1
|
Nakamura T, Shimizu T, Nishinakama N, Takahashi R, Arasaki K, Uda A, Watanabe K, Watarai M. A novel method of Francisella infection of epithelial cells using HeLa cells expressing fc gamma receptor. BMC Infect Dis 2024; 24:1171. [PMID: 39420255 PMCID: PMC11488177 DOI: 10.1186/s12879-024-10083-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Francisella tularensis, the causative agent of tularemia, is a facultative intracellular bacterium. Although the life cycle of this bacterium inside phagocytic cells (e.g., macrophages, neutrophils) has been well analyzed, the difficulty of gene silencing and editing genes in phagocytic cells makes it difficult to analyze host factors important for the infection. On the other hand, epithelial cell lines, such as HeLa, have been established as cell lines that are easy to perform gene editing. However, the infection efficiency of Francisella into these epithelial cells is extremely low. METHODS In order to facilitate the molecular biological analysis of Francisella infection using epithelial cells, we constructed an efficient infection model of F. tularensis subsp. novicida (F. novicida) in HeLa cells expressing mouse FcγRII (HeLa-FcγRII), and the system was applied to evaluate the role of host GLS1 on Francisella infection. RESULTS As a result of colony forming unit count, HeLa-FcγRII cells uptake F. novicida in a serum-dependent manner and demonstrated an approximately 100-fold increase in intracellular bacterial infection compared to parental HeLa cells. Furthermore, taking advantage of the gene silencing capability of HeLa-FcγRII cells, we developed GLS1, a gene encoding glutaminase, knockdown cells using lentiviral sh RNA vector and assessed the impact of GLS1 on F. novicida infection. LDH assay revealed that GLS1-knockdown HeLa-FcγRII cells exhibited increased cytotoxicity during infection with F. novicida compared with control HeLa-FcγRII cells. Furthermore, the cell death was inhibited by the addition of ammonia, the metabolite produced through glutaminase activity. These results suggest that ammonia plays an important role in the proliferation of F. novicida. CONCLUSIONS In this report, we proposed a new cell-based infection system for Francisella infection using HeLa-FcγRII cells and demonstrated its effectiveness. This system has the potential to accelerate cell-based infection assays, such as large-scale genetic screening, and to provide new insights into Francisella infection in epithelial cells, which has been difficult to analyze in phagocytic cells.
Collapse
Affiliation(s)
- Takemasa Nakamura
- Joint Faculty of Veterinary Medicine, Laboratory of Veterinary Public Health, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan
| | - Takashi Shimizu
- Joint Faculty of Veterinary Medicine, Laboratory of Veterinary Public Health, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan
| | - Naho Nishinakama
- Joint Faculty of Veterinary Medicine, Laboratory of Veterinary Public Health, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan
| | - Reika Takahashi
- Joint Faculty of Veterinary Medicine, Laboratory of Veterinary Public Health, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan
| | - Kohei Arasaki
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Akihiko Uda
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo, 162-8640, Japan
| | - Kenta Watanabe
- Joint Faculty of Veterinary Medicine, Laboratory of Veterinary Public Health, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan
| | - Masahisa Watarai
- Joint Faculty of Veterinary Medicine, Laboratory of Veterinary Public Health, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan.
| |
Collapse
|
2
|
Kurtz SL, De Pascalis R, Meierovics AI, Elkins KL. Deficiency in CCR2 increases susceptibility of mice to infection with an intracellular pathogen, Francisella tularensis LVS, but does not impair development of protective immunity. PLoS One 2021; 16:e0249142. [PMID: 33760886 PMCID: PMC7990183 DOI: 10.1371/journal.pone.0249142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/11/2021] [Indexed: 12/24/2022] Open
Abstract
CCR2 is the major chemokine receptor that regulates appropriate trafficking of inflammatory monocytes, but the role of this chemokine receptor and its ligands during primary and secondary infection with intracellular infections remains incompletely understood. Here we used murine infection with the Live Vaccine Strain (LVS) of Francisella tularensis to evaluate the role of CCR2 during primary and secondary parenteral responses to this prototype intracellular bacterium. We find that mice deficient in CCR2 are highly compromised in their ability to survive intradermal infection with LVS, indicating the importance of this receptor during primary parenteral responses. Interestingly, this defect could not be readily attributed to the activities of the known murine CCR2 ligands MCP-1/CCL2, MCP-3/CCL7, or MCP-5/CCL12. Nonetheless, CCR2 knockout mice vaccinated by infection with low doses of LVS generated optimal T cell responses that controlled the intramacrophage replication of Francisella, and LVS-immune CCR2 knockout mice survived maximal lethal Francisella challenge. Thus, fully protective adaptive immune memory responses to this intracellular bacterium can be readily generated in the absence of CCR2.
Collapse
Affiliation(s)
- Sherry L. Kurtz
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
- * E-mail: (KLE); (SLK)
| | - Roberto De Pascalis
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
| | - Anda I. Meierovics
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
| | - Karen L. Elkins
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
- * E-mail: (KLE); (SLK)
| |
Collapse
|
3
|
Hunt D, Drake LA, Drake JR. Murine macrophage TLR2-FcγR synergy via FcγR licensing of IL-6 cytokine mRNA ribosome binding and translation. PLoS One 2018; 13:e0200764. [PMID: 30024985 PMCID: PMC6053178 DOI: 10.1371/journal.pone.0200764] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/02/2018] [Indexed: 12/18/2022] Open
Abstract
Macrophages (MØs) are sentinels of the immune system that use pattern recognition receptors such as Toll-like receptors (TLR) to detect invading pathogens and immune receptors such as FcγR to sense the host’s immune state. Crosstalk between these two signaling pathways allows the MØ to tailor the cell’s overall response to prevailing conditions. However, the molecular mechanisms underlying TLR-FcγR crosstalk are only partially understood. Therefore, we employed an immunologically-relevant MØ stimulus, an inactivated gram-negative bacterium that bears TLR2 agonists but no TLR4 agonist (iBTLR2) opsonized with a monoclonal antibody (mAb-iBTLR2), as a tool to study FcγR regulation of TLR2-driven production of IL-6, a key inflammatory cytokine. We chose this particular agonist as an investigational tool because MØ production of any detectable IL-6 in response to mAb-iBTLR2 requires both TLR2 and FcγR signaling, making it an excellent system for the study of receptor synergy. Using genetic, pharmacological and immunological approaches, we demonstrate that the murine MØ IL-6 response to mAb-iBTLR2 requires activation of both the TLR/NF-κB and FcγR/ITAM signaling pathways. mAb-iBTLR2 engagement of TLR2 drives NF-κB activation and up-regulation of IL-6 mRNA but fails to result in IL-6 cytokine production/release. Here, Src family kinase-driven FcγR ITAM signaling is necessary to enable IL-6 mRNA incorporation into polysomes and translation. These results reveal a novel mechanism by which FcγR ITAM signaling synergizes with TLR signaling, by “licensing” cytokine mRNA ribosome binding/translation to drive a strong murine MØ cytokine response.
