1
|
Li Z, Wang S, Han J, Shi C, Xi L, Cui Y, Zhang H. Expression of cytokine and Apoptosis-Associated genes in mice bone Marrow-Derived Macrophages stimulated with Brucella recombinant type IV secretion effectors. Cytokine 2024; 182:156711. [PMID: 39094437 DOI: 10.1016/j.cyto.2024.156711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/23/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Brucellosis is an economically important infectious caused by most commonly by Brucella. Detection of infected animals at the early stage is important for controlling the disease. The diagnostic antigens, usually protein antigens, have attracted much interest. However, the accurate mechanism of immune response is still unknown. The secretory effectors (BPE005, BPE275, and BPE123) of the type IV secretion system (T4SS) were involved in the intracellular circulation process of Brucella and the immune responses of the host. METHODS Genes encoding three B. abortus effector proteins (BPE005, BPE275, and BPE123) of T4SS were cloned and the recombinant proteins were expressed and purified. The purified recombinant proteins were named rBPE005, rBPE275 and rBPE123. Then, the expressions of Th1- and Th2-related cytokine genes were analyzed in mice bone marrow-derived macrophages (BMDMs) after stimulation with rBPE005, rBPE275, and rBPE123. Furthermore, four apoptosis-associated genes (Caspase-3, Caspase-8, Bax, and Bcl-2) were also detected to explore the damage of the proteins to the cells. RESULTS Expressions of all Th1- and Th2-related cytokine genes were induced with three proteins, and different cytokine expression patterns induced by each protein depend on the stimulation time and dose of protein. However, expressions of apoptosis-related genes did not change. CONCLUSION These results showed that the secreted antigens of Brucella induced an immune reaction via the production of Th1- and Th2-type cytokines in BMDMs without exerting any damage on the cells.
Collapse
Affiliation(s)
- Zhiqiang Li
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Shuli Wang
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Jincheng Han
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Chuanxin Shi
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Li Xi
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Yanyan Cui
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Hui Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang Province, China.
| |
Collapse
|
2
|
Marchesini MI, Spera JM, Comerci DJ. The 'ins and outs' of Brucella intracellular journey. Curr Opin Microbiol 2024; 78:102427. [PMID: 38309247 DOI: 10.1016/j.mib.2024.102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 02/05/2024]
Abstract
Members of the genus Brucella are the causative agents of brucellosis, a worldwide zoonosis affecting wild and domestic animals and humans. These facultative intracellular pathogens cause long-lasting chronic infections by evolving sophisticated strategies to counteract, evade, or subvert host bactericidal mechanisms in order to establish a secure replicative niche necessary for their survival. In this review, we present recent findings on selected Brucella effectors to illustrate how this pathogen modulates host cell signaling pathways to gain control of the vacuole, promote the formation of a safe intracellular replication niche, alter host cell metabolism to its advantage, and exploit various cellular pathways to ensure egress from the infected cell.
Collapse
Affiliation(s)
- María I Marchesini
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Buenos Aires, Argentina
| | - Juan M Spera
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Buenos Aires, Argentina
| | - Diego J Comerci
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Buenos Aires, Argentina; Comisión Nacional de Energía Atómica, Grupo Pecuario, Centro Atómico Ezeiza, Buenos Aires, Argentina.
| |
Collapse
|
3
|
Malmsheimer S, Grin I, Bohn E, Franz-Wachtel M, Macek B, Sahr T, Smollich F, Chetrit D, Meir A, Roy C, Buchrieser C, Wagner S. The T4bSS of Legionella features a two-step secretion pathway with an inner membrane intermediate for secretion of transmembrane effectors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.584949. [PMID: 38559167 PMCID: PMC10980071 DOI: 10.1101/2024.03.14.584949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
To promote intracellular survival and infection, Legionella spp. translocate hundreds of effector proteins into eukaryotic host cells using a type IV b protein secretion system (T4bSS). T4bSS are well known to translocate soluble as well as transmembrane domain-containing effector proteins (TMD-effectors) but the mechanisms of secretion are still poorly understood. Herein we investigated the secretion of hydrophobic TMD-effectors, of which about 80 were previously reported to be encoded by L. pneumophila. A proteomic analysis of fractionated membranes revealed that TMD-effectors are targeted to and inserted into the bacterial inner membranes of L. pneumophila independent of the presence of a functional T4bSS. While the T4bSS chaperones IcmS and IcmW were critical for secretion of all tested TMD-effectors, they did not influence inner membrane targeting of these proteins. As for soluble effector proteins, translocation of TMD-effectors into host cells depended on a C-terminal secretion signal and this signal needed to be presented towards the cytoplasmic side of the inner membrane. A different secretion behavior of TMD- and soluble effectors and the need for small periplasmic loops within TMD-effectors provided strong evidence that TMD-effectors are secreted in a two-step secretion process: Initially, an inner membrane intermediate is formed, that is extracted towards the cytoplasmic side, possibly by the help of the type IV coupling protein complex and subsequently secreted into eukaryotic host cells by the T4bSS core complex. Overall, our study highlights the amazing versatility of T4bSS to secrete soluble and TMD-effectors from different subcellular locations of the bacterial cell.
