1
|
Li J, Zhang G, Zhi F, Zhai Y, Zhou D, Chen H, Lin P, Tang K, Liu W, Jin Y, Wang A. BtpB inhibits innate inflammatory responses in goat alveolar macrophages through the TLR/NF-κB pathway and NLRP3 inflammasome during Brucella infection. Microb Pathog 2022; 166:105536. [DOI: 10.1016/j.micpath.2022.105536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/15/2022] [Accepted: 04/10/2022] [Indexed: 01/19/2023]
|
2
|
Hiyoshi H, English BC, Diaz-Ochoa VE, Wangdi T, Zhang LF, Sakaguchi M, Haneda T, Tsolis RM, Bäumler AJ. Virulence factors perforate the pathogen-containing vacuole to signal efferocytosis. Cell Host Microbe 2022; 30:163-170.e6. [PMID: 34951948 PMCID: PMC8831471 DOI: 10.1016/j.chom.2021.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/20/2021] [Accepted: 11/30/2021] [Indexed: 12/11/2022]
Abstract
Intracellular pathogens commonly reside within macrophages to find shelter from humoral defenses, but host cell death can expose them to the extracellular milieu. We find intracellular pathogens solve this dilemma by using virulence factors to generate a complement-dependent find-me signal that initiates uptake by a new phagocyte through efferocytosis. During macrophage death, Salmonella uses a type III secretion system to perforate the membrane of the pathogen-containing vacuole (PCV), thereby triggering complement deposition on bacteria entrapped in pore-induced intracellular traps (PITs). In turn, complement activation signals neutrophil efferocytosis, a process that shelters intracellular bacteria from the respiratory burst. Similarly, Brucella employs its type IV secretion system to perforate the PCV membrane, which induces complement deposition on bacteria entrapped in PITs. Collectively, this work identifies virulence factor-induced perforation of the PCV as a strategy of intracellular pathogens to generate a find-me signal for efferocytosis.
Collapse
Affiliation(s)
- Hirotaka Hiyoshi
- Department of Bacteriology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Bevin C English
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Vladimir E Diaz-Ochoa
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Tamding Wangdi
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Lillian F Zhang
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Miako Sakaguchi
- Central Laboratory, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Takeshi Haneda
- Laboratory of Microbiology, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Renée M Tsolis
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA.
| |
Collapse
|
3
|
IRE1α-driven inflammation promotes clearance of Citrobacter rodentium infection. Infect Immun 2021; 90:e0048121. [PMID: 34748367 PMCID: PMC8788755 DOI: 10.1128/iai.00481-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is intimately linked with inflammation in response to pathogenic infections. ER stress occurs when cells experience a buildup of misfolded or unfolded protein during times of perturbation, such as infections, which facilitates the unfolded protein response (UPR). The UPR involves multiple host pathways in an attempt to re-establish homeostasis, which oftentimes leads to inflammation and cell death if unresolved. The UPR is activated to help resolve some bacterial infections, and the IRE1α pathway is especially critical in mediating inflammation. To understand the role of the IRE1α pathway of the UPR during enteric bacterial infection, we employed Citrobacter rodentium to study host-pathogen interactions in intestinal epithelial cells and the murine gastrointestinal (GI) tract. C. rodentium is an enteric mouse pathogen that is similar to the human pathogens enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC, respectively), which have limited small animal models. Here, we demonstrate that both C. rodentium and EPEC induced the UPR in intestinal epithelial cells. UPR induction during C. rodentium infection correlated with the onset of inflammation in bone marrow-derived macrophages (BMDMs). Our previous work implicated IRE1α and NOD1/2 in ER stress-induced inflammation, which we observed were also required for pro-inflammatory gene induction during C. rodentium infection. C. rodentium induced IRE1α-dependent inflammation in mice, and inhibiting IRE1α led to a dysregulated inflammatory response and delayed clearance of C. rodentium. This study demonstrates that ER stress aids inflammation and clearance of C. rodentium through a mechanism involving the IRE1α-NOD1/2 axis.
Collapse
|
4
|
Mirzaei R, Sholeh M, Jalalifar S, Zafari E, Kazemi S, Rasouli-Saravani A, Karampoor S, Yousefimashouf R. Immunometabolism in human brucellosis: An emerging field of investigation. Microb Pathog 2021; 158:105115. [PMID: 34332069 DOI: 10.1016/j.micpath.2021.105115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 01/16/2023]
Abstract
In recent years, extreme attention has been focused on the role of immunometabolism in the regulation of immune cell responses in healthy individuals during infection, autoimmunity, and cancer. In the infection biology area, it has been shown that there is a close relationship between the immune system and the host metabolic changes. Brucella species is an intracellular coccobacillus that infects humans and mammals, which led to brucellosis. Brucella species with host-specific evolutionary mechanisms allow it to hide from or manipulate cellular immunity and achieve intracellular persistence. Intracellular bacterial pathogens such as Brucella species also employ host cell resources to replicate and persist inside the host. Targeting these host systems is one promising strategy for developing novel antimicrobials to tackle intracellular infections. This study will summarize the role of metabolic reprogramming in immune cells and their relationship to brucellosis.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Mohammad Sholeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saba Jalalifar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Zafari
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sima Kazemi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ashkan Rasouli-Saravani
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
5
|
Roop RM, Barton IS, Hopersberger D, Martin DW. Uncovering the Hidden Credentials of Brucella Virulence. Microbiol Mol Biol Rev 2021; 85:e00021-19. [PMID: 33568459 PMCID: PMC8549849 DOI: 10.1128/mmbr.00021-19] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Bacteria in the genus Brucella are important human and veterinary pathogens. The abortion and infertility they cause in food animals produce economic hardships in areas where the disease has not been controlled, and human brucellosis is one of the world's most common zoonoses. Brucella strains have also been isolated from wildlife, but we know much less about the pathobiology and epidemiology of these infections than we do about brucellosis in domestic animals. The brucellae maintain predominantly an intracellular lifestyle in their mammalian hosts, and their ability to subvert the host immune response and survive and replicate in macrophages and placental trophoblasts underlies their success as pathogens. We are just beginning to understand how these bacteria evolved from a progenitor alphaproteobacterium with an environmental niche and diverged to become highly host-adapted and host-specific pathogens. Two important virulence determinants played critical roles in this evolution: (i) a type IV secretion system that secretes effector molecules into the host cell cytoplasm that direct the intracellular trafficking of the brucellae and modulate host immune responses and (ii) a lipopolysaccharide moiety which poorly stimulates host inflammatory responses. This review highlights what we presently know about how these and other virulence determinants contribute to Brucella pathogenesis. Gaining a better understanding of how the brucellae produce disease will provide us with information that can be used to design better strategies for preventing brucellosis in animals and for preventing and treating this disease in humans.
Collapse
Affiliation(s)
- R Martin Roop
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Ian S Barton
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Dariel Hopersberger
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Daniel W Martin
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
6
|
B Cells Inhibit CD4 + T Cell-Mediated Immunity to Brucella Infection in a Major Histocompatibility Complex Class II-Dependent Manner. Infect Immun 2020; 88:IAI.00075-20. [PMID: 32071068 DOI: 10.1128/iai.00075-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 01/18/2023] Open
Abstract
Brucella spp. are facultative intracellular bacteria notorious for their ability to induce a chronic, and often lifelong, infection known as brucellosis. To date, no licensed vaccine exists for prevention of human disease, and mechanisms underlying chronic illness and immune evasion remain elusive. We and others have observed that B cell-deficient mice challenged with Brucella display reduced bacterial burden following infection, but the underlying mechanism has not been clearly defined. Here, we show that at 1 month postinfection, B cell deficiency alone enhanced resistance to splenic infection ∼100-fold; however, combined B and T cell deficiency did not impact bacterial burden, indicating that B cells only enhance susceptibility to infection when T cells are present. Therefore, we investigated whether B cells inhibit T cell-mediated protection against Brucella Using B and T cell-deficient Rag1-/- animals as recipients, we demonstrate that adoptive transfer of CD4+ T cells alone confers marked protection against Brucella melitensis that is abrogated by cotransfer of B cells. Interestingly, depletion of CD4+ T cells from B cell-deficient, but not wild-type, mice enhanced susceptibility to infection, further confirming that CD4+ T cell-mediated immunity against Brucella is inhibited by B cells. In addition, we found that the ability of B cells to suppress CD4+ T cell-mediated immunity and modulate CD4+ T cell effector responses during infection was major histocompatibility complex class II (MHCII)-dependent. Collectively, these findings indicate that B cells modulate CD4+ T cell function through an MHCII-dependent mechanism which enhances susceptibility to Brucella infection.