Collapse
Affiliation(s)
- Danielle Hunt
- Albany Medical College, Department of Immunology and Microbial Disease, Albany, NY, United States of America
| | - Lisa A. Drake
- Albany Medical College, Department of Immunology and Microbial Disease, Albany, NY, United States of America
| | - James R. Drake
- Albany Medical College, Department of Immunology and Microbial Disease, Albany, NY, United States of America
- * E-mail:
| |
Collapse
|
4
|
Holland KM, Rosa SJ, Kristjansdottir K, Wolfgeher D, Franz BJ, Zarrella TM, Kumar S, Sunagar R, Singh A, Bakshi CS, Namjoshi P, Barry EM, Sellati TJ, Kron SJ, Gosselin EJ, Reed DS, Hazlett KRO. Differential Growth of Francisella tularensis, Which Alters Expression of Virulence Factors, Dominant Antigens, and Surface-Carbohydrate Synthases, Governs the Apparent Virulence of Ft SchuS4 to Immunized Animals. Front Microbiol 2017; 8:1158. [PMID: 28690600 PMCID: PMC5479911 DOI: 10.3389/fmicb.2017.01158] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 06/07/2017] [Indexed: 12/29/2022] Open
Abstract
The gram-negative bacterium Francisella tularensis (Ft) is both a potential biological weapon and a naturally occurring microbe that survives in arthropods, fresh water amoeba, and mammals with distinct phenotypes in various environments. Previously, we used a number of measurements to characterize Ft grown in Brain-Heart Infusion (BHI) broth as (1) more similar to infection-derived bacteria, and (2) slightly more virulent in naïve animals, compared to Ft grown in Mueller Hinton Broth (MHB). In these studies we observed that the free amino acids in MHB repress expression of select Ft virulence factors by an unknown mechanism. Here, we tested the hypotheses that Ft grown in BHI (BHI-Ft) accurately displays a full protein composition more similar to that reported for infection-derived Ft and that this similarity would make BHI-Ft more susceptible to pre-existing, vaccine-induced immunity than MHB-Ft. We performed comprehensive proteomic analysis of Ft grown in MHB, BHI, and BHI supplemented with casamino acids (BCA) and compared our findings to published “omics” data derived from Ft grown in vivo. Based on the abundance of ~1,000 proteins, the fingerprint of BHI-Ft is one of nutrient-deprived bacteria that—through induction of a stringent-starvation-like response—have induced the FevR regulon for expression of the bacterium's virulence factors, immuno-dominant antigens, and surface-carbohydrate synthases. To test the notion that increased abundance of dominant antigens expressed by BHI-Ft would render these bacteria more susceptible to pre-existing, vaccine-induced immunity, we employed a battery of LVS-vaccination and S4-challenge protocols using MHB- and BHI-grown Ft S4. Contrary to our hypothesis, these experiments reveal that LVS-immunization provides a barrier to infection that is significantly more effective against an MHB-S4 challenge than a BHI-S4 challenge. The differences in apparent virulence to immunized mice are profoundly greater than those observed with primary infection of naïve mice. Our findings suggest that tularemia vaccination studies should be critically evaluated in regard to the growth conditions of the challenge agent.
Collapse
Affiliation(s)
- Kristen M Holland
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Sarah J Rosa
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | | | - Donald Wolfgeher
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicago, IL, United States
| | - Brian J Franz
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Tiffany M Zarrella
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Raju Sunagar
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Anju Singh
- Trudeau InstituteSaranac Lake, NY, United States
| | - Chandra S Bakshi
- Department of Microbiology and Immunology, New York Medical CollegeValhalla, NY, United States
| | - Prachi Namjoshi
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Eileen M Barry
- School of Medicine, University of MarylandBaltimore, MD, United States
| | | | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, University of ChicagoChicago, IL, United States
| | - Edmund J Gosselin
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| | - Douglas S Reed
- Center for Vaccine Research, University of PittsburghPittsburgh, PA, United States
| | - Karsten R O Hazlett
- Department of Immunology and Microbial Disease, Albany Medical CollegeAlbany, NY, United States
| |
Collapse
|
5
|
Kumar S, Sunagar R, Pham G, Franz BJ, Rosa SJ, Hazlett KRO, Gosselin EJ. Differential Cultivation of Francisella tularensis Induces Changes in the Immune Response to and Protective Efficacy of Whole Cell-Based Inactivated Vaccines. Front Immunol 2017; 7:677. [PMID: 28119692 PMCID: PMC5222797 DOI: 10.3389/fimmu.2016.00677] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/21/2016] [Indexed: 01/06/2023] Open
Abstract
Francisella tularensis (Ft) is a category A biothreat agent for which there is no Food and Drug Administration-approved vaccine. Ft can survive in a variety of habitats with a remarkable ability to adapt to changing environmental conditions. Furthermore, Ft expresses distinct sets of antigens (Ags) when inside of macrophages (its in vivo host) as compared to those grown in vitro with Mueller Hinton Broth (MHB). However, in contrast to MHB-grown Ft, Ft grown in Brain-Heart Infusion (BHI) more closely mimics the antigenic profile of macrophage-grown Ft. Thus, we anticipated that when used as a vaccine, BHI-grown Ft would provide better protection compared to MHB-grown Ft, primarily due to its greater antigenic similarity to Ft circulating inside the host (macrophages) during natural infection. Our investigation, however, revealed that inactivated Ft (iFt) grown in MHB (iFt-MHB) exhibited superior protective activity when used as a vaccine, as compared to iFt grown in BHI (iFt-BHI). The superior protection afforded by iFt-MHB compared to that of iFt-BHI was associated with significantly lower bacterial burden and inflammation in the lungs and spleens of vaccinated mice. Moreover, iFt-MHB also induced increased levels of Ft-specific IgG. Further evaluation of early immunological cues also revealed that iFt-MHB exhibits increased engagement of Ag-presenting cells including increased iFt binding to dendritic cells, increased expression of costimulatory markers, and increased secretion of pro-inflammatory cytokines. Importantly, these studies directly demonstrate that Ft growth conditions strongly impact Ft vaccine efficacy and that the growth medium used to produce whole cell vaccines to Ft must be a key consideration in the development of a tularemia vaccine.