Collapse
Affiliation(s)
- Silke Malmsheimer
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), Section of Cellular and Molecular Microbiology, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- Current address: Institut de Recherche en Infectiologie de Montpellier, Equipe Kremer, UMR 9004 - CNRS / UM, 1919 route de Mende, 34293 Montpellier cedex 5, France
| | - Iwan Grin
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), Section of Cellular and Molecular Microbiology, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - Erwin Bohn
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), Institute of Medical Microbiology and Hygiene, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - Mirita Franz-Wachtel
- University of Tübingen, Proteome Center Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Boris Macek
- University of Tübingen, Proteome Center Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Tobias Sahr
- Institute Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires, 75015 Paris, France
| | - Fabian Smollich
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), Section of Cellular and Molecular Microbiology, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - David Chetrit
- Yale University, Department of Microbial Pathogenesis, 295 Congress Avenue, New Haven, CT 06536-0812, USA
| | - Amit Meir
- Yale University, Department of Microbial Pathogenesis, 295 Congress Avenue, New Haven, CT 06536-0812, USA
- Birkbeck Institute of Structural and Molecular Biology, Birkbeck and UCL, Malet Street, London WC1E 7HX, UK
- Current address: University of Glasgow, MRC Centre for Virus Research, School of Infection and Immunity, Glasgow, UK
| | - Craig Roy
- Yale University, Department of Microbial Pathogenesis, 295 Congress Avenue, New Haven, CT 06536-0812, USA
| | - Carmen Buchrieser
- Institute Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires, 75015 Paris, France
| | - Samuel Wagner
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), Section of Cellular and Molecular Microbiology, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), Elfriede-Aulhorn-Str. 6, 72076 Tébingen, Germany
- German Center for Infection Research (DZIF), partner-site Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| |
Collapse
|
4
|
Anbazhagan S, Himani KM, Karthikeyan R, Prakasan L, Dinesh M, Nair SS, Lalsiamthara J, Abhishek, Ramachandra SG, Chaturvedi VK, Chaudhuri P, Thomas P. Comparative genomics of Brucella abortus and Brucella melitensis unravels the gene sharing, virulence factors and SNP diversity among the standard, vaccine and field strains. Int Microbiol 2024; 27:101-111. [PMID: 37202587 DOI: 10.1007/s10123-023-00374-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/29/2023] [Accepted: 05/09/2023] [Indexed: 05/20/2023]
Abstract
Brucella abortus and Brucella melitensis are the primary etiological agents of brucellosis in large and small ruminants, respectively. There are limited comparative genomic studies involving Brucella strains that explore the relatedness among both species. In this study, we involved strains (n=44) representing standard, vaccine and Indian field origin for pangenome, single nucleotide polymorphism (SNP) and phylogenetic analysis. Both species shared a common gene pool representing 2884 genes out of a total 3244 genes. SNP-based phylogenetic analysis indicated higher SNP diversity among B. melitensis (3824) strains in comparison to B. abortus (540) strains, and a clear demarcation was identified between standard/vaccine and field strains. The analysis for virulence genes revealed that virB3, virB7, ricA, virB5, ipx5, wbkC, wbkB, and acpXL genes were highly conserved in most of the Brucella strains. Interestingly, virB10 gene was found to have high variability among the B. abortus strains. The cgMLST analysis revealed distinct sequence types for the standard/vaccine and field strains. B. abortus strains from north-eastern India fall within similar sequence type differing from other strains. In conclusion, the analysis revealed a highly shared core genome among two Brucella species. SNP analysis revealed B. melitensis strains exhibit high diversity as compared to B. abortus strains. Strains with absence or high polymorphism of virulence genes can be exploited for the development of novel vaccine candidates effective against both B. abortus and B. melitensis.