Collapse
|
7
|
Stranahan LW, Khalaf OH, Garcia-Gonzalez DG, Arenas-Gamboa AM. Characterization of Brucella canis infection in mice. PLoS One 2019; 14:e0218809. [PMID: 31220185 PMCID: PMC6586350 DOI: 10.1371/journal.pone.0218809] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/10/2019] [Indexed: 01/18/2023] Open
Abstract
Canine brucellosis, caused by Brucella canis, is a disease of dogs and represents a public health concern as it can be transmitted to humans. Canine brucellosis is on the rise in the United States and there is currently no vaccine for use in dogs. Mice have been extensively utilized to investigate host-pathogen interactions and vaccine candidates for smooth Brucella species and could serve a similar role for studying B. canis. However, comparatively little is known about B. canis infection in mice. The objective of this study was to characterize the kinetics of colonization and pathogenicity of B. canis in mice in order to evaluate the mouse as a model for studying this pathogen. C57BL/6 mice were inoculated intraperitoneally with 105, 107, or 109 CFU of Brucella canis RM6/66 and euthanized 1-, 2-, 4-, 6-, 9-, and 12-weeks post-inoculation. B. canis induced splenomegaly in mice infected with 109 CFU at 1- and 2 weeks post-inoculation while no gross lesions were observed in other dose groups. Infection at the two higher doses resulted in dose-dependent granulomatous hepatitis and histiocytic infiltration of the spleen and mesenteric lymph nodes by 1-2 weeks. B. canis was cultured from the liver, spleen, uterus, bone marrow, lung, and kidney in all groups with colonization declining at a slow but steady rate throughout the experiment. Clearance was achieved by 9 weeks 105 CFU group and by 12 weeks in the 107 CFU group, while B. canis persisted in the spleen until 12 weeks in the highest dose group. Although B. canis does not demonstrate significant replication in C57BL/6 mice, it has the ability to establish an infection, induce splenomegaly, and persist for several weeks in multiple organs. Moreover, 1 x 107 CFU appears to be a suitable challenge dose for investigating vaccine safety.
Collapse
Affiliation(s)
- Lauren W. Stranahan
- Department of Veterinary Pathobiology, Texas A&M University, College of Veterinary Medicine, College Station, TX, United States of America
| | - Omar H. Khalaf
- Department of Veterinary Pathobiology, Texas A&M University, College of Veterinary Medicine, College Station, TX, United States of America
- Department of Veterinary Pathology & Poultry Diseases, College of Veterinary Medicine, University of Baghdad, Baghdad, Iraq
| | - Daniel G. Garcia-Gonzalez
- Department of Veterinary Pathobiology, Texas A&M University, College of Veterinary Medicine, College Station, TX, United States of America
| | - Angela M. Arenas-Gamboa
- Department of Veterinary Pathobiology, Texas A&M University, College of Veterinary Medicine, College Station, TX, United States of America
- * E-mail:
| |
Collapse
|
8
|
Brucella neotomae Recapitulates Attributes of Zoonotic Human Disease in a Murine Infection Model. Infect Immun 2018; 87:IAI.00255-18. [PMID: 30373892 DOI: 10.1128/iai.00255-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 10/23/2018] [Indexed: 01/03/2023] Open
Abstract
Members of the genus Brucella are Gram-negative pathogens that cause chronic systemic infection in farm animals and zoonotic infection in humans. Study of the genus Brucella has been hindered by the need for biosafety level 3 select agent containment. Brucella neotomae, originally isolated from the desert pack rat, presented an opportunity to develop an alternative, non-select agent experimental model. Our prior in vitro work indicated that the cell biology and type IV secretion system (T4SS) dependence of B. neotomae intracellular replication were similar to observations for human-pathogenic select agent Brucella species. Therefore, here, we investigated the pathobiology of B. neotomae infection in the BALB/c mouse. During a sustained infectious course, B. neotomae replicated and persisted in reticuloendothelial organs. Bioluminescent imaging and histopathological and PCR-based analysis demonstrated that the T4SS contributed to efficient early infection of the liver, spleen, and lymph nodes; granuloma formation and hepatosplenomegaly; and early induction of Th1-associated cytokine gene expression. The infectious course and pathologies in the murine model showed similarity to prior observations of primate and native host infection with zoonotic Brucella species. Therefore, the B. neotomae BALB/c infection model offers a promising system to accelerate and complement experimental work in the genus Brucella.
Collapse
|
9
|
Utilization of Host Polyamines in Alternatively Activated Macrophages Promotes Chronic Infection by Brucella abortus. Infect Immun 2018; 86:IAI.00458-17. [PMID: 29203548 DOI: 10.1128/iai.00458-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/29/2017] [Indexed: 12/15/2022] Open
Abstract
Treatment of intracellular bacterial pathogens with antibiotic therapy often requires a long course of multiple drugs. A barrier to developing strategies that enhance antibiotic efficacy against these pathogens is our poor understanding of the intracellular nutritional environment that maintains bacterial persistence. The intracellular pathogen Brucella abortus survives and replicates preferentially in alternatively activated macrophages (AAMs); however, knowledge of the metabolic adaptations promoting exploitation of this niche is limited. Here we show that one mechanism promoting enhanced survival in AAMs is a shift in macrophage arginine utilization from production of nitric oxide (NO) to biosynthesis of polyamines, induced by interleukin 4 (IL-4)/IL-13 treatment. Production of polyamines by infected AAMs promoted both intracellular survival of B. abortus and chronic infection in mice, as inhibition of macrophage polyamine synthesis or inactivation of the putative putrescine transporter encoded by potIHGF reduced both intracellular survival in AAMs and persistence in mice. These results demonstrate that increased intracellular availability of polyamines induced by arginase-1 expression in IL-4/IL-13-induced AAMs promotes chronic persistence of B. abortus within this niche and suggest that targeting of this pathway may aid in eradicating chronic infection.
Collapse
|
10
|
Rossetti CA, Drake KL, Lawhon SD, Nunes JS, Gull T, Khare S, Adams LG. Systems Biology Analysis of Temporal In vivo Brucella melitensis and Bovine Transcriptomes Predicts host:Pathogen Protein-Protein Interactions. Front Microbiol 2017; 8:1275. [PMID: 28798726 PMCID: PMC5529337 DOI: 10.3389/fmicb.2017.01275] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/26/2017] [Indexed: 01/13/2023] Open
Abstract
To date, fewer than 200 gene-products have been identified as Brucella virulence factors, and most were characterized individually without considering how they are temporally and coordinately expressed or secreted during the infection process. Here, we describe and analyze the in vivo temporal transcriptional profile of Brucella melitensis during the initial 4 h interaction with cattle. Pathway analysis revealed an activation of the "Two component system" providing evidence that the in vivo Brucella sense and actively regulate their metabolism through the transition to an intracellular lifestyle. Contrarily, other Brucella pathways involved in virulence such as "ABC transporters" and "T4SS system" were repressed suggesting a silencing strategy to avoid stimulation of the host innate immune response very early in the infection process. Also, three flagellum-encoded loci (BMEII0150-0168, BMEII1080-1089, and BMEII1105-1114), the "flagellar assembly" pathway and the cell components "bacterial-type flagellum hook" and "bacterial-type flagellum" were repressed in the tissue-associated B. melitensis, while RopE1 sigma factor, a flagellar repressor, was activated throughout the experiment. These results support the idea that Brucella employ a stealthy strategy at the onset of the infection of susceptible hosts. Further, through systems-level in silico host:pathogen protein-protein interactions simulation and correlation of pathogen gene expression with the host gene perturbations, we identified unanticipated interactions such as VirB11::MAPK8IP1; BtaE::NFKBIA, and 22 kDa OMP precursor::BAD and MAP2K3. These findings are suggestive of new virulence factors and mechanisms responsible for Brucella evasion of the host's protective immune response and the capability to maintain a dormant state. The predicted protein-protein interactions and the points of disruption provide novel insights that will stimulate advanced hypothesis-driven approaches toward revealing a clearer understanding of new virulence factors and mechanisms influencing the pathogenesis of brucellosis.
Collapse
Affiliation(s)
- Carlos A Rossetti
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Science, Texas A&M UniversityCollege Station, TX, United States
| | | | - Sara D Lawhon
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Science, Texas A&M UniversityCollege Station, TX, United States
| | - Jairo S Nunes
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Science, Texas A&M UniversityCollege Station, TX, United States
| | - Tamara Gull
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Science, Texas A&M UniversityCollege Station, TX, United States
| | - Sangeeta Khare
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Science, Texas A&M UniversityCollege Station, TX, United States
| | - Leslie G Adams
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Science, Texas A&M UniversityCollege Station, TX, United States
| |
Collapse
|
11
|
Abstract
Many bacterial pathogens can cause acute infections that are cleared with the onset of adaptive immunity, but a subset of these pathogens can establish persistent, and sometimes lifelong, infections. While bacteria that cause chronic infections are phylogenetically diverse, they share common features in their interactions with the host that enable a protracted period of colonization. This article will compare the persistence strategies of two chronic pathogens from the Proteobacteria, Brucella abortus and Salmonella enterica serovar Typhi, to consider how these two pathogens, which are very different at the genomic level, can utilize common strategies to evade immune clearance to cause chronic intracellular infections of the mononuclear phagocyte system.
