-Kyoung Kang
- School of Medicine and Brain Korea 21 PLUS Program, Jeju National University, Jeju, South Korea
- Institute of Medical Science, Jeju National University, Jeju, South Korea
| | - Jin-Won Hyun
- School of Medicine and Brain Korea 21 PLUS Program, Jeju National University, Jeju, South Korea
- Institute of Medical Science, Jeju National University, Jeju, South Korea
| | - Nam-Ho Lee
- Department of Chemistry and Cosmetics, College of Natural Sciences, Jeju National University, Jeju, South Korea
| | - Young-Sang Koh
- School of Medicine and Brain Korea 21 PLUS Program, Jeju National University, Jeju, South Korea
- Institute of Medical Science, Jeju National University, Jeju, South Korea
- CONTACT Young-Sang KohDepartment of Microbiology and Immunology, Jeju National University School of Medicine, 102 Jejudaehakno, Jeju63243, South Korea
| |
Collapse
|
18
|
Wallqvist A, Wang H, Zavaljevski N, Memišević V, Kwon K, Pieper R, Rajagopala SV, Reifman J. Mechanisms of action of Coxiella burnetii effectors inferred from host-pathogen protein interactions. PLoS One 2017; 12:e0188071. [PMID: 29176882 PMCID: PMC5703456 DOI: 10.1371/journal.pone.0188071] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/31/2017] [Indexed: 02/06/2023] Open
Abstract
Coxiella burnetii is an obligate Gram-negative intracellular pathogen and the etiological agent of Q fever. Successful infection requires a functional Type IV secretion system, which translocates more than 100 effector proteins into the host cytosol to establish the infection, restructure the intracellular host environment, and create a parasitophorous vacuole where the replicating bacteria reside. We used yeast two-hybrid (Y2H) screening of 33 selected C. burnetii effectors against whole genome human and murine proteome libraries to generate a map of potential host-pathogen protein-protein interactions (PPIs). We detected 273 unique interactions between 20 pathogen and 247 human proteins, and 157 between 17 pathogen and 137 murine proteins. We used orthology to combine the data and create a single host-pathogen interaction network containing 415 unique interactions between 25 C. burnetii and 363 human proteins. We further performed complementary pairwise Y2H testing of 43 out of 91 C. burnetii-human interactions involving five pathogen proteins. We used the combined data to 1) perform enrichment analyses of target host cellular processes and pathways, 2) examine effectors with known infection phenotypes, and 3) infer potential mechanisms of action for four effectors with uncharacterized functions. The host-pathogen interaction profiles supported known Coxiella phenotypes, such as adapting cell morphology through cytoskeletal re-arrangements, protein processing and trafficking, organelle generation, cholesterol processing, innate immune modulation, and interactions with the ubiquitin and proteasome pathways. The generated dataset of PPIs-the largest collection of unbiased Coxiella host-pathogen interactions to date-represents a rich source of information with respect to secreted pathogen effector proteins and their interactions with human host proteins.
Collapse
Affiliation(s)
- Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland, United States of America
| | - Hao Wang
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland, United States of America
| | - Nela Zavaljevski
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland, United States of America
| | - Vesna Memišević
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland, United States of America
| | - Keehwan Kwon
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Rembert Pieper
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | | | - Jaques Reifman
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
19
|
Abnave P, Muracciole X, Ghigo E. Coxiella burnetii Lipopolysaccharide: What Do We Know? Int J Mol Sci 2017; 18:ijms18122509. [PMID: 29168790 PMCID: PMC5751112 DOI: 10.3390/ijms18122509] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/28/2022] Open
Abstract
A small gram-negative bacterium, Coxiella burnetii (C. burnetii), is responsible for a zoonosis called Q fever. C. burnetii is an intracellular bacterium that can survive inside microbicidal cells like monocytes and macrophages by hijacking several functions of the immune system. Among several virulence factors, the lipopolysaccharide (LPS) of C. burnetii is one of the major factors involved in this immune hijacking because of its atypical composition and structure. Thus, the aim of this mini-review is to summarize the repressive effects of C. burnetii LPS on the antibacterial immunity of cells.
Collapse
Affiliation(s)
- Prasad Abnave
- Department of Zoology, University of Oxford, Tinbergen Building, South Parks Road, Oxford OX1 3PS, UK.
| | - Xavier Muracciole
- Department of Radiotherapy Oncology, CHU de la Timone, Assistance Publique-Hopitaux Marseille, 13385 Marseille, France.
| | - Eric Ghigo
- Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes (URMITE), Institut Hospitalier Universitaire Méditerranée-Infection, 19-21 Bd Jean Moulin, CEDEX 05, 13385 Marseille, France.
| |
Collapse
|
20
|
Ying S, Guo J, Dai Z, Zhu H, Yu J, Ma W, Li J, Akhtar MF, Shi Z. Time course effect of lipopolysaccharide on Toll-like receptors expression and steroidogenesis in the Chinese goose ovary. Reproduction 2017; 153:509-518. [DOI: 10.1530/rep-17-0011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 01/06/2017] [Accepted: 02/07/2017] [Indexed: 01/18/2023]
Abstract
The ovary of Chinese goose is easily infected by microorganisms because of the mating behaviour in water, which causes decreased laying performance. This study investigated the time course effect of lipopolysaccharide (LPS) on the steroidogenesis and mRNA expression of Toll-like receptors (TLRs), a class of key pattern recognition receptor, in the breeding goose ovary. The laying geese were treated intravenously with LPS for 0, 6, 12, 24 and 36 h, and all birds were slaughtered approximately 8 h after oviposition. The expression levels of TLRs in the white and yellowish follicles, and granulosa and theca layers of hierarchical follicles were examined by real-time PCR. All 10 members of avian TLR family were differentially expressed among the different follicular tissues. Moreover, at 24 and 36 h after LPS treatment, the hierarchical follicle morphological structure was altered, but the expression levels of TLRs were still higher than the control. Furthermore, during LPS treatment period, the expression pattern of TLRs 2A and 4 genes was similar to that of TLR15 in the white follicles, TLRs 1B, 5 and 15 in the yellowish follicles, TLRs 7 and 15 in the granulosa layer, and TLRs 1A, 2B, 3, 7 and 15 in the theca layer, which had a negative correlation with the kinetics of plasma P4 and E2 concentrations. In conclusion, the mechanism by which pathogen infection inhibited goose follicular growth and further decreased egg production may involve a gradually enhanced inflammatory response and reduced endocrine function. This may be due to stimulated TLRs in the ovary.