Collapse
Affiliation(s)
- S Anbazhagan
- Division of Bacteriology and Mycology, ICAR- Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
- ICMR-National Animal Resource Facility for Biomedical Research, Hyderabad, India
| | - K M Himani
- Division of Bacteriology and Mycology, ICAR- Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - R Karthikeyan
- Division of Epidemiology, ICAR- Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Lakshmi Prakasan
- Division of Bacteriology and Mycology, ICAR- Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - M Dinesh
- Division of Pathology, ICAR- Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Sonu S Nair
- Division of Bacteriology and Mycology, ICAR- Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Jonathan Lalsiamthara
- Department of Molecular Microbiology & Immunology, SOM, OHSU, Portland, OR, US, 97239, USA
| | - Abhishek
- Division of Bacteriology and Mycology, ICAR- Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - S G Ramachandra
- ICMR-National Animal Resource Facility for Biomedical Research, Hyderabad, India
| | - V K Chaturvedi
- Division of Bacteriology and Mycology, ICAR- Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Pallab Chaudhuri
- Division of Bacteriology and Mycology, ICAR- Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India.
| | - Prasad Thomas
- Division of Bacteriology and Mycology, ICAR- Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India.
| |
Collapse
|
5
|
Zheng M, Lin R, Zhu J, Dong Q, Chen J, Jiang P, Zhang H, Liu J, Chen Z. Effector Proteins of Type IV Secretion System: Weapons of Brucella Used to Fight Against Host Immunity. Curr Stem Cell Res Ther 2024; 19:145-153. [PMID: 36809969 DOI: 10.2174/1574888x18666230222124529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/15/2022] [Accepted: 12/29/2022] [Indexed: 02/24/2023]
Abstract
Brucella is an intracellular bacterial pathogen capable of long-term persistence in the host, resulting in chronic infections in livestock and wildlife. The type IV secretion system (T4SS) is an important virulence factor of Brucella and is composed of 12 protein complexes encoded by the VirB operon. T4SS exerts its function through its secreted 15 effector proteins. The effector proteins act on important signaling pathways in host cells, inducing host immune responses and promoting the survival and replication of Brucella in host cells to promote persistent infection. In this article, we describe the intracellular circulation of Brucella-infected cells and survey the role of Brucella VirB T4SS in regulating inflammatory responses and suppressing host immune responses during infection. In addition, the important mechanisms of these 15 effector proteins in resisting the host immune response during Brucella infection are elucidated. For example, VceC and VceA assist in achieving sustained survival of Brucella in host cells by affecting autophagy and apoptosis. BtpB, together with BtpA, controls the activation of dendritic cells during infection, induces inflammatory responses, and controls host immunity. This article reviews the effector proteins secreted by Brucella T4SS and their involvement in immune responses, which can provide a reliable theoretical basis for the subsequent mechanism of hijacking the host cell signaling pathway by bacteria and contribute to the development of better vaccines to effectively treat Brucella bacterial infection.
Collapse
Affiliation(s)
- Min Zheng
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, 110866, Shenyang, China
| | - Ruiqi Lin
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, 110866, Shenyang, China
| | - Jinying Zhu
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, 110866, Shenyang, China
| | - Qiao Dong
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, 110866, Shenyang, China
| | - Jingjing Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, 110866, Shenyang, China
| | - Pengfei Jiang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, 110866, Shenyang, China
| | - Huan Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, 110866, Shenyang, China
| | - Jinling Liu
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, 110866, Shenyang, China
| | - Zeliang Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, 110866, Shenyang, China
| |
Collapse
|
6
|
Kambarev S, Borghesan E, Miller CN, Myeni S, Celli J. The Brucella abortus Type IV Effector BspA Inhibits MARCH6-Dependent ERAD To Promote Intracellular Growth. Infect Immun 2023; 91:e0013023. [PMID: 37129527 PMCID: PMC10187129 DOI: 10.1128/iai.00130-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/03/2023] Open
Abstract
Brucella abortus, the intracellular causative agent of brucellosis, relies on type IV secretion system (T4SS) effector-mediated modulation of host cell functions to establish a replicative niche, the Brucella-containing vacuole (BCV). Brucella exploits the host's endocytic, secretory, and autophagic pathways to modulate the nature and function of its vacuole from an endocytic BCV (eBCV) to an endoplasmic reticulum (ER)-derived replicative BCV (rBCV) to an autophagic egress BCV (aBCV). A role for the host ER-associated degradation pathway (ERAD) in the B. abortus intracellular cycle was recently uncovered, as it is enhanced by the T4SS effector BspL to control the timing of aBCV-mediated egress. Here, we show that the T4SS effector BspA also interferes with ERAD, yet to promote B. abortus intracellular proliferation. BspA was required for B. abortus replication in bone marrow-derived macrophages and interacts with membrane-associated RING-CH-type finger 6 (MARCH6), a host E3 ubiquitin ligase involved in ERAD. Pharmacological inhibition of ERAD and small interfering RNA (siRNA) depletion of MARCH6 did not affect the replication of wild-type B. abortus but rescued the replication defect of a bspA deletion mutant, while depletion of the ERAD component UbxD8 affected replication of B. abortus and rescued the replication defect of the bspA mutant. BspA affected the degradation of ERAD substrates and destabilized the MARCH6 E3 ligase complex. Taken together, these findings indicate that BspA inhibits the host ERAD pathway via targeting of MARCH6 to promote B. abortus intracellular growth. Our data reveal that targeting ERAD components by type IV effectors emerges as a multifaceted theme in Brucella pathogenesis.
