1
|
Chugh S, Létisse F, Neyrolles O. The exometabolome as a hidden driver of bacterial virulence and pathogenesis. Trends Microbiol 2025; 33:546-557. [PMID: 39701858 DOI: 10.1016/j.tim.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024]
Abstract
The traditional view of metabolism as merely supplying energy and biosynthetic precursors is undergoing a paradigm shift. Metabolic dynamics not only regulates gene expression but also orchestrates cellular processes with remarkable precision. Most bacterial pathogens exhibit exceptional metabolic plasticity, enabling them to adapt to diverse environments, including hostile conditions within a host. While the role of intracellular bacterial metabolism in pathogen-host interactions has been extensively studied, the contributions of the extracellularly released or secreted bacterial metabolites (referred to here as the bacterial 'exometabolome') to metabolic adaptations and disease pathogenesis remain largely unexplored. In this review, we highlight the significant and intriguing roles of bacterial exometabolomes in drug tolerance, immune suppression, and disease pathogenesis, opening a new frontier in our understanding of bacterial-host interactions.
Collapse
Affiliation(s)
- Saurabh Chugh
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Fabien Létisse
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
2
|
Kopping EJ, Benziger PT, Thanassi DG. TolC and EmrA1 contribute to Francisella novicida multidrug resistance and modulation of host cell death. J Bacteriol 2024; 206:e0024624. [PMID: 39194223 PMCID: PMC11411944 DOI: 10.1128/jb.00246-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Francisella spp. are Gram-negative, facultative intracellular pathogens. Francisella tularensis causes the human disease tularemia and is considered a biological threat agent due to its high infectivity and virulence. A central aspect of Francisella virulence is its ability to dampen host immune responses. We previously identified the outer membrane channel (OMC) protein TolC as a critical F. tularensis virulence factor required for suppression of apoptotic and proinflammatory responses during macrophage infection. TolC functions as part of multidrug efflux systems and the type I secretion pathway that exports bacterial effector proteins. In these systems, TolC forms tripartite complexes together with an inner membrane transporter and periplasmic membrane fusion protein (MFP). To advance understanding of TolC function in Francisella, we analyzed OMC and MFP homologs in Francisella novicida, a widely used model species that causes a tularemia-like disease in mice. In agreement with the previous F. tularensis studies, all three OMCs present in F. novicida contributed to multidrug resistance, but only TolC was important for suppressing macrophage cell death. In addition, we identified the EmrA1 MFP as important for resisting antimicrobial compounds and dampening host cell death. In contrast to results obtained with F. tularensis, the cell death triggered during infection with the F. novicida tolC and emrA1 mutants was dominated by pyroptosis rather than apoptosis. These data expand our understanding of TolC function in Francisella and underscore both conserved and differential aspects of F. novicida and F. tularensis. IMPORTANCE Francisella tularensis is a Gram-negative intracellular bacterial pathogen and causative agent of tularemia. We previously identified the outer membrane channel protein TolC as contributing to antimicrobial resistance and subversion of host responses by F. tularensis. To advance understanding of TolC function in Francisella and to identify components that might work together with TolC, we took advantage of a transposon mutant library in F. novicida, a model species that causes a tularemia-like disease in mice. Our findings identify TolC and the membrane fusion protein EmrA1 as important for both antimicrobial resistance and suppression of macrophage cell death. This study also revealed differences in cell death pathways triggered by F. novicida versus F. tularensis infection that may relate to differences in virulence.
Collapse
Affiliation(s)
- Erik J Kopping
- Department of Microbiology and Immunology, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - P Todd Benziger
- Department of Microbiology and Immunology, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - David G Thanassi
- Department of Microbiology and Immunology, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
3
|
Benziger PT, Kopping EJ, McLaughlin PA, Thanassi DG. Francisella tularensis disrupts TLR2-MYD88-p38 signaling early during infection to delay apoptosis of macrophages and promote virulence in the host. mBio 2023; 14:e0113623. [PMID: 37404047 PMCID: PMC10470500 DOI: 10.1128/mbio.01136-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/24/2023] [Indexed: 07/06/2023] Open
Abstract
Francisella tularensis is a zoonotic pathogen and the causative agent of tularemia. F. tularensis replicates to high levels within the cytosol of macrophages and other host cells while subverting the host response to infection. Critical to the success of F. tularensis is its ability to delay macrophage apoptosis to maintain its intracellular replicative niche. However, the host-signaling pathway(s) modulated by F. tularensis to delay apoptosis are poorly characterized. The outer membrane channel protein TolC is required for F. tularensis virulence and its ability to suppress apoptosis and cytokine expression during infection of macrophages. We took advantage of the F. tularensis ∆tolC mutant phenotype to identify host pathways that are important for activating macrophage apoptosis and that are disrupted by the bacteria. Comparison of macrophages infected with wild-type or ∆tolC F. tularensis revealed that the bacteria interfere with TLR2-MYD88-p38 signaling at early times post infection to delay apoptosis, dampen innate host responses, and preserve the intracellular replicative niche. Experiments using the mouse pneumonic tularemia model confirmed the in vivo relevance of these findings, revealing contributions of TLR2 and MYD88 signaling to the protective host response to F. tularensis, which is modulated by the bacteria to promote virulence. IMPORTANCE Francisella tularensis is a Gram-negative intracellular bacterial pathogen and the causative agent of the zoonotic disease tularemia. F. tularensis, like other intracellular pathogens, modulates host-programmed cell death pathways to ensure its replication and survival. We previously identified the outer membrane channel protein TolC as required for the ability of F. tularensis to delay host cell death. However, the mechanism by which F. tularensis delays cell death pathways during intracellular replication is unclear despite being critical to pathogenesis. In the present study, we address this gap in knowledge by taking advantage of ∆tolC mutants of F. tularensis to uncover signaling pathways governing host apoptotic responses to F. tularensis and which are modulated by the bacteria during infection to promote virulence. These findings reveal mechanisms by which intracellular pathogens subvert host responses and enhance our understanding of the pathogenesis of tularemia.
Collapse
Affiliation(s)
- P. Todd Benziger
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Erik J. Kopping
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Patrick A. McLaughlin
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - David G. Thanassi
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
4
|
Alqahtani M, Ma Z, Miller J, Yu J, Malik M, Bakshi CS. Comparative analysis of absent in melanoma 2-inflammasome activation in Francisella tularensis and Francisella novicida. Front Microbiol 2023; 14:1188112. [PMID: 37266012 PMCID: PMC10230036 DOI: 10.3389/fmicb.2023.1188112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/02/2023] [Indexed: 06/03/2023] Open
Abstract
Francisella tularensis is a highly virulent Gram-negative bacterium that causes the fatal zoonotic disease tularemia. The mechanisms and signaling pathways leading to the absent in melanoma 2 (Aim2) inflammasome activation have been elegantly elucidated using Francisella novicida as a model. Although not pathogenic for humans, F. novicida can cause tularemia in mice, and the inflammatory response it triggers is the polar opposite to that observed in mice infected with F. tularensis strains. This study aimed to understand the mechanisms of Aim2 inflammasome activation in F. tularensis-infected macrophages. The results reveal that macrophages infected with the F. tularensis live vaccine strain (LVS) induce lower levels of Aim2-dependent IL-1β than those infected with F. novicida. The suppression/weak activation of Aim2 in F. tularensis LVS-infected macrophages is due to the suppression of the cGAS-STING DNA-sensing pathway. Furthermore, the introduction of exogenous F. tularensis LVS DNA into the cytosol of the F. tularensis LVS-infected macrophages, alone or in conjunction with a priming signal, failed to restore IL-1β levels similar to those observed for F. novicida-infected macrophages. These results indicated that, in addition to the bacterial DNA, DNA from some other sources, specifically from the damaged mitochondria, might contribute to the robust Aim2-dependent IL-1β levels observed in F. novicida-infected macrophages. The results indicate that F. tularensis LVS induces mitophagy that may potentially prevent the leakage of mitochondrial DNA and the subsequent activation of the Aim2 inflammasome. Collectively, this study demonstrates that the mechanisms of Aim2 inflammasome activation established for F. novicida are not operative in F. tularensis.