Collapse
|
21
|
Eldin C, Mélenotte C, Mediannikov O, Ghigo E, Million M, Edouard S, Mege JL, Maurin M, Raoult D. From Q Fever to Coxiella burnetii Infection: a Paradigm Change. Clin Microbiol Rev 2017; 30:115-190. [PMID: 27856520 PMCID: PMC5217791 DOI: 10.1128/cmr.00045-16] [Citation(s) in RCA: 558] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Coxiella burnetii is the agent of Q fever, or "query fever," a zoonosis first described in Australia in 1937. Since this first description, knowledge about this pathogen and its associated infections has increased dramatically. We review here all the progress made over the last 20 years on this topic. C. burnetii is classically a strict intracellular, Gram-negative bacterium. However, a major step in the characterization of this pathogen was achieved by the establishment of its axenic culture. C. burnetii infects a wide range of animals, from arthropods to humans. The genetic determinants of virulence are now better known, thanks to the achievement of determining the genome sequences of several strains of this species and comparative genomic analyses. Q fever can be found worldwide, but the epidemiological features of this disease vary according to the geographic area considered, including situations where it is endemic or hyperendemic, and the occurrence of large epidemic outbreaks. In recent years, a major breakthrough in the understanding of the natural history of human infection with C. burnetii was the breaking of the old dichotomy between "acute" and "chronic" Q fever. The clinical presentation of C. burnetii infection depends on both the virulence of the infecting C. burnetii strain and specific risks factors in the infected patient. Moreover, no persistent infection can exist without a focus of infection. This paradigm change should allow better diagnosis and management of primary infection and long-term complications in patients with C. burnetii infection.
Collapse
Affiliation(s)
- Carole Eldin
- URMITE, UMR CNRS 7278, IRD 198, INSERM U1095, Faculté de Médecine, Marseille, France
| | - Cléa Mélenotte
- URMITE, UMR CNRS 7278, IRD 198, INSERM U1095, Faculté de Médecine, Marseille, France
| | - Oleg Mediannikov
- URMITE, UMR CNRS 7278, IRD 198, INSERM U1095, Faculté de Médecine, Marseille, France
| | - Eric Ghigo
- URMITE, UMR CNRS 7278, IRD 198, INSERM U1095, Faculté de Médecine, Marseille, France
| | - Matthieu Million
- URMITE, UMR CNRS 7278, IRD 198, INSERM U1095, Faculté de Médecine, Marseille, France
| | - Sophie Edouard
- URMITE, UMR CNRS 7278, IRD 198, INSERM U1095, Faculté de Médecine, Marseille, France
| | - Jean-Louis Mege
- URMITE, UMR CNRS 7278, IRD 198, INSERM U1095, Faculté de Médecine, Marseille, France
| | - Max Maurin
- Institut de Biologie et de Pathologie, CHU de Grenoble, Grenoble, France
| | - Didier Raoult
- URMITE, UMR CNRS 7278, IRD 198, INSERM U1095, Faculté de Médecine, Marseille, France
| |
Collapse
|
22
|
Mahapatra S, Gallaher B, Smith SC, Graham JG, Voth DE, Shaw EI. Coxiella burnetii Employs the Dot/Icm Type IV Secretion System to Modulate Host NF-κB/RelA Activation. Front Cell Infect Microbiol 2016; 6:188. [PMID: 28066723 PMCID: PMC5165255 DOI: 10.3389/fcimb.2016.00188] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 12/02/2016] [Indexed: 12/17/2022] Open
Abstract
Coxiella burnetii is the causative agent of Q fever and an obligate intracellular pathogen in nature that survives and grows in a parasitophorous vacuole (PV) within eukaryotic host cells. C. burnetii promotes intracellular survival by subverting apoptotic and pro-inflammatory signaling pathways that are typically regulated by nuclear transcription factor-κB (NF-κB). We and others have demonstrated that C. burnetii NMII proteins inhibit expression of pro-inflammatory cytokines and induce expression of anti-apoptotic genes during infection. Here, we demonstrate that C. burnetii promotes intracellular survival by modulating NF-κB subunit p65 (RelA) phosphorylation, and thus activation, in a Type Four B Secretion System (T4BSS)-dependent manner. Immunoblot analysis of RelA phosphorylated at serine-536 demonstrated that C. burnetii increases NF-κB activation via the canonical pathway. However, RelA phosphorylation levels were even higher in infected cells where bacterial protein or mRNA synthesis was inhibited. Importantly, we demonstrate that inhibition of RelA phosphorylation impairs PV formation and C. burnetii growth. We found that a T4BSS-defective mutant (CbΔdotA) elicited phosphorylated RelA levels similar to those of wild type C. burnetii infection treated with Chloramphenicol. Moreover, cells infected with CbΔdotA or wild type C. burnetii treated with Chloramphenicol showed similar levels of GFP-RelA nuclear localization, and significantly increased localization compared to wild type C. burnetii infection. These data indicate that without de novo protein synthesis and a functional T4BSS, C. burnetii is unable to modulate NF-κB activation, which is crucial for optimal intracellular growth.
Collapse
Affiliation(s)
- Saugata Mahapatra
- Department of Microbiology and Molecular genetics, Oklahoma State UniversityStillwater, OK, USA
| | - Brandi Gallaher
- Department of Microbiology and Molecular genetics, Oklahoma State UniversityStillwater, OK, USA
| | - Sydni Caet Smith
- Department of Microbiology and Molecular genetics, Oklahoma State UniversityStillwater, OK, USA
| | - Joseph G. Graham
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS)Little Rock, AR, USA
| | - Daniel E. Voth
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS)Little Rock, AR, USA
| | - Edward I. Shaw
- Department of Microbiology and Molecular genetics, Oklahoma State UniversityStillwater, OK, USA
| |
Collapse
|
23
|
Ryu KA, Slowinska K, Moore T, Esser-Kahn A. Immune Response Modulation of Conjugated Agonists with Changing Linker Length. ACS Chem Biol 2016; 11:3347-3352. [PMID: 27749034 DOI: 10.1021/acschembio.6b00895] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We report immune response modulation with linked Toll-like receptor (TLR) agonists. Conjugating two agonists of synergistic TLRs induce an increase in immune activity compared to equal molarity of soluble agonists. Additionally, varying the distance between the agonists by changing the linker length alters the level of macrophage NF-κB activity as well as primary bone marrow derived dendritic cell IL-6 production. This modulation is effected by the size of the agonists and the pairing of the stimulated TLRs. The sensitivity of linker-length-dependent immune activity of conjugated agonists provides the potential for developing application specific therapeutics.