Collapse
Affiliation(s)
- Stanimir Kambarev
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Elizabeth Borghesan
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Cheryl N. Miller
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Sebenzile Myeni
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Jean Celli
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
- Department of Microbiology and Molecular Genetics, Larner College of Medicine at the University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
7
|
Orsini M, Ianni A, Zinzula L. Brucella ceti and Brucella pinnipedialis genome characterization unveils genetic features that highlight their zoonotic potential. Microbiologyopen 2022; 11:e1329. [PMID: 36314752 PMCID: PMC9597259 DOI: 10.1002/mbo3.1329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
The Gram-negative bacteria Brucella ceti and Brucella pinnipedialis circulate in marine environments primarily infecting marine mammals, where they cause an often-fatal disease named brucellosis. The increase of brucellosis among several species of cetaceans and pinnipeds, together with the report of sporadic human infections, raises concerns about the zoonotic potential of these pathogens on a large scale and may pose a threat to coastal communities worldwide. Therefore, the characterization of the B. ceti and B. pinnipedialis genetic features is a priority to better understand the pathological factors that may impact global health. Moreover, an in-depth functional analysis of the B. ceti and B. pinnipedialis genome in the context of virulence and pathogenesis was not undertaken so far. Within this picture, here we present the comparative whole-genome characterization of all B. ceti and B. pinnipedialis genomes available in public resources, uncovering a collection of genetic tools possessed by these aquatic bacterial species compared to their zoonotic terrestrial relatives. We show that B. ceti and B. pinnipedialis genomes display a wide host-range infection capability and a polyphyletic phylogeny within the genus, showing a genomic structure that fits the canonical definition of closeness. Functional genome annotation led to identifying genes related to several pathways involved in mechanisms of infection, others conferring pan-susceptibility to antimicrobials and a set of virulence genes that highlight the similarity of B. ceti and B. pinnipedialis genotypes to those of Brucella spp. displaying human-infecting phenotypes.