Collapse
Affiliation(s)
- Maha Alqahtani
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Zhuo Ma
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, United States
| | - Jacob Miller
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, United States
| | - Jen Yu
- Electron Microscopy Laboratory, Department of Pathology, Westchester Medical Center, Valhalla, NY, United States
| | - Meenakshi Malik
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, United States
| | - Chandra Shekhar Bakshi
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
5
|
Kinkead LC, Krysa SJ, Allen LAH. Neutrophil Survival Signaling During Francisella tularensis Infection. Front Cell Infect Microbiol 2022; 12:889290. [PMID: 35873156 PMCID: PMC9299441 DOI: 10.3389/fcimb.2022.889290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/07/2022] [Indexed: 12/18/2022] Open
Abstract
Neutrophils are the most abundant and shortest-lived leukocytes in humans and tight regulation of neutrophil turnover via constitutive apoptosis is essential for control of infection and resolution of inflammation. Accordingly, aberrant neutrophil turnover is hallmark of many disease states. We have shown in previous work that the intracellular bacterial pathogen Francisella tularensis markedly prolongs human neutrophil lifespan. This is achieved, in part, by changes in neutrophil gene expression. Still unknown is the contribution of major neutrophil pro-survival signaling cascades to this process. The objective of this study was to interrogate the contributions of ERK and p38 MAP kinase, Class I phosphoinositide 3-kinases (PI3K), AKT, and NF-κB to neutrophil survival in our system. We demonstrate that both ERK2 and p38α were activated in F. tularensis-infected neutrophils, but only p38α MAPK was required for delayed apoptosis and the rate of cell death in the absence of infection was unchanged. Apoptosis of both infected and uninfected neutrophils was markedly accelerated by the pan-PI3K inhibitor LY2094002, but AKT phosphorylation was not induced, and neutrophil death was not enhanced by AKT inhibitors. In addition, isoform specific and selective inhibitors revealed a unique role for PI3Kα in neutrophil survival after infection, whereas only simultaneous inhibition of PI3Kα and PI3kδ accelerated death of the uninfected controls. Finally, we show that inhibition of NF-κB triggered rapid death of neutrophil after infection. Thus, we defined roles for p38α, PI3Kα and NF-κB delayed apoptosis of F. tularensis-infected cells and advanced understanding of Class IA PI3K isoform activity in human neutrophil survival.
Collapse
Affiliation(s)
- Lauren C. Kinkead
- Inflammation Program, University of Iowa, Iowa City, IA, United States
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
- Iowa City VA Health Care System, Iowa City, IA, United States
| | - Samantha J. Krysa
- Inflammation Program, University of Iowa, Iowa City, IA, United States
- Iowa City VA Health Care System, Iowa City, IA, United States
- Molecular Medicine Training Program, University of Iowa, Iowa City, IA, United States
| | - Lee-Ann H. Allen
- Inflammation Program, University of Iowa, Iowa City, IA, United States
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
- Iowa City VA Health Care System, Iowa City, IA, United States
- Molecular Medicine Training Program, University of Iowa, Iowa City, IA, United States
- Department of Medicine, Division of Infectious Diseases, University of Iowa, Iowa City, IA, United States
- Harry S. Truman Memorial VA Hospital, Columbia, MO, United States
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
6
|
Bradford MK, Elkins KL. Immune lymphocytes halt replication of Francisella tularensis LVS within the cytoplasm of infected macrophages. Sci Rep 2020; 10:12023. [PMID: 32694562 PMCID: PMC7374111 DOI: 10.1038/s41598-020-68798-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 06/26/2020] [Indexed: 12/18/2022] Open
Abstract
Francisella tularensis is a highly infectious intracellular bacterium that causes tularemia by invading and replicating in mammalian myeloid cells. Francisella primarily invades host macrophages, where it escapes phagosomes within a few hours and replicates in the cytoplasm. Less is known about how Francisella traffics within macrophages or exits into the extracellular environment for further infection. Immune T lymphocytes control the replication of Francisella within macrophages in vitro by a variety of mechanisms, but nothing is known about intracellular bacterial trafficking in the face of such immune pressure. Here we used a murine model of infection with a Francisella attenuated live vaccine strain (LVS), which is under study as a human vaccine, to evaluate the hypothesis that immune T cells control intramacrophage bacterial growth by re-directing bacteria into toxic intracellular compartments of infected macrophages. We visualized the interactions of lymphocytes and LVS-infected macrophages using confocal microscopy and characterized LVS intramacrophage trafficking when co-cultured with immune lymphocytes. We focused on the late stages of infection after bacteria escape from phagosomes, through bacterial replication and the death of macrophages. We found that the majority of LVS remained cytosolic in the absence of immune pressure, eventually resulting in macrophage death. In contrast, co-culture of LVS-infected macrophages with LVS-immune lymphocytes halted LVS replication and inhibited the spread of LVS infection between macrophages, but bacteria did not return to vacuoles such as lysosomes or autophagosomes and macrophages did not die. Therefore, immune lymphocytes directly limit intracellular bacterial replication within the cytoplasm of infected macrophages.
Collapse
Affiliation(s)
- Mary Katherine Bradford
- 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, 20993, USA.,Johns Hopkins University Professional Development and Career Office, 1830 E. Monument, 2-107, Baltimore, MD, 21287, USA
| | - 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, 20993, USA.
| |
Collapse
|
7
|
Contributions of TolC Orthologs to Francisella tularensis Schu S4 Multidrug Resistance, Modulation of Host Cell Responses, and Virulence. Infect Immun 2019; 87:IAI.00823-18. [PMID: 30670554 DOI: 10.1128/iai.00823-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023] Open
Abstract
Francisella tularensis is a Gram-negative, facultative intracellular pathogen and the causative agent of tularemia. Previous studies with the attenuated live vaccine strain (LVS) identified a role for the outer membrane protein TolC in modulation of host cell responses during infection and virulence in the mouse model of tularemia. TolC is an integral part of efflux pumps that export small molecules and type I secretion systems that export a range of bacterial virulence factors. In this study, we analyzed TolC and its two orthologs, FtlC and SilC, present in the fully virulent F. tularensis Schu S4 strain for their contributions to multidrug efflux, suppression of innate immune responses, and virulence. We found that each TolC ortholog participated in multidrug efflux, with overlapping substrate specificities for TolC and FtlC and a distinct substrate profile for SilC. In contrast to their shared roles in drug efflux, only TolC functioned in the modulation of macrophage apoptotic and proinflammatory responses to Schu S4 infection, consistent with a role in virulence factor delivery to host cells. In agreement with previous results with the LVS, the Schu S4 ΔtolC mutant was highly attenuated for virulence in mice by both the intranasal and intradermal routes of infection. Unexpectedly, FtlC was also critical for Schu S4 virulence, but only by the intradermal route. Our data demonstrate a conserved and critical role for TolC in modulation of host immune responses and Francisella virulence and also highlight strain- and route-dependent differences in the pathogenesis of tularemia.
Collapse
|
8
|
Carruthers J, López-García M, Gillard JJ, Laws TR, Lythe G, Molina-París C. A Novel Stochastic Multi-Scale Model of Francisella tularensis Infection to Predict Risk of Infection in a Laboratory. Front Microbiol 2018; 9:1165. [PMID: 30034369 PMCID: PMC6043654 DOI: 10.3389/fmicb.2018.01165] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/14/2018] [Indexed: 12/30/2022] Open
Abstract
We present a multi-scale model of the within-phagocyte, within-host and population-level infection dynamics of Francisella tularensis, which extends the mechanistic one proposed by Wood et al. (2014). Our multi-scale model incorporates key aspects of the interaction between host phagocytes and extracellular bacteria, accounts for inter-phagocyte variability in the number of bacteria released upon phagocyte rupture, and allows one to compute the probability of response, and mean time until response, of an infected individual as a function of the initial infection dose. A Bayesian approach is applied to parameterize both the within-phagocyte and within-host models using infection data. Finally, we show how dose response probabilities at the individual level can be used to estimate the airborne propagation of Francisella tularensis in indoor settings (such as a microbiology laboratory) at the population level, by means of a deterministic zonal ventilation model.