Collapse
Affiliation(s)
- Sudeep Kumar
- Center for Immunology and Microbial Diseases, Albany Medical College , Albany, NY , USA
| | - Raju Sunagar
- Center for Immunology and Microbial Diseases, Albany Medical College , Albany, NY , USA
| | - Giang Pham
- Center for Immunology and Microbial Diseases, Albany Medical College , Albany, NY , USA
| | - Brian J Franz
- Center for Immunology and Microbial Diseases, Albany Medical College , Albany, NY , USA
| | - Sarah J Rosa
- Center for Immunology and Microbial Diseases, Albany Medical College , Albany, NY , USA
| | - Karsten R O Hazlett
- Center for Immunology and Microbial Diseases, Albany Medical College , Albany, NY , USA
| | - Edmund J Gosselin
- Center for Immunology and Microbial Diseases, Albany Medical College , Albany, NY , USA
| |
Collapse
|
6
|
Holland KM, Rosa SJ, Hazlett KRO. Francisella tularensis - Immune Cell Activator, Suppressor, or Stealthy Evader: The Evolving View from the Petri Dish. ACTA ACUST UNITED AC 2016; 7. [PMID: 27695643 PMCID: PMC5042348 DOI: 10.4172/2157-2526.1000144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
One of the hallmarks of pulmonary tularemia, which results from inhalation of Francisella tularensis - a significant bioterrorism concern, is the lack of an acute TH1-biased inflammatory response in the early phase of disease (days 1–3) despite significant bacterial loads. In an effort to understand this apparent hypo-responsiveness, many laboratories have utilized in vitro cell-based models as tools to probe the nature and consequences of host cell interactions with F. tularensis. The first uses of this model suggested that mammalian host cells recognize this bacterium principally through TLR2 to evoke a robust, classical TH1-biased cytokine response including TNF, IL-6, IL-1β, and IFN-γ. Others used this model in concert with a variety of non-genetic perturbations of the bacterial-host cell interaction and suggested that F. tularensis actively-suppressed the cellular response. Consistent with this notion, others engaged this model to assess isogenic mutants and, in many cases, found the mutant bacteria to be more pro-inflammatory than their WT counter-parts. Frequently, these observations were interpreted as evidence for the immunosuppressive function of the gene of interest. However, recently appreciated roles of the health of the bacterium and the impact of host factors have refined this model to suggest a “stealthy” mode of bacterial-host cell interaction (rather than one involving active suppression) consistent with the observations during early phase disease.
Collapse
Affiliation(s)
- Kristen M Holland
- Center for Immunology & Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | - Sarah J Rosa
- Center for Immunology & Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| | - Karsten R O Hazlett
- Center for Immunology & Microbial Disease, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
7
|
Abstract
Francisella tularensis (Ft) is a gram-negative intercellular pathogen and category A biothreat agent. However, despite 15 years of strong government investment and intense research focused on the development of a US Food and Drug Administration-approved vaccine against Ft, the primary goal remains elusive. This article reviews research efforts focused on developing an Ft vaccine, as well as a number of important factors, some only recently recognized as such, which can significantly impact the development and evaluation of Ft vaccine efficacy. Finally, an assessment is provided as to whether a US Food and Drug Administration-approved Ft vaccine is likely to be forthcoming and the potential means by which this might be achieved.
Collapse
Affiliation(s)
- Raju Sunagar
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Sudeep Kumar
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Brian J Franz
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Edmund J Gosselin
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| |
Collapse
|
8
|
Steiner DJ, Furuya Y, Metzger DW. Host-pathogen interactions and immune evasion strategies in Francisella tularensis pathogenicity. Infect Drug Resist 2014; 7:239-51. [PMID: 25258544 PMCID: PMC4173753 DOI: 10.2147/idr.s53700] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Francisella tularensis is an intracellular Gram-negative bacterium that causes life-threatening tularemia. Although the prevalence of natural infection is low, F. tularensis remains a tier I priority pathogen due to its extreme virulence and ease of aerosol dissemination. F. tularensis can infect a host through multiple routes, including the intradermal and respiratory routes. Respiratory infection can result from a very small inoculum (ten organisms or fewer) and is the most lethal form of infection. Following infection, F. tularensis employs strategies for immune evasion that delay the immune response, permitting systemic distribution and induction of sepsis. In this review we summarize the current knowledge of F. tularensis in an immunological context, with emphasis on the host response and bacterial evasion of that response.
Collapse
Affiliation(s)
- Don J Steiner
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Yoichi Furuya
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Dennis W Metzger
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| |
Collapse
|
9
|
Edwards MW, Aultman JA, Harber G, Bhatt JM, Sztul E, Xu Q, Zhang P, Michalek SM, Katz J. Role of mTOR downstream effector signaling molecules in Francisella tularensis internalization by murine macrophages. PLoS One 2013; 8:e83226. [PMID: 24312679 PMCID: PMC3849438 DOI: 10.1371/journal.pone.0083226] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 11/11/2013] [Indexed: 02/03/2023] Open
Abstract
Francisella tularensis is an infectious, gram-negative, intracellular microorganism, and the cause of tularemia. Invasion of host cells by intracellular pathogens like Francisella is initiated by their interaction with different host cell membrane receptors and the rapid phosphorylation of different downstream signaling molecules. PI3K and Syk have been shown to be involved in F. tularensis host cell entry, and both of these signaling molecules are associated with the master regulator serine/threonine kinase mTOR; yet the involvement of mTOR in F. tularensis invasion of host cells has not been assessed. Here, we report that infection of macrophages with F. tularensis triggers the phosphorylation of mTOR downstream effector molecules, and that signaling via TLR2 is necessary for these events. Inhibition of mTOR or of PI3K, ERK, or p38, but not Akt signaling, downregulates the levels of phosphorylation of mTOR downstream targets, and significantly reduces the number of F. tularensis cells invading macrophages. Moreover, while phosphorylation of mTOR downstream effectors occurs via the PI3K pathway, it also involves PLCγ1 and Ca(2+) signaling. Furthermore, abrogation of PLC or Ca(2+) signaling revealed their important role in the ability of F. tularensis to invade host cells. Together, these findings suggest that F. tularensis invasion of primary macrophages utilize a myriad of host signaling pathways to ensure effective cell entry.