Collapse
Affiliation(s)
- Massimiliano Orsini
- Istituto Zooprofilattico Sperimentale delle Venezie, Laboratory of Microbial Ecology and GenomicsLegnaroItaly
| | - Andrea Ianni
- Research Unit in Hygiene, Statistics and Public HealthCampus Bio‐Medico di Roma UniversityRomeItaly
| | - Luca Zinzula
- Department of Molecular Structural BiologyMax Planck Institute of BiochemistryMartinsriedGermany
- Centro di Educazione Ambientale e alla Sostenibilità (CEAS) Laguna di NoraPulaItaly
| |
Collapse
|
8
|
Mode S, Ketterer M, Québatte M, Dehio C. Antibiotic persistence of intracellular Brucella abortus. PLoS Negl Trop Dis 2022; 16:e0010635. [PMID: 35881641 PMCID: PMC9355222 DOI: 10.1371/journal.pntd.0010635] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 08/05/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022] Open
Abstract
Background Human brucellosis caused by the facultative intracellular pathogen Brucella spp. is an endemic bacterial zoonosis manifesting as acute or chronic infections with high morbidity. Treatment typically involves a combination therapy of two antibiotics for several weeks to months, but despite this harsh treatment relapses occur at a rate of 5–15%. Although poor compliance and reinfection may account for a fraction of the observed relapse cases, it is apparent that the properties of the infectious agent itself may play a decisive role in this phenomenon. Methodology/Principal findings We used B. abortus carrying a dual reporter in a macrophage infection model to gain a better understanding of the efficacy of recommended therapies in cellulo. For this we used automated fluorescent microscopy as a prime read-out and developed specific CellProfiler pipelines to score infected macrophages at the population and the single cell level. Combining microscopy of constitutive and induced reporters with classical CFU determination, we quantified the protective nature of the Brucella intracellular lifestyle to various antibiotics and the ability of B. abortus to persist in cellulo despite harsh antibiotic treatments. Conclusion/Significance We demonstrate that treatment of infected macrophages with antibiotics at recommended concentrations fails to fully prevent growth and persistence of B. abortus in cellulo, which may be explained by a protective nature of the intracellular niche(s). Moreover, we show the presence of bona fide intracellular persisters upon antibiotic treatment, which are metabolically active and retain the full infectious potential, therefore constituting a plausible reservoir for reinfection and relapse. In conclusion, our results highlight the need to extend the spectrum of models to test new antimicrobial therapies for brucellosis to better reflect the in vivo infection environment, and to develop therapeutic approaches targeting the persister subpopulation. Brucellosis is a zoonosis endemic to many low- and middle-income countries around the world. Therapies recommended by the WHO are comprised of at least two antibiotics for several weeks, sometimes months. Relapses are frequent despite these harsh treatments. The underlying reasons for these relapses, besides reinfection and non-compliance to treatment, are unknown. Our study shows that Brucella abortus can form so called “persisters” in rich broth but also inside macrophages. This small bacterial subpopulation survives antibiotic treatment and resumes growth after removal of the antibiotics and could therefore serve as a reservoir for relapses in human brucellosis. Furthermore, we show that the intracellular lifestyle of Brucella has protective properties against recommended antibiotics as observed for other intracellular pathogens, highlighting the necessity to develop new infection models to assess antibiotic efficacy.
Collapse
Affiliation(s)
- Selma Mode
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - Maxime Québatte
- Biozentrum, University of Basel, Basel, Switzerland
- * E-mail: (MQ); (CD)
| | - Christoph Dehio
- Biozentrum, University of Basel, Basel, Switzerland
- * E-mail: (MQ); (CD)
| |
Collapse
|
9
|
Wells KM, He K, Pandey A, Cabello A, Zhang D, Yang J, Gomez G, Liu Y, Chang H, Li X, Zhang H, Feng X, da Costa LF, Metz R, Johnson CD, Martin CL, Skrobarczyk J, Berghman LR, Patrick KL, Leibowitz J, Ficht A, Sze SH, Song J, Qian X, Qin QM, Ficht TA, de Figueiredo P. Brucella activates the host RIDD pathway to subvert BLOS1-directed immune defense. eLife 2022; 11:e73625. [PMID: 35587649 PMCID: PMC9119680 DOI: 10.7554/elife.73625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 04/26/2022] [Indexed: 11/18/2022] Open
Abstract
The phagocytosis and destruction of pathogens in lysosomes constitute central elements of innate immune defense. Here, we show that Brucella, the causative agent of brucellosis, the most prevalent bacterial zoonosis globally, subverts this immune defense pathway by activating regulated IRE1α-dependent decay (RIDD) of Bloc1s1 mRNA encoding BLOS1, a protein that promotes endosome-lysosome fusion. RIDD-deficient cells and mice harboring a RIDD-incompetent variant of IRE1α were resistant to infection. Inactivation of the Bloc1s1 gene impaired the ability to assemble BLOC-1-related complex (BORC), resulting in differential recruitment of BORC-related lysosome trafficking components, perinuclear trafficking of Brucella-containing vacuoles (BCVs), and enhanced susceptibility to infection. The RIDD-resistant Bloc1s1 variant maintains the integrity of BORC and a higher-level association of BORC-related components that promote centrifugal lysosome trafficking, resulting in enhanced BCV peripheral trafficking and lysosomal destruction, and resistance to infection. These findings demonstrate that host RIDD activity on BLOS1 regulates Brucella intracellular parasitism by disrupting BORC-directed lysosomal trafficking. Notably, coronavirus murine hepatitis virus also subverted the RIDD-BLOS1 axis to promote intracellular replication. Our work establishes BLOS1 as a novel immune defense factor whose activity is hijacked by diverse pathogens.