Collapse
Affiliation(s)
- Jonathan Carruthers
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds, United Kingdom
| | - Martín López-García
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds, United Kingdom
| | - Joseph J. Gillard
- Defence Science and Technology Laboratory, Salisbury, United Kingdom
| | - Thomas R. Laws
- Defence Science and Technology Laboratory, Salisbury, United Kingdom
| | - Grant Lythe
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds, United Kingdom
| | - Carmen Molina-París
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
9
|
Abstract
The coevolution of intracellular bacteria with their eukaryotic hosts has presented these pathogens with numerous challenges for their evolutionary progress and survival. Chief among these is the ability to exit from host cells, an event that is fundamentally linked to pathogen dissemination and transmission. Recent years have witnessed a major expansion of research in this area, and this chapter summarizes our current understanding of the spectrum of exit strategies that are exploited by intracellular pathogens. Clear themes regarding the mechanisms of microbial exit have emerged and are most easily conceptualized as (i) lysis of the host cell, (ii) nonlytic exit of free bacteria, and (iii) release of microorganisms into membrane-encased compartments. The adaptation of particular exit strategies is closely linked with additional themes in microbial pathogenesis, including host cell death, manipulation of host signaling pathways, and coincident activation of proinflammatory responses. This chapter will explore the molecular determinants used by intracellular pathogens to promote host cell escape and the infectious advantages each exit pathway may confer, and it will provide an evolutionary framework for the adaptation of these mechanisms.
Collapse
|
10
|
Vaccinia Virus Encodes a Novel Inhibitor of Apoptosis That Associates with the Apoptosome. J Virol 2017; 91:JVI.01385-17. [PMID: 28904196 DOI: 10.1128/jvi.01385-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/08/2017] [Indexed: 12/29/2022] Open
Abstract
Apoptosis is an important antiviral host defense mechanism. Here we report the identification of a novel apoptosis inhibitor encoded by the vaccinia virus (VACV) M1L gene. M1L is absent in the attenuated modified vaccinia virus Ankara (MVA) strain of VACV, a strain that stimulates apoptosis in several types of immune cells. M1 expression increased the viability of MVA-infected THP-1 and Jurkat cells and reduced several biochemical hallmarks of apoptosis, such as PARP-1 and procaspase-3 cleavage. Furthermore, ectopic M1L expression decreased staurosporine-induced (intrinsic) apoptosis in HeLa cells. We then identified the molecular basis for M1 inhibitory function. M1 allowed mitochondrial depolarization but blocked procaspase-9 processing, suggesting that M1 targeted the apoptosome. In support of this model, we found that M1 promoted survival in Saccharomyces cerevisiae overexpressing human Apaf-1 and procaspase-9, critical components of the apoptosome, or overexpressing only conformationally active caspase-9. In mammalian cells, M1 coimmunoprecipitated with Apaf-1-procaspase-9 complexes. The current model is that M1 associates with and allows the formation of the apoptosome but prevents apoptotic functions of the apoptosome. The M1 protein features 14 predicted ankyrin (ANK) repeat domains, and M1 is the first ANK-containing protein reported to use this inhibitory strategy. Since ANK-containing proteins are encoded by many large DNA viruses and found in all domains of life, studies of M1 may lead to a better understanding of the roles of ANK proteins in virus-host interactions.IMPORTANCE Apoptosis selectively eliminates dangerous cells such as virus-infected cells. Poxviruses express apoptosis antagonists to neutralize this antiviral host defense. The vaccinia virus (VACV) M1 ankyrin (ANK) protein, a protein with no previously ascribed function, inhibits apoptosis. M1 interacts with the apoptosome and prevents procaspase-9 processing as well as downstream procaspase-3 cleavage in several cell types and under multiple conditions. M1 is the first poxviral protein reported to associate with and prevent the function of the apoptosome, giving a more detailed picture of the threats VACV encounters during infection. Dysregulation of apoptosis is associated with several human diseases. One potential treatment of apoptosis-related diseases is through the use of designed ANK repeat proteins (DARPins), similar to M1, as caspase inhibitors. Thus, the study of the novel antiapoptosis effects of M1 via apoptosome association will be helpful for understanding how to control apoptosis using either natural or synthetic molecules.
Collapse
|
11
|
Fabrik I, Link M, Putzova D, Plzakova L, Lubovska Z, Philimonenko V, Pavkova I, Rehulka P, Krocova Z, Hozak P, Santic M, Stulik J. The Early Dendritic Cell Signaling Induced by Virulent Francisella tularensis Strain Occurs in Phases and Involves the Activation of Extracellular Signal-Regulated Kinases (ERKs) and p38 In the Later Stage. Mol Cell Proteomics 2017; 17:81-94. [PMID: 29046388 DOI: 10.1074/mcp.ra117.000160] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/22/2017] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) infected by Francisella tularensis are poorly activated and do not undergo classical maturation process. Although reasons of such unresponsiveness are not fully understood, their impact on the priming of immunity is well appreciated. Previous attempts to explain the behavior of Francisella-infected DCs were hypothesis-driven and focused on events at later stages of infection. Here, we took an alternative unbiased approach by applying methods of global phosphoproteomics to analyze the dynamics of cell signaling in primary DCs during the first hour of infection by Francisella tularensis Presented results show that the early response of DCs to Francisella occurs in phases and that ERK and p38 signaling modules induced at the later stage are differentially regulated by virulent and attenuated ΔdsbA strain. These findings imply that the temporal orchestration of host proinflammatory pathways represents the integral part of Francisella life-cycle inside hijacked DCs.
Collapse
Affiliation(s)
- Ivo Fabrik
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Marek Link
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Daniela Putzova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Lenka Plzakova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Zuzana Lubovska
- §Institute of Molecular Genetics ASCR v.v.i., Microscopy Centre, Electron Microscopy Core Facility, 142 20 Prague 4, Czech Republic
| | - Vlada Philimonenko
- §Institute of Molecular Genetics ASCR v.v.i., Microscopy Centre, Electron Microscopy Core Facility, 142 20 Prague 4, Czech Republic.,¶Institute of Molecular Genetics ASCR v.v.i., Department of Biology of the Cell Nucleus, 142 20 Prague 4, Czech Republic
| | - Ivona Pavkova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Pavel Rehulka
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Zuzana Krocova
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic
| | - Pavel Hozak
- §Institute of Molecular Genetics ASCR v.v.i., Microscopy Centre, Electron Microscopy Core Facility, 142 20 Prague 4, Czech Republic.,¶Institute of Molecular Genetics ASCR v.v.i., Department of Biology of the Cell Nucleus, 142 20 Prague 4, Czech Republic
| | - Marina Santic
- ‖Department of Microbiology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Jiri Stulik
- From the ‡Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic;
| |
Collapse
|
12
|
Complement C3 as a Prompt for Human Macrophage Death during Infection with Francisella tularensis Strain SCHU S4. Infect Immun 2017; 85:IAI.00424-17. [PMID: 28739830 DOI: 10.1128/iai.00424-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 07/18/2017] [Indexed: 02/06/2023] Open
Abstract
Tularemia is caused by the Gram-negative bacterial pathogen Francisella tularensis Infection of macrophages and their subsequent death are believed to play important roles in the progression of disease. Because complement is a particularly effective opsonin for Francisella, we asked whether complement-dependent uptake of F. tularensis strain SCHU S4 affects the survival of primary human macrophages during infection. Complement component C3 was found to be an essential opsonin in human serum not only for greatly increased uptake of SCHU S4 but also for the induction of macrophage death. Single-cell analysis also revealed that macrophage death did not require a high intracellular bacterial burden. In the presence of C3, macrophage death was observed at 24 h postinfection in a quarter of the macrophages that contained only 1 to 5 bacterial cells. Macrophages infected in the absence of C3 rarely underwent cell death, even when they contained large numbers of bacteria. The need for C3, but not extensive replication of the pathogen, was confirmed by infections with SCHU S4 ΔpurMCD, a mutant capable of phagosome escape but of only limited cytosolic replication. C3-dependent Francisella uptake alone was insufficient to induce macrophage death, as evidenced by the failure of the phagosome escape-deficient mutant SCHU S4 ΔfevR to induce cell death despite opsonization with C3. Together, these findings indicate that recognition of C3-opsonized F. tularensis, but not extensive cytosolic replication, plays an important role in regulating macrophage viability during intracellular infections with type A F. tularensis.