Collapse
Affiliation(s)
- Keun Ah Ryu
- Department
of Chemistry, University of California−Irvine, Irvine, California 92697, United States
| | - Katarzyna Slowinska
- Department
of Chemistry and Biochemistry, California State University−Long Beach, Long Beach, California 90840, United States
| | - Troy Moore
- Department
of Chemistry, University of California−Irvine, Irvine, California 92697, United States
| | - Aaron Esser-Kahn
- Department
of Chemistry, University of California−Irvine, Irvine, California 92697, United States
| |
Collapse
|
24
|
Battisti JM, Watson LA, Naung MT, Drobish AM, Voronina E, Minnick MF. Analysis of the Caenorhabditis elegans innate immune response to Coxiella burnetii. Innate Immun 2016; 23:111-127. [PMID: 27884946 DOI: 10.1177/1753425916679255] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The nematode Caenorhabditis elegans is well established as a system for characterization and discovery of molecular mechanisms mediating microbe-specific inducible innate immune responses to human pathogens. Coxiella burnetii is an obligate intracellular bacterium that causes a flu-like syndrome in humans (Q fever), as well as abortions in domesticated livestock, worldwide. Initially, when wild type C. elegans (N2 strain) was exposed to mCherry-expressing C. burnetii (CCB) a number of overt pathological manifestations resulted, including intestinal distension, deformed anal region and a decreased lifespan. However, nematodes fed autoclave-killed CCB did not exhibit these symptoms. Although vertebrates detect C. burnetii via TLRs, pathologies in tol-1(-) mutant nematodes were indistinguishable from N2, and indicate nematodes do not employ this orthologue for detection of C. burnetii. sek-1(-) MAP kinase mutant nematodes succumbed to infection faster, suggesting that this signaling pathway plays a role in immune activation, as previously shown for orthologues in vertebrates during a C. burnetii infection. C. elegans daf-2(-) mutants are hyper-immune and exhibited significantly reduced pathological consequences during challenge. Collectively, these results demonstrate the utility of C. elegans for studying the innate immune response against C. burnetii and could lead to discovery of novel methods for prevention and treatment of disease in humans and livestock.
Collapse
Affiliation(s)
- James M Battisti
- Program in Cellular, Molecular and Microbial Biology, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Lance A Watson
- Program in Cellular, Molecular and Microbial Biology, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Myo T Naung
- Program in Cellular, Molecular and Microbial Biology, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Adam M Drobish
- Program in Cellular, Molecular and Microbial Biology, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Ekaterina Voronina
- Program in Cellular, Molecular and Microbial Biology, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Michael F Minnick
- Program in Cellular, Molecular and Microbial Biology, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| |
Collapse
|
25
|
Gonnissen D, Qu Y, Langer K, Öztürk C, Zhao Y, Chen C, Seebohm G, Düfer M, Fuchs H, Galla HJ, Riehemann K. Comparison of cellular effects of starch-coated SPIONs and poly(lactic-co-glycolic acid) matrix nanoparticles on human monocytes. Int J Nanomedicine 2016; 11:5221-5236. [PMID: 27789942 PMCID: PMC5072557 DOI: 10.2147/ijn.s106540] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Within the last years, progress has been made in the knowledge of the properties of medically used nanoparticles and their toxic effects, but still, little is known about their influence on cellular processes of immune cells. The aim of our comparative study was to present the influence of two different nanoparticle types on subcellular processes of primary monocytes and the leukemic monocyte cell line MM6. We used core-shell starch-coated superparamagnetic iron oxide nanoparticles (SPIONs) and matrix poly(lactic-co-glycolic acid) (PLGA) nanoparticles for our experiments. In addition to typical biocompatibility testing like the detection of necrosis or secretion of interleukins (ILs), we investigated the impact of these nanoparticles on the actin cytoskeleton and the two voltage-gated potassium channels Kv1.3 and Kv7.1. Induction of necrosis was not seen for PLGA nanoparticles and SPIONs in primary monocytes and MM6 cells. Likewise, no alteration in secretion of IL-1β and IL-10 was detected under the same experimental conditions. In contrast, IL-6 secretion was exclusively downregulated in primary monocytes after contact with both nanoparticles. Two-electrode voltage clamp experiments revealed that both nanoparticles reduce currents of the aforementioned potassium channels. The two nanoparticles differed significantly in their impact on the actin cytoskeleton, demonstrated via atomic force microscopy elasticity measurement and phalloidin staining. While SPIONs led to the disruption of the respective cytoskeleton, PLGA did not show any influence in both experimental setups. The difference in the effects on ion channels and the actin cytoskeleton suggests that nanoparticles affect these subcellular components via different pathways. Our data indicate that the alteration of the cytoskeleton and the effect on ion channels are new parameters that describe the influence of nanoparticles on cells. The results are highly relevant for medical application and further evaluation of nanomaterial biosafety.
Collapse
Affiliation(s)
- Dominik Gonnissen
- Center for Nanotechnology, Institute of Physics, University of Münster, Münster, Germany
| | - Ying Qu
- Center for Nanotechnology, Institute of Physics, University of Münster, Münster, Germany; National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University of Münster, Münster
| | | | - Yuliang Zhao
- National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Chunying Chen
- National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Guiscard Seebohm
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases, University Hospital Münster
| | - Martina Düfer
- Department of Pharmacology, Institute of Pharmaceutical and Medicinal Chemistry
| | - Harald Fuchs
- Center for Nanotechnology, Institute of Physics, University of Münster, Münster, Germany
| | - Hans-Joachim Galla
- Department of Cell Biology/Biophysics, Institute of Biochemistry, University of Münster, Münster, Germany
| | - Kristina Riehemann
- Center for Nanotechnology, Institute of Physics, University of Münster, Münster, Germany
| |
Collapse
|
26
|
Roles of Toll-Like Receptor 2 (TLR2), TLR4, and MyD88 during Pulmonary Coxiella burnetii Infection. Infect Immun 2016; 84:940-949. [PMID: 26787722 DOI: 10.1128/iai.00898-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 01/14/2016] [Indexed: 01/15/2023] Open
Abstract
Coxiella burnetii, the causative agent of Q fever, is an obligate intracellular, primarily pulmonary, bacterial pathogen. Although much is known about adaptive immune responses against this bacterium, our understanding of innate immune responses against C. burnetii is not well defined, particularly within the target tissue for infection, the lung. Previous studies examined the roles of the innate immune system receptors Toll-like receptor 2 (TLR2) and TLR4 in peripheral infection models and described minimal phenotypes in specific gene deletion animals compared to those of their wild-type controls (S. Meghari et al., Ann N Y Acad Sci 1063:161-166, 2005,http://dx.doi.org/10.1196/annals.1355.025; A. Honstettre et al., J Immunol 172:3695-3703, 2004,http://dx.doi.org/10.4049/jimmunol.172.6.3695) . Here, we assessed the roles for TLR2, TLR4, and MyD88 in pulmonary C. burnetii infection and compared responses to those that occurred in TLR2- and TLR4-deficient animals following peripheral infection. As observed previously, neither TLR2 nor TLR4 was needed for limiting bacterial growth after peripheral infection. In contrast, TLR2 and, to a lesser extent, TLR4 limited growth (or dissemination) of the bacterium in the lung and spleen after pulmonary infection. TLR2, TLR4, and MyD88 were not required for the general inflammatory response in the lungs after pulmonary infection. However, MyD88 signaling was important for infection-induced morbidity. Finally, TLR2 expression on hematopoietic cells was most important for limiting bacterial growth in the lung. These results expand on our knowledge of the roles for TLR2 and TLR4 in C. burnetii infection and suggest various roles for these receptors that are dictated by the site of infection.