Collapse
Affiliation(s)
- Kelsey Michelle Wells
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
| | - Kai He
- Department of Electrical and Computer Engineering, Texas A&M UniversityCollege StationUnited States
| | - Aseem Pandey
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
- Department of Veterinary Pathobiology, Texas A&M UniversityCollege StationUnited States
| | - Ana Cabello
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
- Department of Veterinary Pathobiology, Texas A&M UniversityCollege StationUnited States
| | - Dongmei Zhang
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
| | - Jing Yang
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
| | - Gabriel Gomez
- Texas A&M Veterinary Medical Diagnostic Laboratory, Texas A&M UniversityCollege StationUnited States
| | - Yue Liu
- College of Plant Sciences, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin UniversityJilinChina
| | - Haowu Chang
- Key Laboratory of Symbolic Computation and Knowledge Engineering, Ministry of Education, College of Computer Science and Technology, Jilin UniversityChangchunChina
| | - Xueqiang Li
- Key Laboratory of Symbolic Computation and Knowledge Engineering, Ministry of Education, College of Computer Science and Technology, Jilin UniversityChangchunChina
| | - Hao Zhang
- Key Laboratory of Symbolic Computation and Knowledge Engineering, Ministry of Education, College of Computer Science and Technology, Jilin UniversityChangchunChina
| | - Xuehuang Feng
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
| | | | - Richard Metz
- Genomics and Bioinformatics Services, Texas A&M UniversityCollege StationUnited States
| | - Charles D Johnson
- Genomics and Bioinformatics Services, Texas A&M UniversityCollege StationUnited States
| | - Cameron Lee Martin
- Department of Poultry Science, Texas A&M UniversityCollege StationUnited States
| | - Jill Skrobarczyk
- Department of Poultry Science, Texas A&M UniversityCollege StationUnited States
| | - Luc R Berghman
- Department of Poultry Science, Texas A&M UniversityCollege StationUnited States
| | - Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
| | - Julian Leibowitz
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
| | - Allison Ficht
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science CenterCollege StationUnited States
| | - Sing-Hoi Sze
- Department of Computer Science and Engineering, Dwight Look College of Engineering, Texas A&M UniversityCollege StationUnited States
- Department of Biochemistry & Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
| | - Xiaoning Qian
- Department of Electrical and Computer Engineering, Texas A&M UniversityCollege StationUnited States
- TEES-AgriLife Center for Bioinformatics & Genomic Systems Engineering, Texas A&M UniversityCollege StationUnited States
| | - Qing-Ming Qin
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
- College of Plant Sciences, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin UniversityJilinChina
| | - Thomas A Ficht
- Department of Veterinary Pathobiology, Texas A&M UniversityCollege StationUnited States
| | - Paul de Figueiredo
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science CenterBryanUnited States
- Department of Veterinary Pathobiology, Texas A&M UniversityCollege StationUnited States
| |
Collapse
|
10
|
Zhang X, Chen J, Dong Q, Zhu J, Peng R, He C, Li Y, Lin R, Jiang P, Zheng M, Zhang H, Liu S, Chen Z. Lysine Acylation Modification Landscape of Brucella abortus Proteome and its Virulent Proteins. Front Cell Dev Biol 2022; 10:839822. [PMID: 35300419 PMCID: PMC8921143 DOI: 10.3389/fcell.2022.839822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
The myriad of posttranslational modifications (PTMs) of proteins that occur in all living cells are crucial to all kinds of biological processes. Brucella is an intracellular parasitic bacterium that can cause chronic diseases in both humans and livestock. To reveal the relationship between PTMs and the virulence and survival of Brucella, we described the first comprehensive multiple PTM-omics atlas of B. abortus 2308. Five PTMs involving lysine, namely 2-hydroxyisobutyrylation, succinylation, crotonylation, acetylation, and malonylation were identified. Nearly 2,000 modified proteins were observed, and these proteins took part in many biological processes, with a variety of molecular functions. In addition, we detected many significant virulence factors of Brucella among the modified proteins. 10 of the 15 T4SS effector proteins were detected with one or more PTMs. Moreover, abundant PTMs were detected in other typical virulence factors. Considering the role of PTMs in various biological processes of Brucella virulence and survival, we propose that the virulence of Brucella is associated with the PTMs of proteins. Taken together, this study provides the first global survey of PTMs in Brucella. This is a prospective starting point for further functional analysis of PTMs during the survival of Brucella in hosts, interpretation of the function of Brucella proteins, and elucidation of the pathogenic mechanism of Brucella.