Collapse
|
12
|
NOD1 and NOD2 signalling links ER stress with inflammation. Nature 2016; 532:394-7. [PMID: 27007849 PMCID: PMC4869892 DOI: 10.1038/nature17631] [Citation(s) in RCA: 362] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 03/03/2016] [Indexed: 12/18/2022]
Abstract
Endoplasmic reticulum (ER) stress is a major contributor to inflammatory diseases, such as Crohn’s disease and type 2 diabetes1,2. ER stress induces the unfolded protein response (UPR), which involves activation of three transmembrane receptors, ATF6 (activating transcription factor 6), PERK (protein kinase RNA-like endoplasmic reticulum kinase) and IRE1α (inositol-requiring enzyme 1α)3 (Extended Data figure 1a). Once activated, IRE1α recruits TRAF2 (TNF receptor-associated factor 2) to the ER membrane to initiate inflammatory responses via the nuclear factor kappa B (NF-κB) pathway4. Inflammation is commonly triggered when pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs) or nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), detect tissue damage or microbial infection. However, it is not clear which PRRs play a major role in inducing inflammation during ER stress. Here we show that NOD1 and NOD2, two members of the NLR family of PRRs, are important mediators of ER stress-induced inflammation. The ER stress inducers thapsigargin and dithiothreitol (DTT) triggered production of the pro-inflammatory cytokine interleukin (IL)-6 in a NOD1/2-dependent fashion. Inflammation and IL-6 production triggered by infection with Brucella abortus, which induces ER stress by injecting the type IV secretion system (T4SS) effector protein VceC into host cells5, was TRAF2, NOD1/2 and RIP2-dependent and could be blunted by treatment with the ER-stress inhibitor tauroursodeoxycholate (TUDCA) or an IRE1α kinase inhibitor. The association of NOD1 and NOD2 with pro-inflammatory responses induced by the IRE1α/TRAF2 signaling pathway provides a novel link between innate immunity and ER stress-induced inflammation.
Collapse
|
13
|
Ahmed W, Zheng K, Liu ZF. Establishment of Chronic Infection: Brucella's Stealth Strategy. Front Cell Infect Microbiol 2016; 6:30. [PMID: 27014640 PMCID: PMC4791395 DOI: 10.3389/fcimb.2016.00030] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/29/2016] [Indexed: 01/18/2023] Open
Abstract
Brucella is a facultative intracellular pathogen that causes zoonotic infection known as brucellosis which results in abortion and infertility in natural host. Humans, especially in low income countries, can acquire infection by direct contact with infected animal or by consumption of animal products and show high morbidity, severe economic losses and public health problems. However for survival, host cells develop complex immune mechanisms to defeat and battle against attacking pathogens and maintain a balance between host resistance and Brucella virulence. On the other hand as a successful intracellular pathogen, Brucella has evolved multiple strategies to evade immune response mechanisms to establish persistent infection and replication within host. In this review, we mainly summarize the "Stealth" strategies employed by Brucella to modulate innate and the adaptive immune systems, autophagy, apoptosis and possible role of small noncoding RNA in the establishment of chronic infection. The purpose of this review is to give an overview for recent understanding how this pathogen evades immune response mechanisms of host, which will facilitate to understanding the pathogenesis of brucellosis and the development of novel, more effective therapeutic approaches to treat brucellosis.
Collapse
Affiliation(s)
- Waqas Ahmed
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| | - Ke Zheng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| | - Zheng-Fei Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| |
Collapse
|
14
|
Abstract
Brucellosis, caused by bacteria of the genus Brucella, is an important zoonotic infection that causes reproductive disease in domestic animals and chronic debilitating disease in humans. An intriguing aspect of Brucella infection is the ability of these bacteria to evade the host immune response, leading to pathogen persistence. Conversely, in the reproductive tract of infected animals, this stealthy pathogen is able to cause an acute severe inflammatory response. In this review, we discuss the different mechanisms used by Brucella to cause disease, with emphasis on its virulence factors and the dichotomy between chronic persistence and reproductive disease.
Collapse
Affiliation(s)
| | - Renee M Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616; ,
| |
Collapse
|
15
|
Ke Y, Wang Y, Li W, Chen Z. Type IV secretion system of Brucella spp. and its effectors. Front Cell Infect Microbiol 2015; 5:72. [PMID: 26528442 PMCID: PMC4602199 DOI: 10.3389/fcimb.2015.00072] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 09/28/2015] [Indexed: 11/13/2022] Open
Abstract
Brucella spp. are intracellular bacterial pathogens that cause infection in domestic and wild animals. They are often used as model organisms to study intracellular bacterial infections. Brucella VirB T4SS is a key virulence factor that plays important roles in mediating intracellular survival and manipulating host immune response to infection. In this review, we discuss the roles of Brucella VirB T4SS and 15 effectors that are proposed to be crucial for Brucella pathogenesis. VirB T4SS regulates the inflammation response and manipulates vesicle trafficking inside host cells. VirB T4SS also plays crucial roles in the inhibition of the host immune response and intracellular survival during infection. Here, we list the key molecular events in the intracellular life cycle of Brucella that are potentially targeted by the VirB T4SS effectors. Elucidating the functions of these effectors will help clarify the molecular role of T4SS during infection. Furthermore, studying the effectors secreted by Brucella spp. might provide insights into the mechanisms used by the bacteria to hijack the host signaling pathways and aid in the development of better vaccines and therapies against brucellosis.
Collapse
Affiliation(s)
- Yuehua Ke
- Institute of Disease Control and Prevention, AMMS Beijing, China
| | - Yufei Wang
- Department of Laboratory Medicine, General Hospital of Chinese People's Armed Police Forces Beijing, China
| | - Wengfeng Li
- Department of Orthopedics, The First Affiliated Hospital of General Hospital of People's Liberation Army Beijing, China
| | - Zeliang Chen
- Institute of Disease Control and Prevention, AMMS Beijing, China
| |
Collapse
|
16
|
Silva TMA, Mol JPS, Winter MG, Atluri V, Xavier MN, Pires SF, Paixão TA, Andrade HM, Santos RL, Tsolis RM. The predicted ABC transporter AbcEDCBA is required for type IV secretion system expression and lysosomal evasion by Brucella ovis. PLoS One 2014; 9:e114532. [PMID: 25474545 PMCID: PMC4256435 DOI: 10.1371/journal.pone.0114532] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 11/10/2014] [Indexed: 12/23/2022] Open
Abstract
Brucella ovis is a major cause of reproductive failure in rams and it is one of the few well-described Brucella species that is not zoonotic. Previous work showed that a B. ovis mutant lacking a species-specific ABC transporter (ΔabcBA) was attenuated in mice and was unable to survive in macrophages. The aim of this study was to evaluate the role of this ABC transporter during intracellular survival of B. ovis. In HeLa cells, B. ovis WT was able to survive and replicate at later time point (48 hpi), whereas an ΔabcBA mutant was attenuated at 24 hpi. The reduced survival of the ΔabcBA mutant was associated with a decreased ability to exclude the lysosomal marker LAMP1 from its vacuolar membrane, suggesting a failure to establish a replicative niche. The ΔabcBA mutant showed a reduced abundance of the Type IV secretion system (T4SS) proteins VirB8 and VirB11 in both rich and acid media, when compared to WT B. ovis. However, mRNA levels of virB1, virB8, hutC, and vjbR were similar in both strains. These results support the notion that the ABC transporter encoded by abcEDCBA or its transported substrate acts at a post-transcriptional level to promote the optimal expression of the B. ovis T4SS within infected host cells.
Collapse
Affiliation(s)
- Teane M. A. Silva
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana P. S. Mol
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria G. Winter
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, California, United States of America
| | - Vidya Atluri
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, California, United States of America
| | - Mariana N. Xavier
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, California, United States of America
| | - Simone F. Pires
- Departamento de Parasitologia, Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Tatiane A. Paixão
- Departamento de Patologia Geral, Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Hélida M. Andrade
- Departamento de Parasitologia, Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Renato L. Santos
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail: (RLS); (RMT)
| | - Renee M. Tsolis
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, California, United States of America
- * E-mail: (RLS); (RMT)
| |
Collapse
|
17
|
Xavier MN, Winter MG, Spees AM, den Hartigh AB, Nguyen K, Roux CM, Silva TMA, Atluri VL, Kerrinnes T, Keestra AM, Monack DM, Luciw PA, Eigenheer RA, Bäumler AJ, Santos RL, Tsolis RM. PPARγ-mediated increase in glucose availability sustains chronic Brucella abortus infection in alternatively activated macrophages. Cell Host Microbe 2013; 14:159-70. [PMID: 23954155 PMCID: PMC3777723 DOI: 10.1016/j.chom.2013.07.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 05/17/2013] [Accepted: 06/20/2013] [Indexed: 01/16/2023]
Abstract
Eradication of persistent intracellular bacterial pathogens with antibiotic therapy is often slow or incomplete. However, strategies to augment antibiotics are hampered by our poor understanding of the nutritional environment that sustains chronic infection. Here we show that the intracellular pathogen Brucella abortus survives and replicates preferentially in alternatively activated macrophages (AAMs), which are more abundant during chronic infection. A metabolic shift induced by peroxisome proliferator-activated receptor γ (PPARγ), which increases intracellular glucose availability, is identified as a causal mechanism promoting enhanced bacterial survival in AAMs. Glucose uptake was crucial for increased replication of B. abortus in AAMs, and for chronic infection, as inactivation of the bacterial glucose transporter gluP reduced both intracellular survival in AAMs and persistence in mice. Thus, a shift in intracellular nutrient availability induced by PPARγ promotes chronic persistence of B. abortus within AAMs, and targeting this pathway may aid in eradicating chronic infection.