Collapse
Affiliation(s)
- Michael W. Edwards
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - James A. Aultman
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Gregory Harber
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jay M. Bhatt
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Elizabeth Sztul
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Qingan Xu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ping Zhang
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Suzanne M. Michalek
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jannet Katz
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
10
|
Singh A, Rahman T, Malik M, Hickey AJ, Leifer CA, Hazlett KRO, Sellati TJ. Discordant results obtained with Francisella tularensis during in vitro and in vivo immunological studies are attributable to compromised bacterial structural integrity. PLoS One 2013; 8:e58513. [PMID: 23554897 PMCID: PMC3595284 DOI: 10.1371/journal.pone.0058513] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 02/05/2013] [Indexed: 11/18/2022] Open
Abstract
Francisella tularensis (Ft) is a highly infectious intracellular pathogen and the causative agent of tularemia. Because Ft can be dispersed via small droplet-aerosols and has a very low infectious dose it is characterized as a category A Select Agent of biological warfare. Respiratory infection with the attenuated Live Vaccine Strain (LVS) and the highly virulent SchuS4 strain of Ft engenders intense peribronchiolar and perivascular inflammation, but fails to elicit select pro-inflammatory mediators (e.g., TNF, IL-1β, IL-6, IL-12, and IFN-γ) within the first ∼72 h. This in vivo finding is discordant with the principally TH1-oriented response to Ft frequently observed in cell-based studies wherein the aforementioned cytokines are produced. An often overlooked confounding factor in the interpretation of experimental results is the influence of environmental cues on the bacterium's capacity to elicit certain host responses. Herein, we reveal that adaptation of Ft to its mammalian host imparts an inability to elicit select pro-inflammatory mediators throughout the course of infection. Furthermore, in vitro findings that non-host adapted Ft elicits such a response from host cells reflect aberrant recognition of the DNA of structurally-compromised bacteria by AIM2-dependent and -independent host cell cytosolic DNA sensors. Growth of Ft in Muller-Hinton Broth or on Muller-Hinton-based chocolate agar plates or genetic mutation of Ft was found to compromise the structural integrity of the bacterium thus rendering it capable of aberrantly eliciting pro-inflammatory mediators (e.g., TNF, IL-1β, IL-6, IL-12, and IFN-γ). Our studies highlight the profound impact of different growth conditions on host cell response to infection and demonstrate that not all in vitro-derived findings may be relevant to tularemia pathogenesis in the mammalian host. Rational development of a vaccine and immunotherapeutics can only proceed from a foundation of knowledge based upon in vitro findings that recapitulate those observed during natural infection.
Collapse
Affiliation(s)
- Anju Singh
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Tabassum Rahman
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Meenakshi Malik
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Anthony J. Hickey
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Cynthia A. Leifer
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, New York, United States of America
| | - Karsten R. O. Hazlett
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Timothy J. Sellati
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
- * E-mail:
| |
Collapse
|
11
|
Schmitt DM, O'Dee DM, Horzempa J, Carlson PE, Russo BC, Bales JM, Brown MJ, Nau GJ. A Francisella tularensis live vaccine strain that improves stimulation of antigen-presenting cells does not enhance vaccine efficacy. PLoS One 2012; 7:e31172. [PMID: 22355343 PMCID: PMC3280287 DOI: 10.1371/journal.pone.0031172] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 01/04/2012] [Indexed: 11/25/2022] Open
Abstract
Vaccination is a proven strategy to mitigate morbidity and mortality of infectious diseases. The methodology of identifying and testing new vaccine candidates could be improved with rational design and in vitro testing prior to animal experimentation. The tularemia vaccine, Francisella tularensis live vaccine strain (LVS), does not elicit complete protection against lethal challenge with a virulent type A Francisella strain. One factor that may contribute to this poor performance is limited stimulation of antigen-presenting cells. In this study, we examined whether the interaction of genetically modified LVS strains with human antigen-presenting cells correlated with effectiveness as tularemia vaccine candidates. Human dendritic cells infected with wild-type LVS secrete low levels of proinflammatory cytokines, fail to upregulate costimulatory molecules, and activate human T cells poorly in vitro. One LVS mutant, strain 13B47, stimulated higher levels of proinflammatory cytokines from dendritic cells and macrophages and increased costimulatory molecule expression on dendritic cells compared to wild type. Additionally, 13B47-infected dendritic cells activated T cells more efficiently than LVS-infected cells. A deletion allele of the same gene in LVS displayed similar in vitro characteristics, but vaccination with this strain did not improve survival after challenge with a virulent Francisella strain. In vivo, this mutant was attenuated for growth and did not stimulate T cell responses in the lung comparable to wild type. Therefore, stimulation of antigen-presenting cells in vitro was improved by genetic modification of LVS, but did not correlate with efficacy against challenge in vivo within this model system.
Collapse
Affiliation(s)
- Deanna M. Schmitt
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Dawn M. O'Dee
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Joseph Horzempa
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Natural Sciences and Mathematics, West Liberty University, West Liberty, West Virginia, United States of America
| | - Paul E. Carlson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Brian C. Russo
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jacqueline M. Bales
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Matthew J. Brown
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Gerard J. Nau
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Medicine – Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
12
|
Zarrella TM, Singh A, Bitsaktsis C, Rahman T, Sahay B, Feustel PJ, Gosselin EJ, Sellati TJ, Hazlett KRO. Host-adaptation of Francisella tularensis alters the bacterium's surface-carbohydrates to hinder effectors of innate and adaptive immunity. PLoS One 2011; 6:e22335. [PMID: 21799828 PMCID: PMC3142145 DOI: 10.1371/journal.pone.0022335] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 06/27/2011] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The gram-negative bacterium Francisella tularensis survives in arthropods, fresh water amoeba, and mammals with both intracellular and extracellular phases and could reasonably be expected to express distinct phenotypes in these environments. The presence of a capsule on this bacterium has been controversial with some groups finding such a structure while other groups report that no capsule could be identified. Previously we reported in vitro culture conditions for this bacterium which, in contrast to typical methods, yielded a bacterial phenotype that mimics that of the bacterium's mammalian, extracellular phase. METHODS/FINDINGS SDS-PAGE and carbohydrate analysis of differentially-cultivated F. tularensis LVS revealed that bacteria displaying the host-adapted phenotype produce both longer polymers of LPS O-antigen (OAg) and additional HMW carbohydrates/glycoproteins that are reduced/absent in non-host-adapted bacteria. Analysis of wildtype and OAg-mutant bacteria indicated that the induced changes in surface carbohydrates involved both OAg and non-OAg species. To assess the impact of these HMW carbohydrates on the access of outer membrane constituents to antibody we used differentially-cultivated bacteria in vitro to immunoprecipitate antibodies directed against outer membrane moieties. We observed that the surface-carbohydrates induced during host-adaptation shield many outer membrane antigens from binding by antibody. Similar assays with normal mouse serum indicate that the induced HMW carbohydrates also impede complement deposition. Using an in vitro macrophage infection assay, we find that the bacterial HMW carbohydrate impedes TLR2-dependent, pro-inflammatory cytokine production by macrophages. Lastly we show that upon host-adaptation, the human-virulent strain, F. tularensis SchuS4 also induces capsule production with the effect of reducing macrophage-activation and accelerating tularemia pathogenesis in mice. CONCLUSION F. tularensis undergoes host-adaptation which includes production of multiple capsular materials. These capsules impede recognition of bacterial outer membrane constituents by antibody, complement, and Toll-Like Receptor 2. These changes in the host-pathogen interface have profound implications for pathogenesis and vaccine development.