Collapse
|
13
|
Bolz C, Bach NC, Meyer H, Müller G, Dawidowski M, Popowicz G, Sieber SA, Skerra A, Gerhard M. Comparison of enzymatic properties and small molecule inhibition of γ-glutamyltranspeptidases from pathogenic and commensal bacteria. Biol Chem 2017; 398:341-357. [PMID: 27636829 DOI: 10.1515/hsz-2016-0198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/24/2016] [Indexed: 01/06/2023]
Abstract
Helicobacter pylori infects the stomach of 50% of the population worldwide, thus causing chronic gastritis. Although this infection can be cured by antibiotic treatment, therapeutic options are increasingly limited due to the development of resistances. The γ-glutamyl-transpeptidase (gGT) of H. pylori (HpgGT) is a virulence factor important for colonization and contributes to bacterial immune evasion. Therefore, this enzyme is a potential target for developing new anti-infectives. As species specificity of such compounds is required in order to avoid off-target or adverse effects, comparative analysis of the gGTs from different organisms is a prerequisite for drug development. To allow detailed biochemical and enzymatic characterization, recombinant gGTs from five different bacteria as well as Homo sapiens were characterized and compared. Investigation of the enzymatic activity, the binding modes of known inhibitors to the catalytic center, and a high resolution X-ray structure of the HpgGT provided a starting point for the identification of new inhibitory substances targeting HpgGT. Inhibitors with Ki values in the nm to mm range were identified and their binding modes were analyzed by mass spectrometry. The results of this study provide a basis for the development of species-specific lead compounds with anti-infective potential by effectively inhibiting HpgGT.
Collapse
|
14
|
Di Venanzio G, Lazzaro M, Morales ES, Krapf D, García Véscovi E. A pore-forming toxin enables Serratia a nonlytic egress from host cells. Cell Microbiol 2016; 19. [PMID: 27532510 DOI: 10.1111/cmi.12656] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/28/2016] [Accepted: 08/12/2016] [Indexed: 01/01/2023]
Abstract
Several pathogens co-opt host intracellular compartments to survive and replicate, and they thereafter disperse progeny to prosper in a new niche. Little is known about strategies displayed by Serratia marcescens to defeat immune responses and disseminate afterwards. Upon invasion of nonphagocytic cells, Serratia multiplies within autophagosome-like vacuoles. These Serratia-containing vacuoles (SeCV) circumvent progression into acidic/degradative compartments, avoiding elimination. In this work, we show that ShlA pore-forming toxin (PFT) commands Serratia escape from invaded cells. While ShlA-dependent, Ca2+ local increase was shown in SeCVs tight proximity, intracellular Ca2+ sequestration prevented Serratia exit. Accordingly, a Ca2+ surge rescued a ShlA-deficient strain exit capacity, demonstrating that Ca2+ mobilization is essential for egress. As opposed to wild-type-SeCV, the mutant strain-vacuole was wrapped by actin filaments, showing that ShlA expression rearranges host actin. Moreover, alteration of actin polymerization hindered wild-type Serratia escape, while increased intracellular Ca2+ reorganized the mutant strain-SeCV actin distribution, restoring wild-type-SeCV phenotype. Our results demonstrate that, by ShlA expression, Serratia triggers a Ca2+ signal that reshapes cytoskeleton dynamics and ends up pushing the SeCV load out of the cell, in an exocytic-like process. These results disclose that PFTs can be engaged in allowing bacteria to exit without compromising host cell integrity.
Collapse
Affiliation(s)
| | | | - Enrique S Morales
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Darío Krapf
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Eleonora García Véscovi
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
15
|
Dissection of Francisella-Host Cell Interactions in Dictyostelium discoideum. Appl Environ Microbiol 2015; 82:1586-1598. [PMID: 26712555 DOI: 10.1128/aem.02950-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/22/2015] [Indexed: 12/31/2022] Open
Abstract
Francisella bacteria cause severe disease in both vertebrates and invertebrates and include one of the most infectious human pathogens. Mammalian cell lines have mainly been used to study the mechanisms by which Francisella manipulates its host to replicate within a large variety of hosts and cell types, including macrophages. Here, we describe the establishment of a genetically and biochemically tractable infection model: the amoeba Dictyostelium discoideum combined with the fish pathogen Francisella noatunensis subsp. noatunensis. Phagocytosed F. noatunensis subsp. noatunensis interacts with the endosomal pathway and escapes further phagosomal maturation by translocating into the host cell cytosol. F. noatunensis subsp. noatunensis lacking IglC, a known virulence determinant required for Francisella intracellular replication, follows the normal phagosomal maturation and does not grow in Dictyostelium. The attenuation of the F. noatunensis subsp. noatunensis ΔiglC mutant was confirmed in a zebrafish embryo model, where growth of F. noatunensis subsp. noatunensis ΔiglC was restricted. In Dictyostelium, F. noatunensis subsp. noatunensis interacts with the autophagic machinery. The intracellular bacteria colocalize with autophagic markers, and when autophagy is impaired (Dictyostelium Δatg1), F. noatunensis subsp. noatunensis accumulates within Dictyostelium cells. Altogether, the Dictyostelium-F. noatunensis subsp. noatunensis infection model recapitulates the course of infection described in other host systems. The genetic and biochemical tractability of the system allows new approaches to elucidate the dynamic interactions between pathogenic Francisella and its host organism.
Collapse
|
16
|
Ghonime MG, Mitra S, Eldomany RA, Wewers MD, Gavrilin MA. Inflammasome priming is similar for francisella species that differentially induce inflammasome activation. PLoS One 2015; 10:e0127278. [PMID: 25993107 PMCID: PMC4436270 DOI: 10.1371/journal.pone.0127278] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/13/2015] [Indexed: 01/15/2023] Open
Abstract
Inflammasome activation is a two-step process where step one, priming, prepares the inflammasome for its subsequent activation, by step two. Classically step one can be induced by LPS priming followed by step two, high dose ATP. Furthermore, when IL-18 processing is used as the inflammasome readout, priming occurs before new protein synthesis. In this context, how intracellular pathogens such as Francisella activate the inflammasome is incompletely understood, particularly regarding the relative importance of priming versus activation steps. To better understand these events we compared Francisella strains that differ in virulence and ability to induce inflammasome activation for their relative effects on step one vs. step two. When using the rapid priming model, i.e., 30 min priming by live or heat killed Francisella strains (step 1), followed by ATP (step 2), we found no difference in IL-18 release, p20 caspase-1 release and ASC oligomerization between Francisella strains (F. novicida, F. holarctica -LVS and F. tularensis Schu S4). This priming is fast, independent of bacteria viability, internalization and phagosome escape, but requires TLR2-mediated ERK phosphorylation. In contrast to their efficient priming capacity, Francisella strains LVS and Schu S4 were impaired in inflammasome triggering compared to F. novicida. Thus, observed differences in inflammasome activation by F. novicida, LVS and Schu S4 depend not on differences in priming but rather on their propensity to trigger the primed inflammasome.