Collapse
Affiliation(s)
- Xi Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Jingjing Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Qiao Dong
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Jinying Zhu
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Ruihao Peng
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Chuanyu He
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Yuzhuo Li
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Ruiqi Lin
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Pengfei Jiang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Min Zheng
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Huan Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China
| | - Shiwei Liu
- Department of Nephrology and Endocrinology, Wangjing Hospital, Chinese Academy of Chinese Medical Science, Beijing, China
| | - Zeliang Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang, China.,Department of Nephrology and Endocrinology, Wangjing Hospital, Chinese Academy of Chinese Medical Science, Beijing, China.,Innovative Institute of Zoonoses, Medical College, Inner Mongolia Minzu University, Tongliao, China
| |
Collapse
|
11
|
Ma Z, Deng X, Li R, Hu R, Miao Y, Xu Y, Zheng W, Yi J, Wang Z, Wang Y, Chen C. Crosstalk of Brucella abortus nucleomodulin BspG and host DNA replication process/mitochondrial respiratory pathway promote anti-apoptosis and infection. Vet Microbiol 2022; 268:109414. [DOI: 10.1016/j.vetmic.2022.109414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/18/2022] [Accepted: 03/26/2022] [Indexed: 01/18/2023]
|
12
|
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: 8.0] [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
|
13
|
Xiong X, Li B, Zhou Z, Gu G, Li M, Liu J, Jiao H. The VirB System Plays a Crucial Role in Brucella Intracellular Infection. Int J Mol Sci 2021; 22:ijms222413637. [PMID: 34948430 PMCID: PMC8707931 DOI: 10.3390/ijms222413637] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is a highly prevalent zoonotic disease caused by Brucella. Brucella spp. are gram-negative facultative intracellular parasitic bacteria. Its intracellular survival and replication depend on a functional virB system, an operon encoded by VirB1–VirB12. Type IV secretion system (T4SS) encoded by the virB operon is an important virulence factor of Brucella. It can subvert cellular pathway and induce host immune response by secreting effectors, which promotes Brucella replication in host cells and induce persistent infection. Therefore, this paper summarizes the function and significance of the VirB system, focusing on the structure of the VirB system where VirB T4SS mediates biogenesis of the endoplasmic reticulum (ER)-derived replicative Brucella-containing vacuole (rBCV), the effectors of T4SS and the cellular pathways it subverts, which will help better understand the pathogenic mechanism of Brucella and provide new ideas for clinical vaccine research and development.
Collapse
Affiliation(s)
- Xue Xiong
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (X.X.); (B.L.); (Z.Z.); (G.G.); (M.L.)
| | - Bowen Li
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (X.X.); (B.L.); (Z.Z.); (G.G.); (M.L.)
| | - Zhixiong Zhou
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (X.X.); (B.L.); (Z.Z.); (G.G.); (M.L.)
| | - Guojing Gu
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (X.X.); (B.L.); (Z.Z.); (G.G.); (M.L.)
| | - Mengjuan Li
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (X.X.); (B.L.); (Z.Z.); (G.G.); (M.L.)
| | - Jun Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Yujinxiang Street 573, Changchun 130122, China
- Correspondence: (J.L.); (H.J.)
| | - Hanwei Jiao
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (X.X.); (B.L.); (Z.Z.); (G.G.); (M.L.)
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
- Veterinary Scientific Engineering Research Center, Chongqing 402460, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, China
- Correspondence: (J.L.); (H.J.)