Collapse
Affiliation(s)
- Tiffany M. Zarrella
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Anju Singh
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Constantine Bitsaktsis
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Tabassum Rahman
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Bikash Sahay
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Paul J. Feustel
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York, United States of America
| | - Edmund J. Gosselin
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Timothy J. Sellati
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Karsten R. O. Hazlett
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| |
Collapse
|
13
|
Periasamy S, Singh A, Sahay B, Rahman T, Feustel PJ, Pham GH, Gosselin EJ, Sellati TJ. Development of tolerogenic dendritic cells and regulatory T cells favors exponential bacterial growth and survival during early respiratory tularemia. J Leukoc Biol 2011; 90:493-507. [PMID: 21724804 DOI: 10.1189/jlb.0411197] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Tularemia is a vector-borne zoonosis caused by Ft, a Gram-negative, facultative intracellular bacterium. Ft exists in two clinically relevant forms, the European biovar B (holarctica), which produces acute, although mild, self-limiting infections, and the more virulent United States biovar A (tularensis), which is often associated with pneumonic tularemia and more severe disease. In a mouse model of tularemia, respiratory infection with the virulence-attenuated Type B (LVS) or highly virulent Type A (SchuS4) strain engenders peribronchiolar and perivascular inflammation. Paradoxically, despite an intense neutrophilic infiltrate and high bacterial burden, T(h)1-type proinflammatory cytokines (e.g., TNF, IL-1β, IL-6, and IL-12) are absent within the first ∼72 h of pulmonary infection. It has been suggested that the bacterium has the capacity to actively suppress or block NF-κB signaling, thus causing an initial delay in up-regulation of inflammatory mediators. However, our previously published findings and those presented herein contradict this paradigm and instead, strongly support an alternative hypothesis. Rather than blocking NF-κB, Ft actually triggers TLR2-dependent NF-κB signaling, resulting in the development and activation of tDCs and the release of anti-inflammatory cytokines (e.g., IL-10 and TGF-β). In turn, these cytokines stimulate development and proliferation of T(regs) that may restrain T(h)1-type proinflammatory cytokine release early during tularemic infection. The highly regulated and overall anti-inflammatory milieu established in the lung is permissive for unfettered growth and survival of Ft. The capacity of Ft to evoke such a response represents an important immune-evasive strategy.
Collapse
|
14
|
A Francisella tularensis locus required for spermine responsiveness is necessary for virulence. Infect Immun 2011; 79:3665-76. [PMID: 21670171 DOI: 10.1128/iai.00135-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tularemia is a debilitating febrile illness caused by the category A biodefense agent Francisella tularensis. This pathogen infects over 250 different hosts, has a low infectious dose, and causes high morbidity and mortality. Our understanding of the mechanisms by which F. tularensis senses and adapts to host environments is incomplete. Polyamines, including spermine, regulate the interactions of F. tularensis with host cells. However, it is not known whether responsiveness to polyamines is necessary for the virulence of the organism. Through transposon mutagenesis of F. tularensis subsp. holarctica live vaccine strain (LVS), we identified FTL_0883 as a gene important for spermine responsiveness. In-frame deletion mutants of FTL_0883 and FTT_0615c, the homologue of FTL_0883 in F. tularensis subsp. tularensis Schu S4 (Schu S4), elicited higher levels of cytokines from human and murine macrophages compared to wild-type strains. Although deletion of FTL_0883 attenuated LVS replication within macrophages in vitro, the Schu S4 mutant with a deletion in FTT_0615c replicated similarly to wild-type Schu S4. Nevertheless, both the LVS and the Schu S4 mutants were significantly attenuated in vivo. Growth and dissemination of the Schu S4 mutant was severely reduced in the murine model of pneumonic tularemia. This attenuation depended on host responses to elevated levels of proinflammatory cytokines. These data associate responsiveness to polyamines with tularemia pathogenesis and define FTL_0883/FTT_0615c as an F. tularensis gene important for virulence and evasion of the host immune response.
Collapse
|
15
|
Slight SR, Lin Y, Messmer M, Khader SA. Francisella tularensis LVS-induced Interleukin-12 p40 cytokine production mediates dendritic cell migration through IL-12 Receptor β1. Cytokine 2011; 55:372-9. [PMID: 21669537 DOI: 10.1016/j.cyto.2011.05.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 04/20/2011] [Accepted: 05/18/2011] [Indexed: 12/22/2022]
Abstract
Three cytokines use the IL-12p40 cytokine subunit namely: IL-12p70 (IL-12-comprised of IL-12p40 and IL-12p35), IL-23 (comprised of the IL-12p40 and IL-23p19 subunits) and homodimeric IL-12p40 (IL-12(p40)(2)). Following activation, immature dendritic cells (DCs) upregulate the chemokine receptor Chemokine-C-Receptor 7 (CCR7), and migrate in response to homeostatic chemokines such as chemokine (C-C motif) ligand 19 (CCL19). Induction of the cytokine IL-12p40 in response to pathogen-exposure, likely in its homodimeric form, is one of the primary events that mediates migration of DCs in response to CCL19. Here we show that following exposure to Francisella tularensis Live Vaccine Strain (LVS), DCs produce IL-12p40 and promote the migration of DCs to the chemokine CCL19 in an IL-12Rβ1- and IL-12p(40)(2)-dependent manner. Induction of IL-12p40 and resulting chemokine responsiveness in DCs is TLR2-dependent and coincides with the uptake of F. tularensis LVS and activation of DCs. Importantly, we show that IL-12Rβ1 signaling is required for DC migration from the lung to the draining lymph node following F. tularensis LVS exposure and coincides with accumulation of IL-12p40 expressing DCs in the draining lymph nodes. Together, these findings illustrate that IL-12p40 is induced rapidly in response to F. tularensis LVS and is required for DC migration through an IL-12Rβ1-IL-12(p40)(2) dependent mechanism.