Collapse
Affiliation(s)
- Mariana N. Xavier
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Maria G. Winter
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Alanna M. Spees
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Andreas B. den Hartigh
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Kim Nguyen
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Christelle M. Roux
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Teane M. A. Silva
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Vidya L. Atluri
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Tobias Kerrinnes
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - A. Marijke Keestra
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Denise M. Monack
- Department of Microbiology & Immunology, School of Medicine, Stanford University, Palo Alto, CA, 94305, USA
| | - Paul A. Luciw
- Center for Comparative Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Richard A. Eigenheer
- Proteomics Core Facility, University of California at Davis Genome Center, Davis, CA, 95616, USA
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Renato L. Santos
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Renée M. Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| |
Collapse
|
18
|
Abstract
The complex immune system of mammals is the result of evolutionary forces that include battles against pathogens, as sensing and defeating intruders is a prerequisite to host survival. On the other hand, microorganisms have evolved multiple mechanisms to evade both arms of immunity: the innate and the adaptive immune systems. The successful pathogenic intracellular bacterium Brucella is not an exception to the rule: Brucella displays mechanisms that allow evasion of immune surveillance in order to establish persistent infections in mammals. In this review, we highlight some key mechanisms that pathogenic Brucella use to evade the adaptive immune system.
Collapse
Affiliation(s)
- Anna Martirosyan
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
| | | |
Collapse
|
19
|
Xavier MN, Winter MG, Spees AM, Nguyen K, Atluri VL, Silva TMA, Bäumler AJ, Müller W, Santos RL, Tsolis RM. CD4+ T cell-derived IL-10 promotes Brucella abortus persistence via modulation of macrophage function. PLoS Pathog 2013; 9:e1003454. [PMID: 23818855 PMCID: PMC3688575 DOI: 10.1371/journal.ppat.1003454] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 05/08/2013] [Indexed: 12/21/2022] Open
Abstract
Evasion of host immune responses is a prerequisite for chronic bacterial diseases; however, the underlying mechanisms are not fully understood. Here, we show that the persistent intracellular pathogen Brucella abortus prevents immune activation of macrophages by inducing CD4(+)CD25(+) T cells to produce the anti-inflammatory cytokine interleukin-10 (IL-10) early during infection. IL-10 receptor (IL-10R) blockage in macrophages resulted in significantly higher NF-kB activation as well as decreased bacterial intracellular survival associated with an inability of B. abortus to escape the late endosome compartment in vitro. Moreover, either a lack of IL-10 production by T cells or a lack of macrophage responsiveness to this cytokine resulted in an increased ability of mice to control B. abortus infection, while inducing elevated production of pro-inflammatory cytokines, which led to severe pathology in liver and spleen of infected mice. Collectively, our results suggest that early IL-10 production by CD25(+)CD4(+) T cells modulates macrophage function and contributes to an initial balance between pro-inflammatory and anti-inflammatory cytokines that is beneficial to the pathogen, thereby promoting enhanced bacterial survival and persistent infection.
Collapse
Affiliation(s)
- Mariana N. Xavier
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
- Departamento de Clínica e Cirurgia Veterinárias, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria G. Winter
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Alanna M. Spees
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Kim Nguyen
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Vidya L. Atluri
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Teane M. A. Silva
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
- Departamento de Clínica e Cirurgia Veterinárias, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Werner Müller
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Renato L. Santos
- Departamento de Clínica e Cirurgia Veterinárias, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Renée M. Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| |
Collapse
|
20
|
Abstract
Host cytokine responses to Brucella abortus infection are elicited predominantly by the deployment of a type IV secretion system (T4SS). However, the mechanism by which the T4SS elicits inflammation remains unknown. Here we show that translocation of the T4SS substrate VceC into host cells induces proinflammatory responses. Ectopically expressed VceC interacted with the endoplasmic reticulum (ER) chaperone BiP/Grp78 and localized to the ER of HeLa cells. ER localization of VceC required a transmembrane domain in its N terminus. Notably, the expression of VceC resulted in reorganization of ER structures. In macrophages, VceC was required for B. abortus-induced inflammation by induction of the unfolded protein response by a process requiring inositol-requiring transmembrane kinase/endonuclease 1. Altogether, these findings suggest that translocation of the T4SS effector VceC induces ER stress, which results in the induction of proinflammatory host cell responses during B. abortus infection. IMPORTANCE Brucella species are pathogens that require a type IV secretion system (T4SS) to survive in host cells and to maintain chronic infection. By as-yet-unknown pathways, the T4SS also elicits inflammatory responses in infected cells. Here we show that inflammation caused by the T4SS results in part from the sensing of a T4SS substrate, VceC, that localizes to the endoplasmic reticulum (ER), an intracellular site of Brucella replication. Possibly via binding of the ER chaperone BiP, VceC causes ER stress with concomitant expression of proinflammatory cytokines. Thus, induction of the unfolded protein response may represent a novel pathway by which host cells can detect pathogens deploying a T4SS.
Collapse
|
21
|
Terwagne M, Ferooz J, Rolán HG, Sun YH, Atluri V, Xavier MN, Franchi L, Núñez G, Legrand T, Flavell RA, De Bolle X, Letesson JJ, Tsolis RM. Innate immune recognition of flagellin limits systemic persistence of Brucella. Cell Microbiol 2013; 15:942-960. [PMID: 23227931 DOI: 10.1111/cmi.12088] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 11/01/2012] [Accepted: 11/23/2012] [Indexed: 12/22/2022]
Abstract
Brucella are facultative intracellular bacteria that cause chronic infections by limiting innate immune recognition. It is currently unknown whether Brucella FliC flagellin, the monomeric subunit of flagellar filament, is sensed by the host during infection. Here, we used two mutants of Brucella melitensis, either lacking or overexpressing flagellin, to show that FliC hinders bacterial replication in vivo. The use of cells and mice genetically deficient for different components of inflammasomes suggested that FliC was a target of the cytosolic innate immune receptor NLRC4 in vivo but not in macrophages in vitro where the response to FliC was nevertheless dependent on the cytosolic adaptor ASC, therefore suggesting a new pathway of cytosolic flagellin sensing. However, our work also suggested that the lack of TLR5 activity of Brucella flagellin and the regulation of its synthesis and/or delivery into host cells are both part of the stealthy strategy of Brucella towards the innate immune system. Nevertheless, as a flagellin-deficient mutant of B. melitensis wasfound to cause histologically demonstrable injuries in the spleen of infected mice, we suggested that recognition of FliC plays a role in the immunological stand-off between Brucella and its host, which is characterized by a persistent infection with limited inflammatory pathology.
Collapse
Affiliation(s)
| | | | - Hortensia G Rolán
- Department of Medical Microbiology & Immunology, University of California, Davis, CA, USA
| | - Yao-Hui Sun
- Department of Medical Microbiology & Immunology, University of California, Davis, CA, USA
| | - Vidya Atluri
- Department of Medical Microbiology & Immunology, University of California, Davis, CA, USA
| | - Mariana N Xavier
- Department of Medical Microbiology & Immunology, University of California, Davis, CA, USA
| | - Luigi Franchi
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Gabriel Núñez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | - Renée M Tsolis
- Department of Medical Microbiology & Immunology, University of California, Davis, CA, USA
| |
Collapse
|
22
|
Crucial role of gamma interferon-producing CD4+ Th1 cells but dispensable function of CD8+ T cell, B cell, Th2, and Th17 responses in the control of Brucella melitensis infection in mice. Infect Immun 2012; 80:4271-80. [PMID: 23006848 DOI: 10.1128/iai.00761-12] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Brucella spp. are facultative intracellular bacterial pathogens responsible for brucellosis, a worldwide zoonosis that causes abortion in domestic animals and chronic febrile disease associated with serious complications in humans. There is currently no approved vaccine against human brucellosis, and antibiotic therapy is long and costly. Development of a safe protective vaccine requires a better understanding of the roles played by components of adaptive immunity in the control of Brucella infection. The importance of lymphocyte subsets in the control of Brucella growth has been investigated separately by various research groups and remains unclear or controversial. Here, we used a large panel of genetically deficient mice to compare the importance of B cells, transporter associated with antigen processing (TAP-1), and major histocompatibility complex class II-dependent pathways of antigen presentation as well as T helper 1 (Th1), Th2, and Th17-mediated responses on the immune control of Brucella melitensis 16 M infection. We clearly confirmed the key function played by gamma interferon (IFN-γ)-producing Th1 CD4(+) T cells in the control of B. melitensis infection, whereas IFN-γ-producing CD8(+) T cells or B cell-mediated humoral immunity plays only a modest role in the clearance of bacteria during primary infection. In the presence of a Th1 response, Th2 or Th17 responses do not really develop or play a positive or negative role during the course of B. melitensis infection. On the whole, these results could improve our ability to develop protective vaccines or therapeutic treatments against brucellosis.