Collapse
Affiliation(s)
- Mohammed G. Ghonime
- Davis Heart & Lung Research Institute and Pulmonary Allergy Critical Care and Sleep Medicine Division, The Ohio State University, Columbus, OH, 43210, United States of America
- Microbiology and Immunology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Srabani Mitra
- Davis Heart & Lung Research Institute and Pulmonary Allergy Critical Care and Sleep Medicine Division, The Ohio State University, Columbus, OH, 43210, United States of America
| | - Ramadan A. Eldomany
- Microbiology and Immunology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mark D. Wewers
- Davis Heart & Lung Research Institute and Pulmonary Allergy Critical Care and Sleep Medicine Division, The Ohio State University, Columbus, OH, 43210, United States of America
- * E-mail: and
| | - Mikhail A. Gavrilin
- Davis Heart & Lung Research Institute and Pulmonary Allergy Critical Care and Sleep Medicine Division, The Ohio State University, Columbus, OH, 43210, United States of America
- * E-mail: and
| |
Collapse
|
17
|
Lai XH, Xu Y, Chen XM, Ren Y. Macrophage cell death upon intracellular bacterial infection. ACTA ACUST UNITED AC 2015; 2:e779. [PMID: 26690967 DOI: 10.14800/macrophage.779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Macrophage-pathogen interaction is a complex process and the outcome of this tag-of-war for both sides is to live or die. Without attempting to be comprehensive, this review will discuss the complexity and significance of the interaction outcomes between macrophages and some facultative intracellular bacterial pathogens as exemplified by Francisella, Salmonella, Shigella and Yersinia. Upon bacterial infection, macrophages can die by a variety of ways, such as apoptosis, autophagic cell death, necrosis, necroptosis, oncosis, pyronecrosis, pyroptosis etc, which is the focus of this review.
Collapse
Affiliation(s)
- Xin-He Lai
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Institute of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yunsheng Xu
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Institute of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Department of Dermato-venerology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Ming Chen
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Institute of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Department of Pediatric Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Ren
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ; Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA)
| |
Collapse
|
18
|
Zinman GE, Naiman S, O'Dee DM, Kumar N, Nau GJ, Cohen HY, Bar-Joseph Z. ModuleBlast: identifying activated sub-networks within and across species. Nucleic Acids Res 2015; 43:e20. [PMID: 25428368 PMCID: PMC4330341 DOI: 10.1093/nar/gku1224] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 10/11/2014] [Accepted: 11/09/2014] [Indexed: 11/18/2022] Open
Abstract
Identifying conserved and divergent response patterns in gene networks is becoming increasingly important. A common approach is integrating expression information with gene association networks in order to find groups of connected genes that are activated or repressed. In many cases, researchers are also interested in comparisons across species (or conditions). Finding an active sub-network is a hard problem and applying it across species requires further considerations (e.g. orthology information, expression data and networks from different sources). To address these challenges we devised ModuleBlast, which uses both expression and network topology to search for highly relevant sub-networks. We have applied ModuleBlast to expression and interaction data from mouse, macaque and human to study immune response and aging. The immune response analysis identified several relevant modules, consistent with recent findings on apoptosis and NFκB activation following infection. Temporal analysis of these data revealed cascades of modules that are dynamically activated within and across species. We have experimentally validated some of the novel hypotheses resulting from the analysis of the ModuleBlast results leading to new insights into the mechanisms used by a key mammalian aging protein.
Collapse
Affiliation(s)
- Guy E Zinman
- Lane Center for Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Shoshana Naiman
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Dawn M O'Dee
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Nishant Kumar
- Lane Center for Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Gerard J Nau
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Haim Y Cohen
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Ziv Bar-Joseph
- Lane Center for Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
19
|
Wall DM, McCormick BA. Bacterial secreted effectors and caspase-3 interactions. Cell Microbiol 2014; 16:1746-56. [PMID: 25262664 PMCID: PMC4257569 DOI: 10.1111/cmi.12368] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 09/10/2014] [Accepted: 09/15/2014] [Indexed: 12/29/2022]
Abstract
Apoptosis is a critical process that intrinsically links organism survival to its ability to induce controlled death. Thus, functional apoptosis allows organisms to remove perceived threats to their survival by targeting those cells that it determines pose a direct risk. Central to this process are apoptotic caspases, enzymes that form a signalling cascade, converting danger signals via initiator caspases into activation of the executioner caspase, caspase-3. This enzyme begins disassembly of the cell by activating DNA degrading enzymes and degrading the cellular architecture. Interaction of pathogenic bacteria with caspases, and in particular, caspase-3, can therefore impact both host cell and bacterial survival. With roles outside cell death such as cell differentiation, control of signalling pathways and immunomodulation also being described for caspase-3, bacterial interactions with caspase-3 may be of far more significance in infection than previously recognized. In this review, we highlight the ways in which bacterial pathogens have evolved to subvert caspase-3 both through effector proteins that directly interact with the enzyme or by modulating pathways that influence its activation and activity.
Collapse
Affiliation(s)
- Daniel M Wall
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8QQ, UK
| | | |
Collapse
|
20
|
TolC-dependent modulation of host cell death by the Francisella tularensis live vaccine strain. Infect Immun 2014; 82:2068-78. [PMID: 24614652 DOI: 10.1128/iai.00044-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Francisella tularensis is a facultative intracellular, Gram-negative pathogen and the causative agent of tularemia. We previously identified TolC as a virulence factor of the F. tularensis live vaccine strain (LVS) and demonstrated that a ΔtolC mutant exhibits increased cytotoxicity toward host cells and elicits increased proinflammatory responses compared to those of the wild-type (WT) strain. TolC is the outer membrane channel component used by the type I secretion pathway to export toxins and other bacterial virulence factors. Here, we show that the LVS delays activation of the intrinsic apoptotic pathway in a TolC-dependent manner, both during infection of primary macrophages and during organ colonization in mice. The TolC-dependent delay in host cell death is required for F. tularensis to preserve its intracellular replicative niche. We demonstrate that TolC-mediated inhibition of apoptosis is an active process and not due to defects in the structural integrity of the ΔtolC mutant. These findings support a model wherein the immunomodulatory capacity of F. tularensis relies, at least in part, on TolC-secreted effectors. Finally, mice vaccinated with the ΔtolC LVS are protected from lethal challenge and clear challenge doses faster than WT-vaccinated mice, demonstrating that the altered host responses to primary infection with the ΔtolC mutant led to altered adaptive immune responses. Taken together, our data demonstrate that TolC is required for temporal modulation of host cell death during infection by F. tularensis and highlight how shifts in the magnitude and timing of host innate immune responses may lead to dramatic changes in the outcome of infection.
Collapse
|
21
|
Abstract
A wide spectrum of pathogenic bacteria and protozoa has adapted to an intracellular life-style, which presents several advantages, including accessibility to host cell metabolites and protection from the host immune system. Intracellular pathogens have developed strategies to enter and exit their host cells while optimizing survival and replication, progression through the life cycle, and transmission. Over the last decades, research has focused primarily on entry, while the exit process has suffered from neglect. However, pathogen exit is of fundamental importance because of its intimate association with dissemination, transmission, and inflammation. Hence, to fully understand virulence mechanisms of intracellular pathogens at cellular and systemic levels, it is essential to consider exit mechanisms to be a key step in infection. Exit from the host cell was initially viewed as a passive process, driven mainly by physical stress as a consequence of the explosive replication of the pathogen. It is now recognized as a complex, strategic process termed "egress," which is just as well orchestrated and temporally defined as entry into the host and relies on a dynamic interplay between host and pathogen factors. This review compares egress strategies of bacteria, pathogenic yeast, and kinetoplastid and apicomplexan parasites. Emphasis is given to recent advances in the biology of egress in mycobacteria and apicomplexans.