Collapse
|
27
|
Malov VA, Ponomarev SV, Тarasevich IV, Kubensky EN, Gorobchenko AN, Pantyukhina AN, Nemilostiva EA, Bogdanova MV, Makhmutov YI. [Description of a case of severe Q fever]. TERAPEVT ARKH 2016; 87:84-91. [PMID: 26821422 DOI: 10.17116/terarkh2015871184-91] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The paper considers a rare clinical case of severe Q fever in a young man with no compromised premorbid background. It describes and analyzes clinical manifestations and laboratory findings with consideration for the current data available in the literature. The issues of the differential diagnosis, laboratory diagnosis, and treatment of Q fever are discussed.
Collapse
Affiliation(s)
- V A Malov
- Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - S V Ponomarev
- Acad. N.N. Burdenko Main Military Clinical Hospital, Ministry of Defense of the Russian Federation, Moscow, Russia
| | - I V Тarasevich
- N.F Gamaleya Federal Research Center for Epidemiology and Microbiology, Ministry of Health of Russia, Moscow, Russia
| | - E N Kubensky
- Acad. N.N. Burdenko Main Military Clinical Hospital, Ministry of Defense of the Russian Federation, Moscow, Russia
| | - A N Gorobchenko
- Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - A N Pantyukhina
- N.F Gamaleya Federal Research Center for Epidemiology and Microbiology, Ministry of Health of Russia, Moscow, Russia
| | - E A Nemilostiva
- Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - M V Bogdanova
- Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - Ya I Makhmutov
- Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
28
|
Ali I, Manzoor Z, Koh YS. 3-Hydroxy-4,7-megastigmadien-9-one, Isolated fromUlva pertusaKjellman, Inhibits LPS-Induced Inflammatory Response by Down-Regulating Mitogen-Activated Protein Kinase and NF-κB Pathways. ACTA ACUST UNITED AC 2016. [DOI: 10.4167/jbv.2016.46.3.167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Irshad Ali
- Department of Microbiology and Immunology, School of Medicine and Brain Korea 21 PLUS Program, Jeju National University, Jeju, Korea
- Institute of Medical Science, Jeju National University, Jeju, Korea
| | - Zahid Manzoor
- Department of Microbiology and Immunology, School of Medicine and Brain Korea 21 PLUS Program, Jeju National University, Jeju, Korea
- Institute of Medical Science, Jeju National University, Jeju, Korea
| | - Young-Sang Koh
- Department of Microbiology and Immunology, School of Medicine and Brain Korea 21 PLUS Program, Jeju National University, Jeju, Korea
- Institute of Medical Science, Jeju National University, Jeju, Korea
| |
Collapse
|
29
|
Djaldetti M, Bessler H. High temperature affects the phagocytic activity of human peripheral blood mononuclear cells. Scandinavian Journal of Clinical and Laboratory Investigation 2015; 75:482-6. [PMID: 26067609 DOI: 10.3109/00365513.2015.1052550] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND The ability for engulfment of pathogens and inert particles is the key hallmark of the phagocytic cells. Phagocytes play a significant role in the modulation of local or extended inflammation. Since fever activates a number of factors linked with the immune response it was the goal of this study to examine the in vitro effect of hyperthermia on the phagocytic capacity, the number of phagocytic cells and the viability of human peripheral blood mononuclear cells (PBMC) at 37 and 40°C. METHODS PBMC were incubated with 0.8 μm polysterene latex beads, for 2 hours at 37 and 40°C. The number of phagocytic cells, and that of latex particles internalized by each individual cell was counted with a light microscope. In addition, the percentage of viable cells and the number of active metabolic cells was evaluated. RESULTS A temperature of 40°C significantly increased the number of phagocytic cells and the phagocytic index by 41 and 37% respectively, as compared to cells incubated at 37°C. While the number of vital cells (trypan blue test) did not differ statistically at both temperatures, the number of active metabolic cells (XTT test) after 2 h of incubation at 40°C was 17% higher as compared with that at 37°C. However, the number of active metabolic cells after 24 h of incubation at 40°C was 51% lower compared with cells incubated at 37°C. CONCLUSIONS The increased phagocytic capacity of human peripheral blood monocytes at high temperature further enlightens the immunomodulatory effect of fever in the immune responses during inflammation.
Collapse
Affiliation(s)
- Meir Djaldetti
- Laboratory for Immunology and Hematology Research, Rabin Medical Center, Hasharon Hospital,Petah-Tiqva, and the Sackler School of Medicine, Tel-Aviv University , Ramat-Aviv , Israel
| | | |
Collapse
|
30
|
Neutrophils play an important role in protective immunity against Coxiella burnetii infection. Infect Immun 2015; 83:3104-13. [PMID: 26015476 DOI: 10.1128/iai.00042-15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 05/13/2015] [Indexed: 12/19/2022] Open
Abstract
Coxiella burnetii is an obligate intracellular Gram-negative bacterium that causes the zoonotic disease Q fever. Although Q fever is mainly transmitted by aerosol infection, study of the immune responses in the lung following pulmonary C. burnetii infection is lacking. Neutrophils are considered the first immune cell to migrate into the lung and play an important role in host defense against aerosol infection with microbial pathogens. However, the role of neutrophils in the host defense against C. burnetii infection remains unclear. To determine the role of neutrophils in protective immunity against C. burnetii infection, the RB6-8C5 antibody was used to deplete neutrophils in mice before intranasal infection with C. burnetii. The results indicated that neutrophil-depleted mice developed more severe disease than their wild-type counterparts, suggesting that neutrophils play an important role in host defense against C. burnetii pulmonary infection. We also found that neither CXC chemokine receptor 2 (CXCR2) nor interleukin-17 (IL-17) receptor (IL-17R) deficiency changed the severity of disease following intranasal C. burnetii challenge, suggesting that keratinocyte-derived chemokine and IL-17 may not play essential roles in the response to C. burnetii infection. However, significantly higher C. burnetii genome copy numbers were detected in the lungs of IL-1R(-/-) mice at 14 days postinfection. This indicates that IL-1 may be important for the clearance of C. burnetii from the lungs following intranasal infection. Our results also suggest that neutrophils are involved in protecting vaccinated mice from C. burnetii challenge-induced disease. This is the first study to demonstrate an important role for neutrophils in protective immunity against C. burnetii infection.