Collapse
|
23
|
Identification of genes contributing to the intracellular replication of Brucella abortus within HeLa and RAW 264.7 cells. Vet Microbiol 2012; 158:322-8. [DOI: 10.1016/j.vetmic.2012.02.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 02/07/2012] [Accepted: 02/13/2012] [Indexed: 11/18/2022]
|
24
|
Grilló MJ, Blasco JM, Gorvel JP, Moriyón I, Moreno E. What have we learned from brucellosis in the mouse model? Vet Res 2012; 43:29. [PMID: 22500859 PMCID: PMC3410789 DOI: 10.1186/1297-9716-43-29] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 04/13/2012] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is a zoonosis caused by Brucella species. Brucellosis research in natural hosts is often precluded by practical, economical and ethical reasons and mice are widely used. However, mice are not natural Brucella hosts and the course of murine brucellosis depends on bacterial strain virulence, dose and inoculation route as well as breed, genetic background, age, sex and physiological statu of mice. Therefore, meaningful experiments require a definition of these variables. Brucella spleen replication profiles are highly reproducible and course in four phases: i), onset or spleen colonization (first 48 h); ii), acute phase, from the third day to the time when bacteria reach maximal numbers; iii), chronic steady phase, where bacterial numbers plateaus; and iv), chronic declining phase, during which brucellae are eliminated. This pattern displays clear physiopathological signs and is sensitive to small virulence variations, making possible to assess attenuation when fully virulent bacteria are used as controls. Similarly, immunity studies using mice with known defects are possible. Mutations affecting INF-γ, TLR9, Myd88, Tγδ and TNF-β favor Brucella replication; whereas IL-1β, IL-18, TLR4, TLR5, TLR2, NOD1, NOD2, GM-CSF, IL/17r, Rip2, TRIF, NK or Nramp1 deficiencies have no noticeable effects. Splenomegaly development is also useful: it correlates with IFN-γ and IL-12 levels and with Brucella strain virulence. The genetic background is also important: Brucella-resistant mice (C57BL) yield lower splenic bacterial replication and less splenomegaly than susceptible breeds. When inoculum is increased, a saturating dose above which bacterial numbers per organ do not augment, is reached. Unlike many gram-negative bacteria, lethal doses are large (≥ 108 bacteria/mouse) and normally higher than the saturating dose. Persistence is a useful virulence/attenuation index and is used in vaccine (Residual Virulence) quality control. Vaccine candidates are also often tested in mice by determining splenic Brucella numbers after challenging with appropriate virulent brucellae doses at precise post-vaccination times. Since most live or killed Brucella vaccines provide some protection in mice, controls immunized with reference vaccines (S19 or Rev1) are critical. Finally, mice have been successfully used to evaluate brucellosis therapies. It is concluded that, when used properly, the mouse is a valuable brucellosis model.
Collapse
Affiliation(s)
- María-Jesús Grilló
- Instituto de Agrobiotecnología, CSIC-UPNA-Gobierno de Navarra, Pamplona, Spain
| | - José María Blasco
- Centro de Investigación y Tecnología Agroalimentaria (CITA) de Aragón, Zaragoza, Spain
| | - Jean Pierre Gorvel
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Faculté de Sciences de Luminy, Luminy, France
- Institut National de la Santé et de la Recherche Médicale U631, Marseille, France
- Centre National de la Recherche Scientifique UMR6102, Marseille, France
| | - Ignacio Moriyón
- Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
- Instituto de Salud Tropical, Universidad de Navarra, Pamplona, Spain
| | - Edgardo Moreno
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
25
|
Abstract
Brucellosis is a global disease of domestic and wild mammals that is caused by intracellular bacteria of the genus Brucella. Although humans are not a natural reservoir for Brucella, infection in the human population is common in many countries, and brucellosis is one of the most common zoonotic infections. Brucella species have evolved to avoid the host's immune system and infection is usually characterized by long-term persistence of the bacteria. One important Brucella virulence factor for intracellular survival and persistence in the host is the type IV secretion system. This review will discuss the Brucella type IV secretion system in detail, including current knowledge of architecture and regulation, as well as the newly identified effector substrates that this system transports into host cells.
Collapse
Affiliation(s)
- Maarten F de Jong
- Department of Medical Microbiology & Immunology, University of California, Davis, CA, USA
| | | |
Collapse
|
26
|
Copin R, Vitry MA, Hanot Mambres D, Machelart A, De Trez C, Vanderwinden JM, Magez S, Akira S, Ryffel B, Carlier Y, Letesson JJ, Muraille E. In situ microscopy analysis reveals local innate immune response developed around Brucella infected cells in resistant and susceptible mice. PLoS Pathog 2012; 8:e1002575. [PMID: 22479178 PMCID: PMC3315488 DOI: 10.1371/journal.ppat.1002575] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 01/26/2012] [Indexed: 12/31/2022] Open
Abstract
Brucella are facultative intracellular bacteria that chronically infect humans and animals causing brucellosis. Brucella are able to invade and replicate in a broad range of cell lines in vitro, however the cells supporting bacterial growth in vivo are largely unknown. In order to identify these, we used a Brucella melitensis strain stably expressing mCherry fluorescent protein to determine the phenotype of infected cells in spleen and liver, two major sites of B. melitensis growth in mice. In both tissues, the majority of primary infected cells expressed the F4/80 myeloid marker. The peak of infection correlated with granuloma development. These structures were mainly composed of CD11b⁺ F4/80⁺ MHC-II⁺ cells expressing iNOS/NOS2 enzyme. A fraction of these cells also expressed CD11c marker and appeared similar to inflammatory dendritic cells (DCs). Analysis of genetically deficient mice revealed that differentiation of iNOS⁺ inflammatory DC, granuloma formation and control of bacterial growth were deeply affected by the absence of MyD88, IL-12p35 and IFN-γ molecules. During chronic phase of infection in susceptible mice, we identified a particular subset of DC expressing both CD11c and CD205, serving as a reservoir for the bacteria. Taken together, our results describe the cellular nature of immune effectors involved during Brucella infection and reveal a previously unappreciated role for DC subsets, both as effectors and reservoir cells, in the pathogenesis of brucellosis.
Collapse
Affiliation(s)
- Richard Copin
- Unité de Recherche en Biologie Moléculaire, Laboratoire d'Immunologie et de Microbiologie, Faculté Universitaire Notre Dame de la Paix, Namur, Belgium
| | - Marie-Alice Vitry
- Unité de Recherche en Biologie Moléculaire, Laboratoire d'Immunologie et de Microbiologie, Faculté Universitaire Notre Dame de la Paix, Namur, Belgium
| | - Delphine Hanot Mambres
- Unité de Recherche en Biologie Moléculaire, Laboratoire d'Immunologie et de Microbiologie, Faculté Universitaire Notre Dame de la Paix, Namur, Belgium
| | - Arnaud Machelart
- Unité de Recherche en Biologie Moléculaire, Laboratoire d'Immunologie et de Microbiologie, Faculté Universitaire Notre Dame de la Paix, Namur, Belgium
| | - Carl De Trez
- Department of Molecular and Cellular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jean-Marie Vanderwinden
- Laboratoire de Neurophysiologie, Faculté de Médecine, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Stefan Magez
- Department of Molecular and Cellular Interactions, Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium
| | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University Yamadaoka, Suita City, Osaka, Japan
| | - Bernhard Ryffel
- University of Orleans, Transgenose Institute, Laboratoire d'Immunologie et d'Embryologie Moléculaires, CNRS, UMR 6218, Orleans, France
| | - Yves Carlier
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Jean-Jacques Letesson
- Unité de Recherche en Biologie Moléculaire, Laboratoire d'Immunologie et de Microbiologie, Faculté Universitaire Notre Dame de la Paix, Namur, Belgium
- * E-mail: (JJL); (EM)
| | - Eric Muraille
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Bruxelles, Belgium
- * E-mail: (JJL); (EM)
| |
Collapse
|
27
|
Sá JC, Silva TMA, Costa EA, Silva APC, Tsolis RM, Paixão TA, Carvalho Neta AV, Santos RL. The virB-encoded type IV secretion system is critical for establishment of infection and persistence of Brucella ovis infection in mice. Vet Microbiol 2012; 159:130-40. [PMID: 22483850 DOI: 10.1016/j.vetmic.2012.03.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 03/08/2012] [Accepted: 03/10/2012] [Indexed: 12/13/2022]
Abstract
Brucella spp. are gram-negative intracellular bacterial pathogens that cause chronic infections. Brucella virulence factors include a type IV secretion system (T4SS) and its lipopolysaccharide (LPS), which are essential for persistence. However, the role of the virB-encoded T4SS has not been investigated in naturally rough Brucella species such as Brucella ovis. In this study, male 6-week old BALBc mice were infected with B. ovis, Brucella abortus, and their respective ΔvirB2 mutant strains. During early infection, B. ovis and B. abortus wild type strains were similarly recovered from spleen. Interestingly, in contrast to ΔvirB2 B. abortus that was recovered at similar levels when compared to the wild type strain, the ΔvirB2 B. ovis was markedly attenuated as early as 24h post infection (hpi). The ΔvirB2 B. ovis was unable to survive and multiply in murine peritoneal macrophages and extracellularly within the peritoneal cavity at 12 and 24 hpi with lower splenic colonization than the parental strain at 6, 12 and 24 hpi. In contrast, wild type B. abortus and ΔvirB2 B. abortus had a similar kinetics of infection in this model. As expected, the T4SS was essential for intracellular replication of smooth and rough strains in RAW macrophages at 48 hpi. These results suggest that T4SS is important for survival of B. ovis in murine model, and that a T4SS deficient B. ovis strain is cleared at earlier stages of infection when compared to a similar B. abortus mutant.
Collapse
Affiliation(s)
- Joicy C Sá
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
28
|
von Bargen K, Gorvel JP, Salcedo SP. Internal affairs: investigating the Brucella intracellular lifestyle. FEMS Microbiol Rev 2012; 36:533-62. [PMID: 22373010 DOI: 10.1111/j.1574-6976.2012.00334.x] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 01/10/2012] [Accepted: 02/16/2012] [Indexed: 01/18/2023] Open
Abstract
Bacteria of the genus Brucella are Gram-negative pathogens of several animal species that cause a zoonotic disease in humans known as brucellosis or Malta fever. Within their hosts, brucellae reside within different cell types where they establish a replicative niche and remain protected from the immune response. The aim of this article is to discuss recent advances in the field in the specific context of the Brucella intracellular 'lifestyle'. We initially discuss the different host cell targets and their relevance during infection. As it represents the key to intracellular replication, the focus is then set on the maturation of the Brucella phagosome, with particular emphasis on the Brucella factors that are directly implicated in intracellular trafficking and modulation of host cell signalling pathways. Recent data on the role of the type IV secretion system are discussed, novel effector molecules identified and how some of them impact on trafficking events. Current knowledge on Brucella gene regulation and control of host cell death are summarized, as they directly affect intracellular persistence. Understanding how Brucella molecules interplay with their host cell targets to modulate cellular functions and establish the intracellular niche will help unravel how this pathogen causes disease.