Collapse
|
22
|
Chong A, Child R, Wehrly TD, Rockx-Brouwer D, Qin A, Mann BJ, Celli J. Structure-Function Analysis of DipA, a Francisella tularensis Virulence Factor Required for Intracellular Replication. PLoS One 2013; 8:e67965. [PMID: 23840797 PMCID: PMC3694160 DOI: 10.1371/journal.pone.0067965] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/23/2013] [Indexed: 12/26/2022] Open
Abstract
Francisella tularensis is a highly infectious bacterium whose virulence relies on its ability to rapidly reach the macrophage cytosol and extensively replicate in this compartment. We previously identified a novel Francisella virulence factor, DipA (FTT0369c), which is required for intramacrophage proliferation and survival, and virulence in mice. DipA is a 353 amino acid protein with a Sec-dependent signal peptide, four Sel1-like repeats (SLR), and a C-terminal coiled-coil (CC) domain. Here, we determined through biochemical and localization studies that DipA is a membrane-associated protein exposed on the surface of the prototypical F. tularensis subsp. tularensis strain SchuS4 during macrophage infection. Deletion and substitution mutagenesis showed that the CC domain, but not the SLR motifs, of DipA is required for surface exposure on SchuS4. Complementation of the dipA mutant with either DipA CC or SLR domain mutants did not restore intracellular growth of Francisella, indicating that proper localization and the SLR domains are required for DipA function. Co-immunoprecipitation studies revealed interactions with the Francisella outer membrane protein FopA, suggesting that DipA is part of a membrane-associated complex. Altogether, our findings indicate that DipA is positioned at the host–pathogen interface to influence the intracellular fate of this pathogen.
Collapse
Affiliation(s)
- Audrey Chong
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
- * E-mail:
| | - Robert Child
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Tara D. Wehrly
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Dedeke Rockx-Brouwer
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Aiping Qin
- Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Barbara J. Mann
- Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jean Celli
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| |
Collapse
|
23
|
Mohapatra NP, Soni S, Rajaram MVS, Strandberg KL, Gunn JS. Type A Francisella tularensis acid phosphatases contribute to pathogenesis. PLoS One 2013; 8:e56834. [PMID: 23457625 PMCID: PMC3574111 DOI: 10.1371/journal.pone.0056834] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 01/15/2013] [Indexed: 12/02/2022] Open
Abstract
Different Francisella spp. produce five or six predicted acid phosphatases (AcpA, AcpB, AcpC, AcpD, HapA and HapB). The genes encoding the histidine acid phosphatases (hapA, hapB) and acpD of F. tularensis subsp. Schu S4 strain are truncated or disrupted. However, deletion of HapA (FTT1064) in F. tularensis Schu S4 resulted in a 33% reduction in acid phosphatase activity and loss of the four functional acid phosphatases in F. tularensis Schu S4 (ΔABCH) resulted in a>99% reduction in acid phosphatase activity compared to the wild type strain. All single, double and triple mutants tested, demonstrated a moderate decrease in mouse virulence and survival and growth within human and murine phagocytes, whereas the ΔABCH mutant showed >3.5-fold decrease in intramacrophage survival and 100% attenuation of virulence in mouse. While the Schu S4 ΔABCH strain was attenuated in the mouse model, it showed only limited protection against wild type challenge. F. tularensis Schu S4 failed to stimulate reactive oxygen species production in phagocytes, whereas infection by the ΔABCH strain stimulated 5- and 56-fold increase in reactive oxygen species production in neutrophils and human monocyte-derived macrophages, respectively. The ΔABCH mutant but not the wild type strain strongly co-localized with p47phox and replicated in macrophages isolated from p47phox knockout mice. Thus, F. tularensis Schu S4 acid phosphatases, including the truncated HapA, play a major role in intramacrophage survival and virulence of this human pathogen.
Collapse
Affiliation(s)
- Nrusingh P. Mohapatra
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Shilpa Soni
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Murugesan V. S. Rajaram
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Kristi L. Strandberg
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - John S. Gunn
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
24
|
Production of outer membrane vesicles and outer membrane tubes by Francisella novicida. J Bacteriol 2012; 195:1120-32. [PMID: 23264574 DOI: 10.1128/jb.02007-12] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella spp. are highly infectious and virulent bacteria that cause the zoonotic disease tularemia. Knowledge is lacking for the virulence factors expressed by Francisella and how these factors are secreted and delivered to host cells. Gram-negative bacteria constitutively release outer membrane vesicles (OMV), which may function in the delivery of virulence factors to host cells. We identified growth conditions under which Francisella novicida produces abundant OMV. Purification of the vesicles revealed the presence of tube-shaped vesicles in addition to typical spherical OMV, and examination of whole bacteria revealed the presence of tubes extending out from the bacterial surface. Recently, both prokaryotic and eukaryotic cells have been shown to produce membrane-enclosed projections, termed nanotubes, which appear to function in cell-cell communication and the exchange of molecules. In contrast to these previously characterized structures, the F. novicida tubes are produced in liquid as well as on solid medium and are derived from the OM rather than the cytoplasmic membrane. The production of the OMV and tubes (OMV/T) by F. novicida was coordinately regulated and responsive to both growth medium and growth phase. Proteomic analysis of purified OMV/T identified known Francisella virulence factors among the constituent proteins, suggesting roles for the vesicles in pathogenesis. In support of this, production of OM tubes by F. novicida was stimulated during infection of macrophages and addition of purified OMV/T to macrophages elicited increased release of proinflammatory cytokines. Finally, vaccination with purified OMV/T protected mice from subsequent challenge with highly lethal doses of F. novicida.
Collapse
|
25
|
Silva MT, Pestana NTS. The in vivo extracellular life of facultative intracellular bacterial parasites: role in pathogenesis. Immunobiology 2012; 218:325-37. [PMID: 22795971 DOI: 10.1016/j.imbio.2012.05.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/23/2012] [Accepted: 05/16/2012] [Indexed: 01/14/2023]
Abstract
Classically labeled facultative intracellular pathogens are characterized by the ability to have an intracellular phase in the host, which is required for pathogenicity, while capable of extracellular growth in vitro. The ability of these bacteria to replicate in cell-free conditions is usually assessed by culture in artificial bacteriological media. However, the extracellular growth ability of these pathogens may also be expressed by a phase of extracellular infection in the natural setting of the host with pathologic consequences, an ability that adds to the pathogenic potential of the infectious agent. This infective capability to grow in the extracellular sites of the host represents an additional virulence attribute of those pathogens which may lead to severe outcomes. Here we discuss examples of infectious diseases where the in vivo infective extracellular life is well documented, including infections by Francisella tularensis, Yersinia pestis, Burkholderia pseudomallei, Burkholderia cenocepacia, Salmonella enterica serovar Typhimurium and Edwardsiella tarda. The occurrence of a phase of systemic dissemination with extracellular multiplication during progressive infections by facultative intracellular bacterial pathogens has been underappreciated, with most studies exclusively centered on the intracellular phase of the infections. The investigation of the occurrence of a dual lifestyle in the host among bacterial pathogens in general should be extended and likely will reveal more cases of infectious diseases with a dual infective intracellular/extracellular pattern.