Collapse
|
31
|
Klein DCG, Skjesol A, Kers-Rebel ED, Sherstova T, Sporsheim B, Egeberg KW, Stokke BT, Espevik T, Husebye H. CD14, TLR4 and TRAM Show Different Trafficking Dynamics During LPS Stimulation. Traffic 2015; 16:677-90. [PMID: 25707286 DOI: 10.1111/tra.12274] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 02/19/2015] [Accepted: 02/20/2015] [Indexed: 12/18/2022]
Abstract
Toll-like receptor 4 (TLR4) is responsible for the immediate response to Gram-negative bacteria and signals via two main pathways by recruitment of distinct pairs of adaptor proteins. Mal-MyD88 [Mal (MyD88-adaptor-like) - MYD88 (Myeloid differentiation primary response gene (88))] is recruited to the plasma membrane to initiate the signaling cascade leading to production of pro-inflammatory cytokines while TRAM-TRIF [TRAM (TRIF-related adaptor molecule)-TRIF (TIR-domain-containing adapter-inducing interferon-β)] is recruited to early endosomes to initiate the subsequent production of type I interferons. We have investigated the dynamics of TLR4 and TRAM during lipopolysaccharide (LPS) stimulation. We found that LPS induced a CD14-dependent immobile fraction of TLR4 in the plasma membrane. Total internal reflection fluorescence microscopy (TIRF) revealed that LPS stimulation induced clustering of TLR4 into small punctate structures in the plasma membrane containing CD14/LPS and clathrin, both in HEK293 cells and the macrophage model cell line U373-CD14. These results suggest that laterally immobilized TLR4 receptor complexes are being formed and prepared for endocytosis. RAB11A was found to be involved in localizing TRAM to the endocytic recycling compartment (ERC) and to early sorting endosomes. Moreover, CD14/LPS but not TRAM was immobilized on RAB11A-positive endosomes, which indicates that TRAM and CD14/LPS can independently be recruited to endosomes.
Collapse
Affiliation(s)
- Dionne C G Klein
- Department of Cancer Research and Molecular Medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Astrid Skjesol
- Department of Cancer Research and Molecular Medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Esther D Kers-Rebel
- Graduate School of Life Sciences, University of Utrecht, Utrecht, The Netherlands.,Present address: Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Tatyana Sherstova
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjørnar Sporsheim
- Department of Cancer Research and Molecular Medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kjartan W Egeberg
- Department of Cancer Research and Molecular Medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjørn T Stokke
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Terje Espevik
- Department of Cancer Research and Molecular Medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Harald Husebye
- Department of Cancer Research and Molecular Medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
32
|
Morin SO, Giroux V, Favre C, Bechah Y, Auphan-Anezin N, Roncagalli R, Mège JL, Olive D, Malissen M, Nunès JA. In the absence of its cytosolic domain, the CD28 molecule still contributes to T cell activation. Cell Mol Life Sci 2015; 72:2739-48. [PMID: 25725801 DOI: 10.1007/s00018-015-1873-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 02/04/2015] [Accepted: 02/24/2015] [Indexed: 12/13/2022]
Abstract
The CD28 costimulatory receptor has a pivotal role in T cell biology as this molecule amplifies T cell receptor (TCR) signals to provide an efficient immune T cell response. There is a large debate about how CD28 mediates these signals. Here, we designed a CD28 gene-targeted knock-in mouse strain lacking the cytoplasmic tail of CD28. As is the case in CD28-deficient (CD28 knock-out) mice, regulatory T cell homeostasis and T cell activation are altered in these CD28 knock-in mice. Unexpectedly, the presence of a CD28 molecule deprived of its cytoplasmic tail could partially induce some early activation events in T cells such as signaling events or expression of early activation markers. These results unravel a new mechanism of T cell costimulation by CD28, independent of its cytoplasmic tail.
Collapse
Affiliation(s)
- Stéphanie O Morin
- Centre de Recherche en Cancérologie de Marseille (CRCM), 27 Bd Leï Roure, BP 30059, 13273, Marseille Cedex 09, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Conti F, Boucherit N, Baldassarre V, Trouplin V, Toman R, Mottola G, Mege JL, Ghigo E. Coxiella burnetii lipopolysaccharide blocks p38α-MAPK activation through the disruption of TLR-2 and TLR-4 association. Front Cell Infect Microbiol 2015; 4:182. [PMID: 25610812 PMCID: PMC4285172 DOI: 10.3389/fcimb.2014.00182] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/10/2014] [Indexed: 11/13/2022] Open
Abstract
To survive in macrophages, Coxiella burnetii hijacks the activation pathway of macrophages. Recently, we have demonstrated that C. burnetii, via its lipopolysaccharide (LPS), avoids the activation of p38α-MAPK through an antagonistic engagement of Toll-like receptor (TLR)-4. We investigated the fine-tuned mechanism leading to the absence of activation of the p38α-MAPK despite TLR-4 engagement. In macrophages challenged with LPS from the avirulent variants of C. burnetii, TLR-4 and TLR-2 co-immunoprecipitated. This association was absent in cells challenged by the LPS of pathogenic C. burnetii. The disruption makes TLRs unable to signal during the recognition of the LPS of pathogenic C. burnetii. The disruption of TLR-2 and TLR-4 was induced by the re-organization of the macrophage cytoskeleton by C. burnetii LPS. Interestingly, blocking the actin cytoskeleton re-organization relieved the disruption of the association TLR-2/TLR-4 by pathogenic C. burnetii and rescued the p38α-MAPK activation by C. burnetii. We elucidated an unexpected mechanism allowing pathogenic C. burnetii to avoid macrophage activation by the disruption of the TLR-2 and TLR-4 association.