Collapse
Affiliation(s)
- Kristine von Bargen
- Faculté de Sciences de Luminy, Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, UM 2, Marseille Cedex, France
| | | | | |
Collapse
|
29
|
Atluri VL, Xavier MN, de Jong MF, den Hartigh AB, Tsolis RM. Interactions of the human pathogenic Brucella species with their hosts. Annu Rev Microbiol 2012; 65:523-41. [PMID: 21939378 DOI: 10.1146/annurev-micro-090110-102905] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Brucellosis is a zoonotic infection caused primarily by the bacterial pathogens Brucella melitensis and B. abortus. It is acquired by consumption of unpasteurized dairy products or by contact with infected animals. Globally, it is one of the most widespread zoonoses, with 500,000 new cases reported each year. In endemic areas, Brucella infections represent a serious public health problem that results in significant morbidity and economic losses. An important feature of the disease is persistent bacterial colonization of the reticuloendothelial system. In this review we discuss recent insights into mechanisms of intracellular survival and immune evasion that contribute to systemic persistence by the pathogenic Brucella species.
Collapse
Affiliation(s)
- Vidya L Atluri
- Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616, USA.
| | | | | | | | | |
Collapse
|
30
|
Martirosyan A, Moreno E, Gorvel JP. An evolutionary strategy for a stealthy intracellular Brucella pathogen. Immunol Rev 2011; 240:211-34. [PMID: 21349096 DOI: 10.1111/j.1600-065x.2010.00982.x] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Brucella is an intracellular bacterial pathogen that causes abortion and infertility in mammals and leads to a debilitating febrile illness that can progress into a long lasting disease with severe complications in humans. Its virulence depends on survival and replication properties in host cells. In this review, we describe the stealthy strategy used by Brucella to escape recognition of the innate immunity and the means by which this bacterium evades intracellular destruction. We also discuss the development of adaptive immunity and its modulation during brucellosis that in course leads to chronic infections. Brucella has developed specific strategies to influence antigen presentation mediated by cells. There is increasing evidence that Brucella also modulates signaling events during host adaptive immune responses.
Collapse
Affiliation(s)
- Anna Martirosyan
- Faculté de Sciences de Luminy, Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Marseille, France
| | | | | |
Collapse
|
31
|
Laboratory animal models for brucellosis research. J Biomed Biotechnol 2011; 2011:518323. [PMID: 21403904 PMCID: PMC3043301 DOI: 10.1155/2011/518323] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 11/25/2010] [Accepted: 01/11/2011] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is a chronic infectious disease caused by Brucella spp., a Gram-negative facultative intracellular pathogen that affects humans and animals, leading to significant impact on public health and animal industry. Human brucellosis is considered the most prevalent bacterial zoonosis in the world and is characterized by fever, weight loss, depression, hepato/splenomegaly, osteoarticular, and genital infections. Relevant aspects of Brucella pathogenesis have been intensively investigated in culture cells and animal models. The mouse is the animal model more commonly used to study chronic infection caused by Brucella. This model is most frequently used to investigate specific pathogenic factors of Brucella spp., to characterize the host immune response, and to evaluate therapeutics and vaccines. Other animal species have been used as models for brucellosis including rats, guinea pigs, and monkeys. This paper discusses the murine and other laboratory animal models for human and animal brucellosis.
Collapse
|
32
|
The Vi capsular polysaccharide prevents complement receptor 3-mediated clearance of Salmonella enterica serotype Typhi. Infect Immun 2010; 79:830-7. [PMID: 21098104 DOI: 10.1128/iai.00961-10] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Capsular polysaccharides are important virulence factors of invasive bacterial pathogens. Here we studied the role of the virulence (Vi) capsular polysaccharide of Salmonella enterica serotype Typhi (S. Typhi) in preventing innate immune recognition by complement. Comparison of capsulated S. Typhi with a noncapsulated mutant (ΔtviBCDE vexABCDE mutant) revealed that the Vi capsule interfered with complement component 3 (C3) deposition. Decreased complement fixation resulted in reduced bacterial binding to complement receptor 3 (CR3) on the surface of murine macrophages in vitro and decreased CR3-dependent clearance of Vi capsulated S. Typhi from the livers and spleens of mice. Opsonization of bacteria with immune serum prior to intraperitoneal infection increased clearance of capsulated S. Typhi from the liver. Our data suggest that the Vi capsule prevents CR3-dependent clearance, which can be overcome in part by a specific antibody response.
Collapse
|
33
|
de Jong MF, Rolán HG, Tsolis RM. Innate immune encounters of the (Type) 4th kind: Brucella. Cell Microbiol 2010; 12:1195-202. [PMID: 20670294 DOI: 10.1111/j.1462-5822.2010.01498.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In humans, pathogenic Brucella species cause a febrile illness known as brucellosis. A key pathogenic trait of this group of organisms is their ability to survive in immune cells and persist in tissues of the reticuloendothelial system, a process that requires the function of a Type IV secretion system. In contrast to other well-studied Gram-negative bacteria, Brucella spp. do not cause inflammation at the site of invasion, but have a latency period of 2-4 weeks before the onset of symptoms. This review discusses several mechanisms that allow Brucella spp. both to evade detection by pattern recognition receptors of the innate immune system and suppress their signalling. In contrast to these stealth features, the VirB Type IV secretion system, which mediates survival within phagocytic cells, stimulates innate immune responses in vivo. The responses stimulated by this virulence factor are sufficient to check bacterial growth, but not to elicit sterilizing immunity. The result is a stand-off between host and pathogen that results in persistent infection.
Collapse
Affiliation(s)
- Maarten F de Jong
- Department of Medical Microbiology & Immunology, University of California at Davis, Davis, CA, USA
| | | | | |
Collapse
|
34
|
Neta AVC, Mol JP, Xavier MN, Paixão TA, Lage AP, Santos RL. Pathogenesis of bovine brucellosis. Vet J 2010; 184:146-55. [DOI: 10.1016/j.tvjl.2009.04.010] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 02/05/2009] [Accepted: 04/13/2009] [Indexed: 12/14/2022]
|
35
|
Gao N, Jennings P, Guo Y, Yuan D. Regulatory role of natural killer (NK) cells on antibody responses to Brucella abortus. Innate Immun 2010; 17:152-63. [PMID: 20418255 DOI: 10.1177/1753425910367526] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Our previous studies have indicated an important regulatory role for natural killer (NK) cells, a major constituent of the innate immune system in modulating antigen-specific responses. Herein, we have investigated the possible participation of these cells in regulating the polyclonal response as well. For these studies we have utilized heat-killed Brucella abortus (HKBA). Brucella abortus is a facultative intracellular bacterium that is pathogenic for both humans and animals. An outstanding feature of the infectious process is the rapid production of polyclonal antibodies, particularly of the IgG2c subclass, that bypasses the requirement for clonally specific antigen recognition. We report here that NK-cell depletion profoundly reduced the production of these polyclonal antibodies suggesting that activation of B cells by HKBA requires help from NK cells. This help may not be solely derived from NK-cell amplification of the cytokine circuit initiated by HKBA but may involve direct NK-B-cell interactions as suggested by results of in vitro analyses of NK induction of γ2a mRNA by B cells. These findings have therapeutic implications in that the induction of polyclonal Ig production may be more important for altering the chronic phase rather than the acute stage of infection by B. abortus.
Collapse
Affiliation(s)
- Ning Gao
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, TX 75390, USA
| | | | | | | |
Collapse
|
36
|
Tükel C, Wilson RP, Nishimori JH, Pezeshki M, Chromy BA, Bäumler AJ. Responses to amyloids of microbial and host origin are mediated through toll-like receptor 2. Cell Host Microbe 2009; 6:45-53. [PMID: 19616765 DOI: 10.1016/j.chom.2009.05.020] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Revised: 01/30/2009] [Accepted: 05/01/2009] [Indexed: 10/20/2022]
Abstract
Curli fibrils are proteinaceous bacterial structures formed by amyloid fibrils composed of the major curli subunit CsgA. Like beta-amyloid 1-42, which is associated with brain inflammation and Alzheimer's disease, curli fibrils have been implicated in the induction of host inflammatory responses. However, the underlying mechanisms of amyloid-induced inflammation are not fully understood. In a mouse sepsis model, we show that curli fibrils contributed to Nos2 expression, a hallmark of inflammation, by stimulating Toll-like receptor (TLR) 2. The TLR2 agonist activity was reduced by an amyloidogenicity-lowering amino acid substitution (N122A) in CsgA. Amyloid-forming synthetic peptides corresponding to beta-amyloid 1-42 or CsgA 111-151 stimulated Nos2 production in macrophages and microglia cells through a TLR2-dependent mechanism. This activity was abrogated when an N122A substitution was introduced into the synthetic CsgA peptide. The induction of TLR2-mediated responses by bacterial and eukaryotic amyloids may explain the inflammation associated with amyloids and the resulting pathologies.