Collapse
Affiliation(s)
- Manuel T Silva
- Institute for Molecular and Cell Biology, University of Porto, Porto, Portugal
| | | |
Collapse
|
26
|
Peters KN, Anderson DM. Modulation of host cell death pathways by Yersinia species and the type III effector YopK. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 954:229-36. [PMID: 22782768 DOI: 10.1007/978-1-4614-3561-7_29] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Kristen N Peters
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | | |
Collapse
|
27
|
Jones CL, Weiss DS. TLR2 signaling contributes to rapid inflammasome activation during F. novicida infection. PLoS One 2011; 6:e20609. [PMID: 21698237 PMCID: PMC3116832 DOI: 10.1371/journal.pone.0020609] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 05/05/2011] [Indexed: 11/19/2022] Open
Abstract
Background Early detection of microorganisms by the innate immune system is provided by surface-expressed and endosomal pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs). Detection of microbial components by TLRs initiates a signaling cascade leading to the expression of proinflammatory cytokines including IL-6 and IL-1β. Some intracellular bacteria subvert the TLR response by rapidly escaping the phagosome and entering the cytosol. However, these bacteria may be recognized by the inflammasome, a multi-protein complex comprised of a sensor protein, ASC and the cysteine protease caspase-1. Inflammasome activation leads to release of the proinflammatory cytokines IL-1β and IL-18 and death of the infected cell, an important host defense that eliminates the pathogen's replicative niche. While TLRs and inflammasomes are critical for controlling bacterial infections, it is unknown whether these distinct host pathways cooperate to activate defenses against intracellular bacteria. Methodology/Significant Findings Using the intracellular bacterium Francisella novicida as a model, we show that TLR2−/− macrophages exhibited delayed inflammasome activation compared to wild-type macrophages as measured by inflammasome assembly, caspase-1 activation, cell death and IL-18 release. TLR2 also contributed to inflammasome activation in response to infection by the cytosolic bacterium Listeria monocytogenes. Components of the TLR2 signaling pathway, MyD88 and NF-κB, were required for rapid inflammasome activation. Furthermore, TLR2−/− mice exhibited lower levels of cell death, caspase-1 activation, and IL-18 production than wild-type mice upon F. novicida infection. Conclusions/Significance These results show that TLR2 is required for rapid inflammasome activation in response to infection by cytosolic bacterial pathogens. In addition to further characterizing the role of TLR2 in host defense, these findings broaden our understanding of how the host integrates signals from spatiotemporally separated PRRs to coordinate an innate response against intracellular bacteria.
Collapse
Affiliation(s)
- Crystal L. Jones
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - David S. Weiss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
28
|
Al-Khodor S, Abu Kwaik Y. Triggering Ras signalling by intracellular Francisella tularensis through recruitment of PKCα and βI to the SOS2/GrB2 complex is essential for bacterial proliferation in the cytosol. Cell Microbiol 2011; 12:1604-21. [PMID: 20618341 DOI: 10.1111/j.1462-5822.2010.01494.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intracellular proliferation of Francisella tularensis is essential for manifestation of the fatal disease tularaemia, and is classified as a category A bioterrorism agent. The F. tularensis-containing phagosome (FCP) matures into a late endosome-like phagosome with limited fusion to lysosomes, followed by rapid bacterial escape into the cytosol. The Francisella pathogenicity island (FPI) encodes a type VI-like secretion system, and the FPI-encoded IglC is essential for evasion of lysosomal fusion and phagosomal escape. Many host signalling events are likely to be modulated by F. tularensis to render the cell permissive for intracellular proliferation but they are not fully understood. Here we show that within 15 min of infection, intracellular F. tularensis ssp. novicida triggers IglC-dependent temporal activation of Ras, but attached extracellular bacteria fail to trigger Ras activation, which has never been shown for other intracellular pathogens. Intracellular F. tularensis ssp. novicida triggers activation of Ras through recruitment of PKCα and PKCβI to the SOS2/GrB2 complex. Silencing of SOS2, GrB2 and PKCα and PKCβI by RNAi has no effect on evasion of lysosomal fusion and bacterial escape into the cytosol but renders the cytosol non-permissive for replication of F. tularensis ssp. novicida. Since Ras activation promotes cell survival, we show that silencing of SOS2, GrB2 and PKCα and βI is associated with rapid early activation of caspase-3 within 8 h post infection. However, silencing of SOS2, GrB2 and PKCα and βI does not affect phosphorylation of Akt or Erk, indicating that activation of the PI3K/Akt and the Erk signalling cascade are independent of the F. tularensis-triggered Ras activation. We conclude that intracellular F. tularensis ssp. novicida triggers temporal and early activation of Ras through the SOS2/GrB2/PKCα/PKCβI quaternary complex. Temporal and rapid trigger of Ras signalling by intracellular F. tularensis is essential for intracellular bacterial proliferation within the cytosol, and this is associated with downregulation of early caspase-3 activation.
Collapse
Affiliation(s)
- Souhaila Al-Khodor
- Department of Microbiology and Immunology, College of Medicine, Department of Biology, University of Louisville, Louisville, KY 40202, USA
| | | |
Collapse
|
29
|
Novosad J, Holicka M, Novosadova M, Krejsek J, Krcmova I. Rapid onset of ICAM-1 expression is a marker of effective macrophages activation during infection of Francisella tularensis LVS in vitro. Folia Microbiol (Praha) 2011; 56:149-54. [PMID: 21476047 DOI: 10.1007/s12223-011-0028-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 01/25/2011] [Indexed: 11/26/2022]
Abstract
Francisella tularensis is capable to modulate immunobiological activities of the host cells. We focused on the expression of ICAM-1 (CD54) on J774.2 mouse macrophage cell line infected by F. tularensis live vaccine strain (LVS) in vitro as a putative marker of subsequent elimination of infection. J774.2 cell line cells were infected by F. tularensis LVS strain (multiplicity of infection, 1:100). Cell cultures were stimulated either 3 h before infection or 3 h after infection by either lipopolysaccharide (LPS) or interferon γ (IFN-γ). The expression of ICAM-1 was determined by flow cytometry 6 h after infection. The intensity of ICAM-1 expression after 6 h of J774.2 macrophage cells infection by F. tularensis is very sensitive indicator of the effective macrophages stimulation resulting in the elimination of F. tularensis infection. The mean fluorescence intensity MFI = 49.8 is set-up by our experiments as a reliable threshold of the effective elimination of F. tularensis experimental infection with 83.3% sensitivity and 96.7% specificity, respectively. Simultaneous stimulation of J774.2 macrophage cells by LPS and IFN-γ was essential to elicit the elimination of F. tularensis infection. The ICAM-1 expression determined by flow cytometry can be considered to be highly sensitive and specific approach to predict elimination of F. tularensis infection by J774.2 macrophages.
Collapse
Affiliation(s)
- J Novosad
- Department of Clinical Immunology, Faculty of Medicine and University Hospital in Hradec Kralove, Charles University Prague, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | | | | | | | | |
Collapse
|
30
|
Phagocytic receptors dictate phagosomal escape and intracellular proliferation of Francisella tularensis. Infect Immun 2011; 79:2204-14. [PMID: 21422184 DOI: 10.1128/iai.01382-10] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Francisella tularensis, the causative agent of tularemia, survives and proliferates within macrophages of the infected host as part of its pathogenic strategy, through an intracellular life cycle that includes phagosomal escape and extensive proliferation within the macrophage cytosol. Various in vitro models of Francisella-macrophage interactions have been developed, using either opsonic or nonopsonic phagocytosis, and have generated discrepant results on the timing and extent of Francisella phagosomal escape. Here we have investigated whether either complement or antibody opsonization of the virulent prototypical type A strain Francisella tularensis subsp. tularensis Schu S4 affects its intracellular cycle within primary murine bone marrow-derived macrophages. Opsonization of Schu S4 with either human serum or purified IgG enhanced phagocytosis but restricted phagosomal escape and intracellular proliferation. Opsonization of Schu S4 with either fresh serum or purified antibodies redirected bacteria from the mannose receptor (MR) to the complement receptor CR3, the scavenger receptor A (SRA), and the Fcγ receptor (FcγR), respectively. CR3-mediated uptake delayed maturation of the early Francisella-containing phagosome (FCP) and restricted phagosomal escape, while FcγR-dependent phagocytosis was associated with superoxide production in the early FCP and restricted phagosomal escape and intracellular growth in an NADPH oxidase-dependent manner. Taken together, these results demonstrate that opsonophagocytic receptors alter the intracellular fate of Francisella by delivering bacteria through phagocytic pathways that restrict phagosomal escape and intracellular proliferation.