Collapse
Affiliation(s)
- Filippo Conti
- CNRS UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université Marseille, France
| | - Nicolas Boucherit
- CNRS UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université Marseille, France
| | | | - Virginie Trouplin
- CNRS UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université Marseille, France
| | - Rudolf Toman
- Laboratory for Diagnosis and Prevention of Rickettsial and Chlamydial Infections, Institute of Virology, Slovak Academy of Sciences Bratislava, Slovakia
| | - Giovanna Mottola
- UMR MD2, Faculté de Médecine NORD, Aix-Marseille Université and IRBA (Institute of Research in Biology of the French Army) Marseille, France ; Laboratory of Biochemistry, La Timone University Hospital, Assistance Publique Hôpitaux de Marseille Marseille, France
| | - Jean-Louis Mege
- CNRS UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université Marseille, France
| | - Eric Ghigo
- CNRS UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université Marseille, France
| |
Collapse
|
34
|
Ammerdorffer A, Schoffelen T, Gresnigt MS, Oosting M, den Brok MH, Abdollahi-Roodsaz S, Kanneganti TD, de Jong DJ, van Deuren M, Roest HJ, Rebel JM, Netea MG, Joosten LAB, Sprong T. Recognition of Coxiella burnetii by toll-like receptors and nucleotide-binding oligomerization domain-like receptors. J Infect Dis 2014; 211:978-87. [PMID: 25246533 DOI: 10.1093/infdis/jiu526] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Infection with Coxiella burnetii can lead to acute and chronic Q fever. Toll-like receptor 1 (TLR1), TLR2, TLR4, TLR6, nucleotide-binding oligomerization domain receptor 1 (NOD1), NOD2, and the mitogen-activated protein kinases are central in the innate immune response against microorganisms, but little is known about their role in the recognition of C. burnetii in humans. METHODS Human peripheral blood mononuclear cells (PBMCs) were stimulated with C. burnetii Nine Mile and the Dutch outbreak isolate C. burnetii 3262. TLRs were inhibited using specific antibodies or antagonists. Additionally, the influence of human polymorphisms in TLRs and Nod-like receptors (NLRs) on C. burnetii-induced cytokine production was assessed. RESULTS Inhibition of TLR2, p38, JNK, and ERK led to decreased cytokine responses in C. burnetii-stimulated human PBMCs. Humans with polymorphisms in TLR1 and NOD2 had reduced cytokine production, compared with humans with wild-type genotypes, after stimulation. Interestingly, polymorphisms in TLR6 led to decreased cytokine production after C. burnetii 3262 stimulation but not after C. burnetii Nine Mile stimulation. CONCLUSIONS The TLR1/TLR2 heterodimer and NOD2 are important recognition receptors for the induction of cytokine responses against C. burnetii in humans. Furthermore, an interesting finding was the divergent recognition of C. burnetii Nine Mile and C. burnetii 3262.
Collapse
Affiliation(s)
| | | | | | | | - Martijn H den Brok
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences
| | | | | | - Dirk J de Jong
- Department of Gastroenterology, Radboud University Medical Center
| | | | | | - Johanna M Rebel
- Department of Infection Biology, Central Veterinary Institute, Lelystad, the Netherlands
| | | | | | - Tom Sprong
- Department of Internal Medicine Department of Internal Medicine Department of Medical Microbiology and Infectious Diseases, CWZ, Nijmegen
| |
Collapse
|
35
|
Characterization of a lipopolysaccharide-targeted monoclonal antibody and its variable fragments as candidates for prophylaxis against the obligate intracellular bacterial pathogen Coxiella burnetii. Infect Immun 2014; 82:4530-41. [PMID: 25114119 DOI: 10.1128/iai.01695-14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Our previous study demonstrated that treatment of Coxiella burnetii with the phase I lipopolysaccharide (PI-LPS)-targeted monoclonal antibody (MAb) 1E4 significantly inhibited C. burnetii infection in mice, suggesting that 1E4 is a protective MAb. To determine whether passive transfer of antibodies (Abs) can provide protection against C. burnetii natural infection, we examined if passive transfer of 1E4 would protect SCID mice against C. burnetii aerosol infection. The results indicated that 1E4 conferred significant protection against aerosolized C. burnetii, suggesting that 1E4 may be useful for preventing C. burnetii natural infection. To further understand the mechanisms of 1E4-mediated protection and to test the possibility of using humanized 1E4 to prevent C. burnetii infection, we examined whether the Fab fragment of 1E4 (Fab1E4), a recombinant murine single-chain variable fragment (muscFv1E4), and a humanized single-chain variable fragment (huscFv1E4) retained the ability of 1E4 to inhibit C. burnetii infection. The results indicated that Fab1E4, muscFv1E4, and huscFv1E4 were able to inhibit C. burnetii infection in mice but that their ability to inhibit C. burnetii infection was lower than that of 1E4. In addition, treatment of C. burnetii with Fab1E4, muscFv1E4, or huscFv1E4 can block C. burnetii infection of macrophages. Interestingly, treatment of C. burnetii with huscFv1E4 can significantly reduce C. burnetii infectivity in human macrophages. This report provides the first evidence to demonstrate that the humanized variable fragments of an LPS-specific MAb can neutralize C. burnetii infection and appears to be a promising step toward the potential use of a humanized MAb as emergency prophylaxis against C. burnetii exposure.
Collapse
|
36
|
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: 37] [Impact Index Per Article: 3.4] [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
|
37
|
van Schaik EJ, Chen C, Mertens K, Weber MM, Samuel JE. Molecular pathogenesis of the obligate intracellular bacterium Coxiella burnetii. Nat Rev Microbiol 2013; 11:561-73. [PMID: 23797173 DOI: 10.1038/nrmicro3049] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The agent of Q fever, Coxiella burnetii, is an obligate intracellular bacterium that causes acute and chronic infections. The study of C. burnetii pathogenesis has benefited from two recent fundamental advances: improved genetic tools and the ability to grow the bacterium in extracellular media. In this Review, we describe how these recent advances have improved our understanding of C. burnetii invasion and host cell modulation, including the formation of replication-permissive Coxiella-containing vacuoles. Furthermore, we describe the Dot/Icm (defect in organelle trafficking/intracellular multiplication) system, which is used by C. burnetii to secrete a range of effector proteins into the host cell, and we discuss the role of these effectors in remodelling the host cell.