Collapse
Affiliation(s)
- Cagla Tükel
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA 95616-8645, USA
| | | | | | | | | | | |
Collapse
|
37
|
Establishment of systemic Brucella melitensis infection through the digestive tract requires urease, the type IV secretion system, and lipopolysaccharide O antigen. Infect Immun 2009; 77:4197-208. [PMID: 19651862 DOI: 10.1128/iai.00417-09] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Human brucellosis is caused mainly by Brucella melitensis, which is often acquired by ingesting contaminated goat or sheep milk and cheese. Bacterial factors required for food-borne infection of humans by B. melitensis are poorly understood. In this study, a mouse model of oral infection was characterized to assess the roles of urease, the VirB type IV secretion system, and lipopolysaccharide for establishing infection through the digestive tract. B. melitensis strain 16M was consistently recovered from the mesenteric lymph node (MLN), spleen, and liver beginning at 3 or 7 day postinfection (dpi). In the gut, persistence of the inoculum was observed up to 21 dpi. No inflammatory lesions were observed in the ileum or colon during infection. Mutant strains lacking the ureABC genes of the ure1 operon, virB2, or pmm encoding phosphomannomutase were constructed and compared to the wild-type strain for infectivity through the digestive tract. Mutants lacking the virB2 and pmm genes were attenuated in the spleen (P < 0.05) and MLN (P < 0.001), respectively. The wild-type and mutant strains had similar levels of resistance to low pH and 5 or 10% bile, suggesting that the reduced colonization of mutants was not the result of reduced resistance to acid pH or bile salts. In an in vitro lymphoepithelial cell (M-cell) model, B. melitensis transited rapidly through polarized enterocyte monolayers containing M-like cells; however, transit through monolayers containing only enterocytes was reduced or absent. These results indicate that B. melitensis is able to spread systemically from the digestive tract after infection, most likely through M cells of the mucosa-associated lymphoid tissue.
Collapse
|
38
|
Natural antibody contributes to host defense against an attenuated Brucella abortus virB mutant. Infect Immun 2009; 77:3004-13. [PMID: 19364836 DOI: 10.1128/iai.01114-08] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Brucella abortus is an intracellular pathogen that persists within phagocytic cells of the reticuloendothelial system. To identify in vivo interactions between B. abortus and the host that lead to persistent infection, we studied the persistence of B. abortus and an isogenic virB mutant deficient in the VirB type IV secretion system (T4SS) in knockout mice. In contrast to control mice, mice lacking B cells (Igh6(-/-)) were permissive for infection with the attenuated virB mutant. To determine the basis for this phenotype, we characterized immune functions of Igh6(-/-) mice in the context of B. abortus infection. Igh6(-/-) mice had greater numbers of extracellular bacteria in the spleen and increased early expression of proinflammatory cytokines during B. abortus infection. Further, a virB mutant, despite its wild-type level of survival, failed to elicit microgranuloma formation in the spleens of Igh6(-/-) mice, suggesting a requirement for the T4SS to elicit this pathological change. Passive transfer of immunoglobulin G from naïve mice restored the ability of Igh6(-/-) mice to control the persistence of the virB mutant by a complement-independent mechanism. Further, adoptive transfer of CD11b(+) cells from C57BL/6 mice to Igh6(-/-) mice restored the ability of the knockout mice to limit the replication of the virB mutant in the spleen, suggesting that the Igh6(-)(/)(-) mutation affects phagocyte function and that phagocyte function can be restored by natural antibody.
Collapse
|
39
|
Contribution of flagellin pattern recognition to intestinal inflammation during Salmonella enterica serotype typhimurium infection. Infect Immun 2009; 77:1904-16. [PMID: 19237529 DOI: 10.1128/iai.01341-08] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Salmonella enterica serotype Typhimurium causes acute inflammatory diarrhea in humans. Flagella contribute to intestinal inflammation, but the mechanism remains unclear since most mutations abrogating pattern recognition of flagellin also prevent motility and reduce bacterial invasion. To determine the contribution of flagellin pattern recognition to the generation of innate immune responses, we compared in two animal models a nonmotile, but flagellin-expressing and -secreting serotype Typhimurium strain (flgK mutant) to a nonmotile, non-flagellin-expressing strain (flgK fliC fljB mutant). In vitro, caspase-1 can be activated by cytosolic delivery of flagellin, resulting in release of the interferon gamma inducing factor interleukin-18 (IL-18). Experiments with streptomycin-pretreated caspase-1-deficient mice suggested that induction of gamma interferon expression in the murine cecum early (12 h) after serotype Typhimurium infection was caspase-1 dependent but independent of flagellin pattern recognition. In addition, mRNA levels of the CXC chemokines macrophage inflammatory protein 2 and keratinocyte-derived chemokine were markedly increased early after serotype Typhimurium infection of streptomycin-pretreated wild-type mice regardless of flagellin expression. In contrast, in bovine ligated ileal loops, flagellin pattern recognition contributed to increased mRNA levels of macrophage inflammatory protein 3alpha and more fluid accumulation at 2 h after infection. Collectively, our data suggest that pattern recognition of flagellin contributes to early innate host responses in the bovine ileal mucosa but not in the murine cecal mucosa.
Collapse
|
40
|
Pathological, Immunohistochemical and Bacteriological Study of Tissues and Milk of Cows and Fetuses Experimentally Infected with Brucella abortus. J Comp Pathol 2009; 140:149-57. [DOI: 10.1016/j.jcpa.2008.10.004] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Accepted: 10/05/2008] [Indexed: 11/18/2022]
|
41
|
RNAi screen of endoplasmic reticulum-associated host factors reveals a role for IRE1alpha in supporting Brucella replication. PLoS Pathog 2008; 4:e1000110. [PMID: 18654626 PMCID: PMC2453327 DOI: 10.1371/journal.ppat.1000110] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Accepted: 06/24/2008] [Indexed: 11/20/2022] Open
Abstract
Brucella species are facultative intracellular bacterial pathogens that cause brucellosis, a global zoonosis of profound importance. Although recent studies have demonstrated that Brucella spp. replicate within an intracellular compartment that contains endoplasmic reticulum (ER) resident proteins, the molecular mechanisms by which the pathogen secures this replicative niche remain obscure. Here, we address this issue by exploiting Drosophila S2 cells and RNA interference (RNAi) technology to develop a genetically tractable system that recapitulates critical aspects of mammalian cell infection. After validating this system by demonstrating a shared requirement for phosphoinositide 3-kinase (PI3K) activities in supporting Brucella infection in both host cell systems, we performed an RNAi screen of 240 genes, including 110 ER-associated genes, for molecules that mediate bacterial interactions with the ER. We uncovered 52 evolutionarily conserved host factors that, when depleted, inhibited or increased Brucella infection. Strikingly, 29 of these factors had not been previously suggested to support bacterial infection of host cells. The most intriguing of these was inositol-requiring enzyme 1 (IRE1), a transmembrane kinase that regulates the eukaryotic unfolded protein response (UPR). We employed IRE1α−/− murine embryonic fibroblasts (MEFs) to demonstrate a role for this protein in supporting Brucella infection of mammalian cells, and thereby, validated the utility of the Drosophila S2 cell system for uncovering novel Brucella host factors. Finally, we propose a model in which IRE1α, and other ER-associated genes uncovered in our screen, mediate Brucella replication by promoting autophagosome biogenesis. Brucella spp. are facultative intracellular pathogens that cause brucellosis in a broad range of hosts, including humans. Brucella melitensis, B. abortus, and B. suis are highly infectious and can be readily transmitted in aerosolized form, and a human vaccine against brucellosis is unavailable. Therefore, these pathogens are recognized as potential bioterror agents. Because genetic systems for studying host–Brucella interactions have been unavailable, little is known about the host factors that mediate infection. Here, we demonstrate that a Drosophila S2 cell system and RNA interference can be exploited to study the role that evolutionarily conserved Brucella host proteins play in these processes. We also show that this system provides for the identification and characterization of host factors that mediate Brucella interactions with the host cell endoplasmic reticulum. In fact, we identified 52 host factors that, when depleted, inhibited or increased Brucella infection. Among the identified Brucella host factors, 29 have not been previously shown to support bacterial infection. Finally, we demonstrate that the novel host factor inositol-requiring enzyme 1 (IRE1) and its mammalian ortholog (IRE1α) are required for Brucella infection of Drosophila S2 and mammalian cells, respectively. Therefore, this work contributes to our understanding of host factors mediating Brucella infection.
Collapse
|
42
|
Abstract
Dr David Bruce (1855-1931) first identified the causative agent of brucellosis as a small Gram-negative alpha-Proteobacterium, which was later on called Brucella melitensis in his honor by Meyer and Shaw. Nowadays, four strains exhibit pathogenicity in humans with B. melitensis being the least host specific and also the most infectious for humans. The other strains are Brucella suis and Brucella abortus and more recently human cases being infected with Brucella cetaceae have been reported. Why such a reemerging disease is so difficult to fight, evidence shows that the pathogenic bacterium has developed strategies to hide from immune recognition.