Collapse
Affiliation(s)
- Samantha R Slight
- Division of Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | | | | | | |
Collapse
|
16
|
Valentino MD, Maben ZJ, Hensley LL, Woolard MD, Kawula TH, Frelinger JA, Frelinger JG. Identification of T-cell epitopes in Francisella tularensis using an ordered protein array of serological targets. Immunology 2011; 132:348-60. [PMID: 21214540 DOI: 10.1111/j.1365-2567.2010.03387.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Francisella tularensis is a Gram-negative intracellular bacterium that is the causative agent of tularaemia. Concerns regarding its use as a bioterrorism agent have led to a renewed interest in the biology of infection, host response and pathogenesis. A robust T-cell response is critical to confer protection against F. tularensis. However, characterization of the cellular immune response has been hindered by the paucity of tools to examine the anti-Francisella immune response at the molecular level. We set out to combine recent advances of genomics with solid-phase antigen delivery coupled with a T-cell functional assay to identify T-cell epitopes. A subset of clones, encoding serological targets, was selected from an F. tularensis SchuS4 ordered genomic library and subcloned into a bacterial expression vector to test the feasibility of this approach. Proteins were expressed and purified individually employing the BioRobot 3000 in a semi-automated purification method. The purified proteins were coupled to beads, delivered to antigen-presenting cells for processing, and screened with Francisella-specific T-cell hybridomas of unknown specificity. We identified cellular reactivity against the pathogenicity protein IglB, and the chaperone proteins GroEL and DnaK. Further analyses using genetic deletions and synthetic peptides were performed to identify the minimal peptide epitopes. Priming with the peptide epitopes before infection with F. tularensis LVS increased the frequency of antigen-specific CD4 T cells as assessed by intracellular interferon-γ staining. These results illustrate the feasibility of screening an arrayed protein library that should be applicable to a variety of pathogens.
Collapse
Affiliation(s)
- Michael D Valentino
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Liguori MJ, Ditewig AC, Maddox JF, Luyendyk JP, Lehman-McKeeman LD, Nelson DM, Bhaskaran VM, Waring JF, Ganey PE, Roth RA, Blomme EAG. Comparison of TNFα to lipopolysaccharide as an inflammagen to characterize the idiosyncratic hepatotoxicity potential of drugs: Trovafloxacin as an example. Int J Mol Sci 2010; 11:4697-714. [PMID: 21151465 PMCID: PMC3000109 DOI: 10.3390/ijms11114697] [Citation(s) in RCA: 18] [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: 09/12/2010] [Revised: 10/22/2010] [Accepted: 11/15/2010] [Indexed: 11/20/2022] Open
Abstract
Idiosyncratic drug reactions (IDRs) are poorly understood, unpredictable, and not detected in preclinical studies. Although the cause of these reactions is likely multi-factorial, one hypothesis is that an underlying inflammatory state lowers the tolerance to a xenobiotic. Previously used in an inflammation IDR model, bacterial lipopolysaccharide (LPS) is heterogeneous in nature, making development of standardized testing protocols difficult. Here, the use of rat tumor necrosis factor-α (TNFα) to replace LPS as an inflammatory stimulus was investigated. Sprague-Dawley rats were treated with separate preparations of LPS or TNFα, and hepatic transcriptomic effects were compared. TNFα showed enhanced consistency at the transcriptomic level compared to LPS. TNFα and LPS regulated similar biochemical pathways, although LPS was associated with more robust inflammatory signaling than TNFα. Rats were then codosed with TNFα and trovafloxacin (TVX), an IDR-associated drug, and evaluated by liver histopathology, clinical chemistry, and gene expression analysis. TNFα/TVX induced unique gene expression changes that clustered separately from TNFα/levofloxacin, a drug not associated with IDRs. TNFα/TVX cotreatment led to autoinduction of TNFα resulting in potentiation of underlying gene expression stress signals. Comparison of TNFα/TVX and LPS/TVX gene expression profiles revealed similarities in the regulation of biochemical pathways. In conclusion, TNFα could be used in lieu of LPS as an inflammatory stimulus in this model of IDRs.
Collapse
Affiliation(s)
- Michael J. Liguori
- Department of Cellular, Molecular, & Exploratory Toxicology, Abbott Laboratories; Abbott Park, IL 60064, USA; E-Mails: (A.C.D.); (J.F.W.); (E.A.G.B.)
| | - Amy C. Ditewig
- Department of Cellular, Molecular, & Exploratory Toxicology, Abbott Laboratories; Abbott Park, IL 60064, USA; E-Mails: (A.C.D.); (J.F.W.); (E.A.G.B.)
| | - Jane F. Maddox
- Department of Pharmacology & Toxicology, Center for Integrative Toxicology, Michigan State University; East Lansing, MI 48824, USA; E-Mails: (J.F.M.); (J.P.L.); (P.E.G.); (R.A.R.)
| | - James P. Luyendyk
- Department of Pharmacology & Toxicology, Center for Integrative Toxicology, Michigan State University; East Lansing, MI 48824, USA; E-Mails: (J.F.M.); (J.P.L.); (P.E.G.); (R.A.R.)
| | - Lois D. Lehman-McKeeman
- Discovery Toxicology, Bristol-Myers Squibb; Princeton, NJ 08540, USA; E-Mail: (L.D.L.-M.); (V.M.B.)
| | - David M. Nelson
- The Beatson Institute for Cancer Research, Glasgow, UK; E-Mail:
| | - Vasanthi M. Bhaskaran
- Discovery Toxicology, Bristol-Myers Squibb; Princeton, NJ 08540, USA; E-Mail: (L.D.L.-M.); (V.M.B.)
| | - Jeffrey F. Waring
- Department of Cellular, Molecular, & Exploratory Toxicology, Abbott Laboratories; Abbott Park, IL 60064, USA; E-Mails: (A.C.D.); (J.F.W.); (E.A.G.B.)
| | - Patricia E. Ganey
- Department of Pharmacology & Toxicology, Center for Integrative Toxicology, Michigan State University; East Lansing, MI 48824, USA; E-Mails: (J.F.M.); (J.P.L.); (P.E.G.); (R.A.R.)
| | - Robert A. Roth
- Department of Pharmacology & Toxicology, Center for Integrative Toxicology, Michigan State University; East Lansing, MI 48824, USA; E-Mails: (J.F.M.); (J.P.L.); (P.E.G.); (R.A.R.)
| | - Eric A. G. Blomme
- Department of Cellular, Molecular, & Exploratory Toxicology, Abbott Laboratories; Abbott Park, IL 60064, USA; E-Mails: (A.C.D.); (J.F.W.); (E.A.G.B.)