Collapse
Affiliation(s)
- Erin J van Schaik
- Department of Microbial and Molecular Pathogenesis, Texas A&M Health Science Center College of Medicine, Bryan, Texas 77807-3260, USA
| | | | | | | | | |
Collapse
|
38
|
Orenstein R. Travel in patients receiving TNF-alpha inhibitors. Travel Med Infect Dis 2012; 3:105-9. [PMID: 17292013 DOI: 10.1016/j.tmaid.2004.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2004] [Accepted: 08/10/2004] [Indexed: 01/06/2023]
Abstract
The TNF-alpha inhibitors, infliximab (Remicade), etanercept (Enbrel) and adalimumab (Humira) are increasingly being used for chronic inflammatory diseases and have effectively allowed many patients to return to more active lifestyles. The impact of these agents on immune function has led to the recognition of specific infectious complications. The combination of the increasingly active lifestyles of those receiving these agents and their immunosuppressive effects pose new challenges for practitioners of travel medicine. The purpose of this article is to review the immunologic and infectious consequences of these therapies and their potential impact on travelers.
Collapse
Affiliation(s)
- Robert Orenstein
- Divisions of General Internal Medicine and Infectious Diseases, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
39
|
Delaby A, Gorvel L, Espinosa L, Lépolard C, Raoult D, Ghigo E, Capo C, Mege JL. Defective monocyte dynamics in Q fever granuloma deficiency. J Infect Dis 2012; 205:1086-94. [PMID: 22351939 DOI: 10.1093/infdis/jis013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The outcome of Q fever, an infectious disease caused by Coxiella burnetii, is associated with granuloma formation. Granulomas are present in patients with resolutive Q fever but are lacking in patients with chronic Q fever. METHODS Study of granuloma formation requires invasive approaches. Here, we took advantage of a recently described method that enables in vitro generation of human granulomas specific for C. burnetii. RESULTS Circulating mononuclear cells progressively accumulated around beads coated with C. burnetii extracts, and complete granulomas were generated in 8 days. Granuloma cells consisted of macrophages, lymphocytes, and, to a lesser extent, epithelioid cells and multinucleated giant cells. Early events that govern granuloma formation were studied using live-imaging microscopy. Monocytes migrated toward C. burnetii-coated beads independently of the presence of T lymphocytes and then recruited T lymphocytes. About 90% of patients with chronic Q fever failed to form granulomas. This deficiency was associated with defective migration of monocytes toward coated beads. CONCLUSIONS Monocytes were involved in the early stages of granuloma formation and recruited T lymphocytes to complete granuloma formation. This article describes a direct relationship between defective granuloma formation and defective migration of monocytes.
Collapse
Affiliation(s)
- Amélie Delaby
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses Transmissibles et Emergentes, CNRS-IRD UMR 6236, Faculté de Médecine, Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
40
|
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
|
41
|
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: 1.8] [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
|
42
|
Role of innate and adaptive immunity in the control of Q fever. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 984:273-86. [PMID: 22711637 DOI: 10.1007/978-94-007-4315-1_14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Acute Q fever is commonly resolved without an antibiotic regimen, but a primary infection may develop into a chronic infection in a minority of cases. Coxiella burnetii, the causative agent of Q fever, is known to infect macrophages both in vitro and in vivo. It has been observed that the intracellular survival of C. burnetii requires the subversion of the microbicidal properties of macrophages. Adaptive immunity is also essential to cure C. burnetii infection, as demonstrated by clinical studies and animal models. Indeed, the control of infection in patients with primary Q fever involves a systemic cell-mediated immune response and granuloma formation with an essential role for interferon-γ in the protection against C. burnetii. In contrast, chronic Q fever is characterized by defective cell-mediated immunity with the defective formation of granulomas and over-production of interleukin-10, an immunoregulatory cytokine. Finally, epidemiological data demonstrate that age and gender are risk factors for Q fever. The analysis of gene expression programs in mice reveals the importance of sex-related genes in C. burnetii infection because only 14% of the modulated genes are sex-independent, while the remaining 86% are differentially expressed in males and females. These results open a new field to understand how host metabolism controls C. burnetii infection in humans.
Collapse
|
43
|
The Coxiella burnetii parasitophorous vacuole. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 984:141-69. [PMID: 22711631 DOI: 10.1007/978-94-007-4315-1_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Coxiella burnetii is a bacterial intracellular parasite of eucaryotic cells that replicates within a membrane-bound compartment, or "parasitophorous vacuole" (PV). With the exception of human macrophages/monocytes, the consensus model of PV trafficking in host cells invokes endolysosomal maturation culminating in lysosome fusion. C. burnetii resists the degradative functions of the vacuole while at the same time exploiting the acidic pH for metabolic activation. While at first glance the mature PV resembles a large phagolysosome, an increasing body of evidence indicates the vacuole is in fact a specialized compartment that is actively modified by the pathogen. Adding to the complexity of PV biogenesis is new data showing vacuole engagement with autophagic and early secretory pathways. In this chapter, we review current knowledge of PV nature and development, and discuss disparate data related to the ultimate maturation state of PV harboring virulent or avirulent C. burnetii lipopolysaccharide phase variants in human mononuclear phagocytes.
Collapse
|
44
|
Wang Y, Xiong X, Wu D, Wang X, Wen B. Efficient activation of T cells by human monocyte-derived dendritic cells (HMDCs) pulsed with Coxiella burnetii outer membrane protein Com1 but not by HspB-pulsed HMDCs. BMC Immunol 2011; 12:52. [PMID: 21888659 PMCID: PMC3179940 DOI: 10.1186/1471-2172-12-52] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 09/03/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Coxiella burnetii is an obligate intracellular bacterium and the etiologic agent of Q fever; both coxiella outer membrane protein 1 (Com1) and heat shock protein B (HspB) are its major immunodominant antigens. It is not clear whether Com1 and HspB have the ability to mount immune responses against C. burnetii infection. RESULTS The recombinant proteins Com1 and HspB were applied to pulse human monocyte-derived dendritic cells (HMDCs), and the pulsed HMDCs were used to stimulate isogenic T cells. Com1-pulsed HMDCs expressed substantially higher levels of surface molecules (CD83, CD40, CD80, CD86, CD54, and CD58) and a higher level of interleukin-12 than HspB-pulsed HMDCs. Moreover, Com1-pulsed HMDCs induced high-level proliferation and activation of CD4(+) and CD8(+) cells, which expressed high levels of T-cell activation marker CD69 and inflammatory cytokines IFN-γ and TNF-α. In contrast, HspB-pulsed HMDCs were unable to induce efficient T-cell proliferation and activation. CONCLUSIONS Our results demonstrate that Com1-pulsed HMDCs are able to induce efficient T-cell proliferation and drive T cells toward Th1 and Tc1 polarization; however, HspB-pulsed HMDCs are unable to do so. Unlike HspB, Com1 is a protective antigen, which was demonstrated by the adoptive transfer of Com1-pulsed bone marrow dendritic cells into naive BALB/c mice.