Collapse
Affiliation(s)
- Jean-Pierre Gorvel
- Marseille Aix Marseille Université, Faculté des Sciences de Luminy, F-13288, France.
| |
Collapse
|
43
|
Billard E, Dornand J, Gross A. VirB type IV secretory system does not contribute to Brucella suis' avoidance of human dendritic cell maturation. ACTA ACUST UNITED AC 2008; 53:404-12. [PMID: 18625010 DOI: 10.1111/j.1574-695x.2008.00441.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Dendritic cells (DCs), which are critical components of adaptive immunity, are highly susceptible to infection with the intracellular bacteria Brucella. Infection with living Brucella prevents infected human DCs from engaging in maturation processes, thus impairing their capacity to present antigens to naïve T cells and to secrete IL-12. Recently, we have established that several attenuated mutants of Brucella (rough, omp25, bvrR) are unable to control DCs maturation and thus effectively stimulate naïve T cells, which could be the origin of the protective immunity elicited by these mutants in vivo. In this study, we investigate the interactions of a VirB-defective Brucella mutant with human DCs to determine whether its attenuation could be attributed to the induction of an adaptive immune response. We show here that in contrast to previously studied strains and similar to wild-type strains, this virB mutant was unable to trigger significant DC maturation. Together with recently published data describing infection with virB mutants in vivo, these results suggest that Brucella T4SS VirB is not involved in the control of DC maturation and does not interfere with the establishment of a T-helper type 1 adaptive immune response.
Collapse
Affiliation(s)
- Elisabeth Billard
- Centre d'étude d'agents Pathogènes et Biotechnologies pour la Santé (CPBS) Université Montpellier 1, Montpellier, France
| | | | | |
Collapse
|
44
|
VirB3 to VirB6 and VirB8 to VirB11, but not VirB7, are essential for mediating persistence of Brucella in the reticuloendothelial system. J Bacteriol 2008; 190:4427-36. [PMID: 18469100 DOI: 10.1128/jb.00406-08] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The Brucella abortus virB locus contains 12 open reading frames, termed virB1 through virB12, which encode a type IV secretion system. Polar mutations in the virB locus markedly reduce the ability of B. abortus to survive in cultured macrophages or to persist in organs of mice. While a nonpolar deletion of the virB2 gene reduces survival in cultured macrophages and in organs of mice, a nonpolar deletion of virB1 only reduces survival in macrophages, whereas virB12 is dispensable for either virulence trait. Here we investigated the role of the remaining genes in the virB locus during survival in macrophages and virulence in mice. Mutants carrying nonpolar deletions of the virB3, virB4, virB5, virB6, virB7, virB8, virB9, virB10, or virB11 gene were constructed and characterized. All mutations reduced the ability of B. abortus to survive in J774A.1 mouse macrophage-like cells to a degree similar to that caused by a deletion of the entire virB locus. Deletion of virB3, virB4, virB5, virB6, virB8, virB9, virB10, or virB11 markedly reduced the ability of B. abortus to persist in the spleens of mice at 8 weeks after infection. Interestingly, deletion of virB7 did not reduce the ability of B. abortus to persist in spleens of mice. We conclude that virB2, virB3, virB4, virB5, virB6, virB8, virB9, virB10, and virB11 are essential for virulence of B. abortus in mice, while functions encoded by the virB1, virB7, and virB12 genes are not required for persistence in organs with this animal model.
Collapse
|
45
|
Inactivation of the type IV secretion system reduces the Th1 polarization of the immune response to Brucella abortus infection. Infect Immun 2008; 76:3207-13. [PMID: 18458071 DOI: 10.1128/iai.00203-08] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The Brucella abortus type IV secretion system (T4SS), encoded by the virB operon, is essential for establishing persistent infection in the murine reticuloendothelial system. To gain insight into the in vivo interactions mediated by the T4SS, we compared host responses elicited by B. abortus with those of an isogenic mutant in the virB operon. Mice infected with the B. abortus virB mutant elicited smaller increases in serum levels of immunoglobulin G2a, gamma interferon (IFN-gamma), and interleukin-12p40 than did mice infected with wild-type B. abortus. Despite equal bacterial loads in the spleen, at 3 to 4 days postinfection, levels of IFN-gamma were higher in mice infected with wild-type B. abortus than in mice infected with the virB mutant, as shown by real-time PCR, intracellular cytokine staining, and cytokine levels. IFN-gamma-producing CD4(+) T cells were more abundant in spleens of mice infected with wild-type B. abortus than in virB mutant-infected mice. Similar numbers of IFN-gamma-secreting CD8(+) T cells were observed in the spleens of mice infected with B. abortus 2308 or a virB mutant. These results suggest that early differences in cytokine responses contribute to a stronger Th1 polarization of the immune response in mice infected with wild-type B. abortus than in mice infected with the virB mutant.
Collapse
|
46
|
Nijskens C, Copin R, De Bolle X, Letesson JJ. Intracellular rescuing of a B. melitensis 16M virB mutant by co-infection with a wild type strain. Microb Pathog 2008; 45:134-41. [PMID: 18547782 DOI: 10.1016/j.micpath.2008.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Revised: 04/14/2008] [Accepted: 04/18/2008] [Indexed: 10/22/2022]
Abstract
Brucella is a broad-range, facultative intracellular pathogen that can survive and replicate in an endoplasmic reticulum (ER)-derived replication niche by preventing fusion of its membrane-bound compartment with late endosomes and lysosomes. This vacuolar hijacking was demonstrated to be dependent on the type IV secretion system VirB but no secreted effectors have been identified yet. A virB mutant is unable to reach its ER-derived replicative niche and does not multiply intracellularly. In this paper, we showed that, by co-infecting bovine macrophages or HeLa cells with the wild type (WT) strain of Brucella melitensis 16M and a deletion mutant of the complete virB operon, the replication of DeltavirB is rescued in almost 20% of the co-infected cells. Furthermore, we demonstrated that co-infections with the WT strains of Brucella abortus or Brucella suis were equally able to rescue the replication of the B. melitensis DeltavirB mutant. By contrast, no rescue was observed when the WT strain was given 1h before or after the infection with the DeltavirB mutant. Finally, vacuoles containing the rescued DeltavirB mutant were shown to exclude the LAMP-1 marker in a way similar to the WT containing vacuoles.
Collapse
Affiliation(s)
- C Nijskens
- Unité de Recherche en Biologie Moléculaire (URBM), University of Namur, Namur, Belgium
| | | | | | | |
Collapse
|
47
|
VirB12 is a serological marker of Brucella infection in experimental and natural hosts. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2007; 15:208-14. [PMID: 18077620 DOI: 10.1128/cvi.00374-07] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The Brucella species type IV secretion system, encoded by the virB1-12 locus, is required for intracellular replication and persistent infection in vivo. The requirement of VirB proteins for infection suggests that they are expressed in vivo and may therefore represent serological markers of infection. To test this idea, we purified recombinant VirB1, VirB5, VirB11, and VirB12 and tested for their recognition by antibodies in sera from experimentally infected mice and goats by using an indirect enzyme-linked immunosorbent assay. Antibody responses to VirB12 but not to VirB1, VirB5, or VirB11 were detected in 20/20 mice experimentally inoculated with Brucella abortus and 12/12 goats experimentally infected with Brucella melitensis. The potential use of VirB12 as a serological tool for the diagnosis of brucellosis was evaluated in the natural bovine host. Serum samples from 145 cattle of known serology (29% negative and 71% positive) were analyzed for the production of antibody responses to VirB12. One hundred two cattle samples (70.3%) were positive for antibodies to VirB12, while 43 samples were negative (29.7%). A positive serological response to VirB12 correlated with positive serology to whole B. abortus antigen in 99% of samples tested. These results show that VirB12 is expressed during infection of both experimental and natural hosts of Brucella species, and they suggest that VirB12 may be a useful serodiagnostic marker for brucellosis.
Collapse
|
48
|
Wilson RP, Raffatellu M, Chessa D, Winter SE, Tükel C, Bäumler AJ. The Vi-capsule prevents Toll-like receptor 4 recognition of Salmonella. Cell Microbiol 2007; 10:876-90. [PMID: 18034866 DOI: 10.1111/j.1462-5822.2007.01090.x] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The viaB locus enables Salmonella enterica serotype Typhi to reduce Toll-like receptor (TLR) dependent cytokine production in tissue culture models. This DNA region contains genes involved in the regulation (tviA), biosynthesis (tviBCDE) and export (vexABCDE) of the Vi capsule. Expression of the Vi capsule in S. Typhimurium, but not expression of the TviA regulatory protein, reduced tumour necrosis factor-alpha (TNF-alpha) and IL-6 production by murine bone-marrow derived macrophages. Production of TNF-alpha and IL-6 was dependent on expression of TLR4 as stimulation of macrophages from TLR4(-/-) mice with S. Typhimurium did not result in expression of these cytokines. Intraperitoneal infection of mice with S. Typhimurium induced expression of TNF-alpha and inducible nitric oxide synthase (iNOS) in the liver. Introduction of the cloned viaB region into S. Typhimurium reduced TNF-alpha and iNOS expression to levels observed after infection with a S. Typhimurium msbB mutant. In contrast, no differences in TNF-alpha expression between the S. Typhimurium wild type and strains expressing the Vi-capsule or carrying a mutation in msbB were observed after infection of TLR4(-/-) mice. We conclude that the Vi capsule prevents both in vitro and in vivo recognition of S. Typhimurium lipopolysaccharide by TLR4.
Collapse
Affiliation(s)
- R Paul Wilson
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave., Davis, CA, USA
| | | | | | | | | | | |
Collapse
|