| |
Collapse
|
18
|
J774 macrophage-like cell line cytokine and chemokine patterns are modulated by Francisella tularensis LVS strain infection. Folia Microbiol (Praha) 2010; 55:191-200. [PMID: 20490763 DOI: 10.1007/s12223-010-0028-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 11/26/2009] [Indexed: 10/19/2022]
Abstract
Mutual interactions were investigated between intracellular parasitic bacterium Francisella tularensis (F.t.; highly virulent bacterium responsible for tularemia, replicating within the host macrophages) and murine macrophage-like cell line J774. Recombinant murine lymphokine INF-gamma and/or LPS derived from E. coli were determined to stimulate in vitro antimicrobial activity of macrophage-like J774 cell line against the live vaccine strain (LVS) of F.t. through their ability to produce proinflammatory cytokines and chemokines. F.t. infection up-regulated IL-12 p40 production and down-regulated TNF-alpha production by stimulated macrophages; on the other hand, F.t. infection did not affect the production of IL-8, IL-6, MCP-5, and RANTES by stimulated macrophages. This showed that F.t. infection modulates the cytokine synthesis by J774 macrophage cell line.
Collapse
|
19
|
Contribution of citrulline ureidase to Francisella tularensis strain Schu S4 pathogenesis. J Bacteriol 2009; 191:4798-806. [PMID: 19502406 DOI: 10.1128/jb.00212-09] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The citrulline ureidase (CTU) activity has been shown to be associated with highly virulent Francisella tularensis strains, including Schu S4, while it is absent in avirulent or less virulent strains. A definitive role of the ctu gene in virulence and pathogenesis of F. tularensis Schu S4 has not been assessed; thus, an understanding of the significance of this phenotype is long overdue. CTU is a carbon-nitrogen hydrolase encoded by the citrulline ureidase (ctu) gene (FTT0435) on the F. tularensis Schu S4 genome. In the present study, we evaluated the contribution of the ctu gene in the virulence of category A agent F. tularensis Schu S4 by generating a nonpolar deletion mutant, the Deltactu mutant. The deletion of the ctu gene resulted in loss of CTU activity, which was restored by transcomplementing the ctu gene. The Deltactu mutant did not exhibit any growth defect under acellular growth conditions; however, it was impaired for intramacrophage growth in resting as well as gamma interferon-stimulated macrophages. The Deltactu mutant was further tested for its virulence attributes in a mouse model of respiratory tularemia. Mice infected intranasally with the Deltactu mutant showed significantly reduced bacterial burden in the lungs, liver, and spleen compared to wild-type (WT) Schu S4-infected mice. The reduced bacterial burden in mice infected with the Deltactu mutant was also associated with significantly lower histopathological scores in the lungs. Mice infected with the Deltactu mutant succumbed to infection, but they survived longer and showed significantly extended median time to death compared to that shown by WT Schu S4-infected mice. To conclude, this study demonstrates that ctu contributes to intracellular survival, in vivo growth, and pathogenesis. However, ctu is not an absolute requirement for the virulence of F. tularensis Schu S4 in mice.
Collapse
|
20
|
Lutter D, Langmann T, Ugocsai P, Moehle C, Seibold E, Splettstoesser WD, Gruber P, Lang EW, Schmitz G. Analyzing time-dependent microarray data using independent component analysis derived expression modes from human macrophages infected with F. tularensis holartica. J Biomed Inform 2009; 42:605-11. [PMID: 19535009 DOI: 10.1016/j.jbi.2009.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 12/22/2008] [Accepted: 01/12/2009] [Indexed: 11/26/2022]
Abstract
The analysis of large-scale gene expression profiles is still a demanding and extensive task. Modern machine learning and data mining techniques developed in linear algebra, like Independent Component Analysis (ICA), become increasingly popular as appropriate tools for analyzing microarray data. We applied ICA to analyze kinetic gene expression profiles of human monocyte derived macrophages (MDM) from three different donors infected with Francisella tularensis holartica and compared them to more classical methods like hierarchical clustering. Results were compared using a pathway analysis tool, based on the Gene Ontology and the MeSH database. We could show that both methods lead to time-dependent gene regulatory patterns which fit well to known TNFalpha induced immune responses. In comparison, the nonexclusive attribute of ICA results in a more detailed view and a higher resolution in time dependent behavior of the immune response genes. Additionally, we identified NFkappaB as one of the main regulatory genes during response to F. tularensis infection.
Collapse
Affiliation(s)
- D Lutter
- Clinical Chemistry, University Clinic, 93053 Regensburg, Germany; CIML Group, Institute of Biophysics, University of Regensburg, 93040 Regensburg, Germany; Institute of Bioinformatics and Systems Biology, CMB, Helmholtz Zentrum Muenchen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Shirey KA, Cole LE, Keegan AD, Vogel SN. Francisella tularensis live vaccine strain induces macrophage alternative activation as a survival mechanism. THE JOURNAL OF IMMUNOLOGY 2008; 181:4159-67. [PMID: 18768873 DOI: 10.4049/jimmunol.181.6.4159] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Francisella tularensis (Ft), the causative agent of tularemia, elicits a potent inflammatory response early in infection, yet persists within host macrophages and can be lethal if left unchecked. We report in this study that Ft live vaccine strain (LVS) infection of murine macrophages induced TLR2-dependent expression of alternative activation markers that followed the appearance of classically activated markers. Intraperitoneal infection with Ft LVS also resulted in induction of alternatively activated macrophages (AA-Mphi). Induction of AA-Mphi by treatment of cells with rIL-4 or by infection with Ft LVS promoted replication of intracellular Ftn, in contrast to classically activated (IFN-gamma plus LPS) macrophages that promoted intracellular killing of Ft LVS. Ft LVS failed to induce alternative activation in IL-4Ralpha(-/-) or STAT6(-/-) macrophages and prolonged the classical inflammatory response in these cells, resulting in intracellular killing of Ft. Treatment of macrophages with anti-IL-4 and anti-IL-13 Ab blunted Ft-induced AA-Mphi differentiation and resulted in increased expression of IL-12 p70 and decreased bacterial replication. In vivo, Ft-infected IL-4Ralpha(-/-) mice exhibited increased survival compared with wild-type mice. Thus, redirection of macrophage differentiation by Ft LVS from a classical to an alternative activation state enables the organism to survive at the expense of the host.