Collapse
Affiliation(s)
- Ying Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | | | | | | | | |
Collapse
|
45
|
Abstract
Coxiella burnetii is the causative agent of Q fever, a disease with a spectrum of presentations from the mild to fatal, including chronic sequelae. Since its discovery in 1935, it has been shown to infect a wide range of hosts, including humans. A recent outbreak in Europe reminds us that this is still a significant pathogen of concern, very transmissible and with a very low infectious dose. For these reasons it has also featured regularly on various threat lists, as it may be considered by the unscrupulous for use as a bioweapon. As an intracellular pathogen, it has remained an enigmatic organism due to the inability to culture it on laboratory media. As a result, interactions with the host have been difficult to elucidate and we still have a very limited understanding of the molecular mechanisms of virulence. However, two recent developments will open up our understanding of C. burnetii: the first axenic growth medium capable of supporting cell-free growth, and the production of the first isogenic mutant. We are approaching an exciting time for expanding our knowledge of this organism in the next few years.
Collapse
Affiliation(s)
- P. C. F. Oyston
- Biomedical Sciences, Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| | - C. Davies
- Biomedical Sciences, Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| |
Collapse
|
46
|
Sukocheva OA, Marmion BP, Storm PA, Lockhart M, Turra M, Graves S. Long-term persistence after acute Q fever of non-infective Coxiella burnetii cell components, including antigens. QJM 2010; 103:847-63. [PMID: 20639288 DOI: 10.1093/qjmed/hcq113] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Previous studies of inciting factors for a prolonged post-infection fatigue syndrome after Q fever (variously termed QFS or Q fever associated CFS/ME in the literature) showed that after the acute infection a high proportion of asymptomatic and QFS patients had Q fever antibody and also low levels in PBMC and bone marrow of Coxiella burnetii (C.b.) DNA with PCR assays directed against three different target sequences in different parts of the coxiella genome. Attempts to isolate a strain of C.b. in A/J mice, and cell culture from PCR positive PBMC and bone marrow were consistently negative. The detailed composition of the persisting coxiella residues remains to be defined. AIM To retest and provide detailed results on selected PCR positive samples from the Birmingham Q fever outbreak patients tested by a highly sensitive method to detect viable organisms and to determine the nature of the residual coxiella cell components. DESIGN Laboratory case study. METHODS NOD/SCID mice were inoculated with samples from the 1989 Q fever outbreak in Birmingham and followed for evidence of infection and the presence of coxiella DNA and specific antigens in spleen and liver macrophages. A significant, unexpected finding of specific antigen was followed by assessment of its ability to provoke production of inflammatory and non-inflammatory cytokines in mice, in THP-1 human macrophage cell cultures and to induce inflammatory lesions in the skin of guinea pigs hyperimmunized against Q fever vaccine. RESULTS Culture of samples from 10 Birmingham Q fever patients in NOD/SCID mice, 12 years from infection did not yield viable Coxiella burnetii, as shown earlier. However complexes of material with coxiella antigens were found in mouse spleens in all cases but in significantly greater amounts in samples from those with post Q fever fatigue syndrome. The antigenic complexes [now designated 'immunomodulatory complexes' (IMC)] were shown to stimulate cytokine release in the mice and in the THP-1 macrophages and to provoke an inflammatory reaction on intradermal injection into the skin of Q fever hyperimmunized guinea pigs. CONCLUSION The study identifies a non-infective complex of C.b. antigens able to survive in the host and provoke aberrant humoral and cell medicated immunity responses - a possible pathogenic link between initial infection and a subsequent long-term post Q fever fatigue syndrome.
Collapse
Affiliation(s)
- O A Sukocheva
- Q fever Research Group, Hanson Institute, Adelaide, South Australia
| | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Coxiella burnetii phase I and II variants replicate with similar kinetics in degradative phagolysosome-like compartments of human macrophages. Infect Immun 2010; 78:3465-74. [PMID: 20515926 DOI: 10.1128/iai.00406-10] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Coxiella burnetii infects mononuclear phagocytes, where it directs biogenesis of a vacuolar niche termed the parasitophorous vacuole (PV). Owing to its lumenal pH (approximately 5) and fusion with endolysosomal vesicles, the PV is considered phagolysosome-like. However, the degradative properties of the mature PV are unknown, and there are conflicting reports on the maturation state and growth permissiveness of PV harboring virulent phase I or avirulent phase II C. burnetii variants in human mononuclear phagocytes. Here, we employed infection of primary human monocyte-derived macrophages (HMDMs) and THP-1 cells as host cells to directly compare the PV maturation kinetics and pathogen growth in cells infected with the Nine Mile phase I variant (NMI) or phase II variant (NMII) of C. burnetii. In both cell types, phase variants replicated with similar kinetics, achieving roughly 2 to 3 log units of growth before they reached stationary phase. HMDMs infected by either phase variant secreted similar amounts of the proinflammatory cytokines interleukin-6 and tumor necrosis factor alpha. In infected THP-1 cells, equal percentages of NMI and NMII PVs decorate with the early endosomal marker Rab5, the late endosomal/lysosomal markers Rab7 and CD63, and the lysosomal marker cathepsin D at early (8 h) and late (72 h) time points postinfection (p.i.). Mature PVs (2 to 4 days p.i.) harboring NMI or NMII contained proteolytically active cathepsins and quickly degraded Escherichia coli. These data suggest that C. burnetii does not actively inhibit phagolysosome function as a survival mechanism. Instead, NMI and NMII resist degradation to replicate in indistinguishable digestive PVs that fully mature through the endolysosomal pathway.
Collapse
|
49
|
Angelakis E, Raoult D. Q fever. Vet Microbiol 2010; 140:297-309. [DOI: 10.1016/j.vetmic.2009.07.016] [Citation(s) in RCA: 434] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Revised: 07/03/2009] [Accepted: 07/30/2009] [Indexed: 01/17/2023]
|
50
|
Dual High Titer Antineutrophil Cytoplasmic Autoantibodies in Association With Systemic Q Fever. J Clin Rheumatol 2009; 15:411-3. [PMID: 19956002 DOI: 10.1097/rhu.0b013e3181c3f8a8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|