Collapse
|
31
|
Gavrilin MA, Wewers MD. Francisella Recognition by Inflammasomes: Differences between Mice and Men. Front Microbiol 2011; 2:11. [PMID: 21687407 PMCID: PMC3109362 DOI: 10.3389/fmicb.2011.00011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 01/19/2011] [Indexed: 12/31/2022] Open
Abstract
Pathogen recognition by intracellular sensors involves the assembly of a caspase-1 activation machine termed the inflammasome. Intracellular pathogens like Francisella that gain access to the cytosolic detection systems are useful tools to uncover the details of caspase-1 activation events. This review overviews Francisella function in the mononuclear phagocyte with particular attention to inflammasome versus pyroptosome roles and outlines differences between mouse and human caspase-1 activation pathways. Specific attention is placed on functional differences between human and murine pyrin as an intracellular recognition molecule for Francisella.
Collapse
Affiliation(s)
- Mikhail A Gavrilin
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, The Center for Microbial Interface Biology, The Ohio State University Columbus, OH, USA
| | | |
Collapse
|
32
|
Asare R, Kwaik YA. Exploitation of host cell biology and evasion of immunity by francisella tularensis. Front Microbiol 2011; 1:145. [PMID: 21687747 PMCID: PMC3109322 DOI: 10.3389/fmicb.2010.00145] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 12/21/2010] [Indexed: 12/13/2022] Open
Abstract
Francisella tularensis is an intracellular bacterium that infects humans and many small mammals. During infection, F. tularensis replicates predominantly in macrophages but also proliferate in other cell types. Entry into host cells is mediate by various receptors. Complement-opsonized F. tularensis enters into macrophages by looping phagocytosis. Uptake is mediated in part by Syk, which may activate actin rearrangement in the phagocytic cup resulting in the engulfment of F. tularensis in a lipid raft rich phagosome. Inside the host cells, F. tularensis resides transiently in an acidified late endosome-like compartment before disruption of the phagosomal membrane and escape into the cytosol, where bacterial proliferation occurs. Modulation of phagosome biogenesis and escape into the cytosol is mediated by the Francisella pathogenicity island-encoded type VI-like secretion system. Whilst inside the phagosome, F. tularensis temporarily induce proinflammatory cytokines in PI3K/Akt-dependent manner, which is counteracted by the induction of SHIP that negatively regulates PI3K/Akt activation and promotes bacterial escape into the cytosol. Interestingly, F. tularensis subverts CD4 T cells-mediated killing by inhibiting antigen presentation by activated macrophages through ubiquitin-dependent degradation of MHC II molecules on activated macrophages. In the cytosol, F. tularensis is recognized by the host cell inflammasome, which is down-regulated by F. tularensis that also inhibits caspase-1 and ASC activity. During late stages of intracellular proliferation, caspase-3 is activated but apoptosis is delayed through activation of NF-κB and Ras, which ensures cell viability.
Collapse
Affiliation(s)
- Rexford Asare
- Department of Microbiology and Immunology, School of Medicine, University of Louisville Louisville, KY, USA
| | | |
Collapse
|
33
|
Chong A, Celli J. The francisella intracellular life cycle: toward molecular mechanisms of intracellular survival and proliferation. Front Microbiol 2010; 1:138. [PMID: 21687806 PMCID: PMC3109316 DOI: 10.3389/fmicb.2010.00138] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 12/05/2010] [Indexed: 11/13/2022] Open
Abstract
The tularemia-causing bacterium Francisella tularensis is a facultative intracellular organism with a complex intracellular lifecycle that ensures its survival and proliferation in a variety of mammalian cell types, including professional phagocytes. Because this cycle is essential to Francisella pathogenesis and virulence, much research has focused on deciphering the mechanisms of its intracellular survival and replication and characterizing both bacterial and host determinants of the bacterium's intracellular cycle. Studies of various strains and host cell models have led to the consensual paradigm of Francisella as a cytosolic pathogen, but also to some controversy about its intracellular cycle. In this review, we will detail major findings that have advanced our knowledge of Francisella intracellular survival strategies and also attempt to reconcile discrepancies that exist in our molecular understanding of the Francisella–phagocyte interactions.
Collapse
Affiliation(s)
- Audrey Chong
- Tularemia Pathogenesis Section, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton, MT, USA
| | | |
Collapse
|
34
|
Meibom KL, Charbit A. Francisella tularensis metabolism and its relation to virulence. Front Microbiol 2010; 1:140. [PMID: 21687763 PMCID: PMC3109416 DOI: 10.3389/fmicb.2010.00140] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 12/13/2010] [Indexed: 01/08/2023] Open
Abstract
Francisella tularensis is a Gram-negative bacterium capable of causing the zoonotic disease tularaemia in a large number of mammalian species and in arthropods. F. tularensis is a facultative intracellular bacterium that infects and replicates in vivo mainly inside macrophages. During its systemic dissemination, F. tularensis must cope with very different life conditions (such as survival in different target organs or tissues and/or survival in the blood stream…) and may thus encounter a broad variety of carbon substrates, nitrogen, phosphor, and sulfur sources, as well as very low concentrations of essential ions. The development of recent genome-wide genetic screens have led to the identification of hundreds of genes participating to variable extents to Francisella virulence. Remarkably, an important proportion of the genes identified are related to metabolic and nutritional functions. However, the relationship between nutrition and the in vivo life cycle of F. tularensis is yet poorly understood. In this review, we will address the importance of metabolism and nutrition for F. tularensis pathogenesis, focusing specifically on amino acid and carbohydrate requirements.
Collapse
|
35
|
Barel M, Meibom K, Charbit A. Nucleolin, a shuttle protein promoting infection of human monocytes by Francisella tularensis. PLoS One 2010; 5:e14193. [PMID: 21152024 PMCID: PMC2995743 DOI: 10.1371/journal.pone.0014193] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 11/10/2010] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Francisella tularensis is a highly virulent facultative intracellular bacterium, disseminating in vivo mainly within host mononuclear phagocytes. After entry into macrophages, F. tularensis initially resides in a phagosomal compartment, whose maturation is then arrested. Bacteria escape rapidly into the cytoplasm, where they replicate freely. We recently demonstrated that nucleolin, an eukaryotic protein able to traffic from the nucleus to the cell surface, acted as a surface receptor for F. tularensis LVS on human monocyte-like THP-1 cells. METHODOLOGY/PRINCIPAL FINDINGS Here, we followed the fate of nucleolin once F. tularensis has been endocytosed. We first confirmed by siRNA silencing experiments that expression of nucleolin protein was essential for binding of LVS on human macrophage-type THP-1 cells. We then showed that nucleolin co-localized with intracellular bacteria in the phagosomal compartment. Strikingly, in that compartment, nucleolin also co-localized with LAMP-1, a late endosomal marker. Co-immunoprecipation assays further demonstrated an interaction of nucleolin with LAMP-1. Co-localization of nucleolin with LVS was no longer detectable at 24 h when bacteria were multiplying in the cytoplasm. In contrast, with an iglC mutant of LVS, which remains trapped into the phagosomal compartment, or with inert particles, nucleolin/bacteria co-localization remained almost constant. CONCLUSIONS/SIGNIFICANCE We herein confirm the importance of nucleolin expression for LVS binding and its specificity as nucleolin is not involved in binding of another intracellular pathogen as L. monocytogenes or an inert particle. Association of nucleolin with F. tularensis during infection continues intracellularly after endocytosis of the bacteria. The present work therefore unravels for the first time the presence of nucleolin in the phagosomal compartment of macrophages.
Collapse
Affiliation(s)
- Monique Barel
- Université Paris Descartes, Faculté de Médecine Necker Enfants-Malades, Paris, France.
| | | | | |
Collapse
|