Collapse
Affiliation(s)
- Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
22
|
Adaptation of Francisella tularensis to the mammalian environment is governed by cues which can be mimicked in vitro. Infect Immun 2008; 76:4479-88. [PMID: 18644878 DOI: 10.1128/iai.00610-08] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The intracellular bacterium Francisella tularensis survives in mammals, arthropods, and freshwater amoeba. It was previously established that the conventional media used for in vitro propagation of this microbe do not yield bacteria that mimic those harvested from infected mammals; whether these in vitro-cultivated bacteria resemble arthropod- or amoeba-adapted Francisella is unknown. As a foundation for our goal of identifying F. tularensis outer membrane proteins which are expressed during mammalian infection, we first sought to identify in vitro cultivation conditions that induce the bacterium's infection-derived phenotype. We compared Francisella LVS grown in brain heart infusion broth (BHI; a standard microbiological medium rarely used in Francisella research) to that grown in Mueller-Hinton broth (MHB; the most widely used F. tularensis medium, used here as a negative control) and macrophages (a natural host cell, used here as a positive control). BHI- and macrophage-grown F. tularensis cells showed similar expression of MglA-dependent and MglA-independent proteins; expression of the MglA-dependent proteins was repressed by the supraphysiological levels of free amino acids present in MHB. We observed that during macrophage infection, protein expression by intracellular bacteria differed from that by extracellular bacteria; BHI-grown bacteria mirrored the latter, while MHB-grown bacteria resembled neither. Naïve macrophages responding to BHI- and macrophage-grown bacteria produced markedly lower levels of proinflammatory mediators than those in cells exposed to MHB-grown bacteria. In contrast to MHB-grown bacteria, BHI-grown bacteria showed minimal delay during intracellular replication. Cumulatively, our findings provide compelling evidence that growth in BHI yields bacteria which recapitulate the phenotype of Francisella organisms that have emerged from macrophages.
Collapse
|
23
|
Parsa KVL, Butchar JP, Rajaram MVS, Cremer TJ, Gunn JS, Schlesinger LS, Tridandapani S. Francisella gains a survival advantage within mononuclear phagocytes by suppressing the host IFNgamma response. Mol Immunol 2008; 45:3428-37. [PMID: 18514317 DOI: 10.1016/j.molimm.2008.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 03/31/2008] [Accepted: 04/02/2008] [Indexed: 10/22/2022]
Abstract
Tularemia is a zoonotic disease caused by the Gram-negative intracellular pathogen Francisella tularensis. These bacteria evade phagolysosomal fusion, escape from the phagosome and replicate in the host cell cytoplasm. IFNgamma has been shown to suppress the intra-macrophage growth of Francisella through both nitric oxide-dependent and -independent pathways. Since Francisella is known to subvert host immune responses, we hypothesized that this pathogen could interfere with IFNgamma signaling. Here, we report that infection with Francisella suppresses IFNgamma-induced STAT1 expression and phosphorylation in both human and murine mononuclear phagocytes. This suppressive effect of Francisella is independent of phagosomal escape or replication and is mediated by a heat-stable and constitutively expressed bacterial factor. An analysis of the molecular mechanism of STAT1 inhibition indicated that expression of SOCS3, an established negative regulator of IFNgamma signaling, is highly up-regulated during infection and suppresses STAT1 phosphorylation. Functional analyses revealed that this interference with IFNgamma signaling is accompanied by the suppression of IP-10 production and iNOS induction resulting in increased intracellular bacterial survival. Importantly, the suppressive effect on IFNgamma-mediated host cell protection is most effective when IFNgamma is added post infection, suggesting that the bacteria establish a permissive environment within the host cell.
Collapse
Affiliation(s)
- Kishore V L Parsa
- The Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Activities of daptomycin and comparative antimicrobials, singly and in combination, against extracellular and intracellular Staphylococcus aureus and its stable small-colony variant in human monocyte-derived macrophages and in broth. Antimicrob Agents Chemother 2008; 52:1829-33. [PMID: 18332172 DOI: 10.1128/aac.01480-07] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated the antistaphylococcal activities of daptomycin, gentamicin, and rifampin against two Staphylococcus aureus strains and their stable small-colony variants, singly and in combination, in human monocyte-derived macrophages and in broth. Intracellularly, the three-drug combination and two-drug combinations with rifampin were most effective. Extracellularly, daptomycin, daptomycin plus gentamicin, gentamicin plus rifampin, and the three-drug combination had similar activities.
Collapse
|
25
|
Carlson PE, Carroll JA, O’Dee DM, Nau GJ. Modulation of virulence factors in Francisella tularensis determines human macrophage responses. Microb Pathog 2007; 42:204-14. [PMID: 17369012 PMCID: PMC2699611 DOI: 10.1016/j.micpath.2007.02.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2006] [Revised: 01/24/2007] [Accepted: 02/02/2007] [Indexed: 02/07/2023]
Abstract
Francisella tularensis, the causative agent of tularemia and Category A biodefense agent, is known to replicate within host macrophages, though the pathogenesis of this organism is incompletely understood. We have isolated a variant of F. tularensis live vaccine strain (LVS) based on colony morphology and its effect on macrophages. Human monocyte-derived macrophages produced more tumor necrosis factor alpha (TNFalpha), interleukin (IL)-1beta, IL-6, and IL-12 p40 following exposure to the variant, designated the activating variant (ACV). The immunoreactivity of the lipopolysaccharide (LPS) from both LVS and ACV was comparable to the previously described blue variant and was distinct from the gray variant of LVS. We found, however, the soluble protein fractions of LVS and ACV differed. Further investigation using two-dimensional gel electrophoresis demonstrated higher levels of several proteins in the parental LVS isolate. The differentially expressed proteins featured several associated with virulence in F. tularensis and other pathogens, including intracellular growth locus C (IglC), a sigma(54)-modulation protein family member (YhbH), and aconitase. ACV reverted to the LVS phenotype, indicated by low cytokine induction and high IglC expression, after growth in a chemically defined medium. These data provide evidence that the levels of virulence factors in F. tularensis are modulated based on culture conditions and that this modulation impacts host responses. This work provides a basis for investigation of Francisella virulence factor regulation and the identification of additional factors, co-regulated with IglC, that affect macrophage responses.
Collapse
Affiliation(s)
- Paul E. Carlson
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261, USA
| | - James A. Carroll
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261, USA
| | - Dawn M. O’Dee
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261, USA
| | - Gerard J. Nau
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261, USA
- Department of Medicine - Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261, USA
- Corresponding Author: Gerard J. Nau, Department of Molecular Genetics and Biochemistry, E1256 BSTWR, 200 Lothrop St., Pittsburgh, PA 15261, Tel: 412-383-9986, Fax: 412-624-1401,
| |
Collapse
|