| |
Collapse
|
14
|
Vaughn B, Abu Kwaik Y. Idiosyncratic Biogenesis of Intracellular Pathogens-Containing Vacuoles. Front Cell Infect Microbiol 2021; 11:722433. [PMID: 34858868 PMCID: PMC8632064 DOI: 10.3389/fcimb.2021.722433] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
While most bacterial species taken up by macrophages are degraded through processing of the bacteria-containing vacuole through the endosomal-lysosomal degradation pathway, intravacuolar pathogens have evolved to evade degradation through the endosomal-lysosomal pathway. All intra-vacuolar pathogens possess specialized secretion systems (T3SS-T7SS) that inject effector proteins into the host cell cytosol to modulate myriad of host cell processes and remodel their vacuoles into proliferative niches. Although intravacuolar pathogens utilize similar secretion systems to interfere with their vacuole biogenesis, each pathogen has evolved a unique toolbox of protein effectors injected into the host cell to interact with, and modulate, distinct host cell targets. Thus, intravacuolar pathogens have evolved clear idiosyncrasies in their interference with their vacuole biogenesis to generate a unique intravacuolar niche suitable for their own proliferation. While there has been a quantum leap in our knowledge of modulation of phagosome biogenesis by intravacuolar pathogens, the detailed biochemical and cellular processes affected remain to be deciphered. Here we discuss how the intravacuolar bacterial pathogens Salmonella, Chlamydia, Mycobacteria, Legionella, Brucella, Coxiella, and Anaplasma utilize their unique set of effectors injected into the host cell to interfere with endocytic, exocytic, and ER-to-Golgi vesicle traffic. However, Coxiella is the main exception for a bacterial pathogen that proliferates within the hydrolytic lysosomal compartment, but its T4SS is essential for adaptation and proliferation within the lysosomal-like vacuole.
Collapse
Affiliation(s)
- Bethany Vaughn
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,Center for Predictive Medicine, College of Medicine, University of Louisville, Louisville, KY, United States
| |
Collapse
|
15
|
Borghesan E, Smith EP, Myeni S, Binder K, Knodler LA, Celli J. A Brucella effector modulates the Arf6-Rab8a GTPase cascade to promote intravacuolar replication. EMBO J 2021; 40:e107664. [PMID: 34423453 PMCID: PMC8488576 DOI: 10.15252/embj.2021107664] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 01/15/2023] Open
Abstract
Remodeling of host cellular membrane transport pathways is a common pathogenic trait of many intracellular microbes that is essential to their intravacuolar life cycle and proliferation. The bacterium Brucella abortus generates a host endoplasmic reticulum‐derived vacuole (rBCV) that supports its intracellular growth, via VirB Type IV secretion system‐mediated delivery of effector proteins, whose functions and mode of action are mostly unknown. Here, we show that the effector BspF specifically promotes Brucella replication within rBCVs by interfering with vesicular transport between the trans‐Golgi network (TGN) and recycling endocytic compartment. BspF targeted the recycling endosome, inhibited retrograde traffic to the TGN, and interacted with the Arf6 GTPase‐activating Protein (GAP) ACAP1 to dysregulate Arf6‐/Rab8a‐dependent transport within the recycling endosome, which resulted in accretion of TGN‐associated vesicles by rBCVs and enhanced bacterial growth. Altogether, these findings provide mechanistic insight into bacterial modulation of membrane transport used to promote their own proliferation within intracellular vacuoles.
Collapse
Affiliation(s)
- Elizabeth Borghesan
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Erin P Smith
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Sebenzile Myeni
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kelsey Binder
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Leigh A Knodler
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Jean Celli
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA.,Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
16
|
Karthik K, Anbazhagan S, Thomas P, Ananda Chitra M, Senthilkumar TMA, Sridhar R, Dhinakar Raj G. Genome Sequencing and Comparative Genomics of Indian Isolates of Brucella melitensis. Front Microbiol 2021; 12:698069. [PMID: 34489888 PMCID: PMC8417702 DOI: 10.3389/fmicb.2021.698069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/19/2021] [Indexed: 11/25/2022] Open
Abstract
Brucella melitensis causes small ruminant brucellosis and a zoonotic pathogen prevalent worldwide. Whole genome phylogeny of all available B. melitensis genomes (n = 355) revealed that all Indian isolates (n = 16) clustered in the East Mediterranean lineage except the ADMAS-GI strain. Pangenome analysis indicated the presence of limited accessory genomes with few clades showing specific gene presence/absence pattern. A total of 43 virulence genes were predicted in all the Indian strains of B. melitensis except 2007BM-1 (ricA and wbkA are absent). Multilocus sequence typing (MLST) analysis indicated all except one Indian strain (ADMAS-GI) falling into sequence type (ST 8). In comparison with MLST, core genome phylogeny indicated two major clusters (>70% bootstrap support values) among Indian strains. Clusters with <70% bootstrap support values represent strains with diverse evolutionary origins present among animal and human hosts. Genetic relatedness among animal (sheep and goats) and human strains with 100% bootstrap values shows its zoonotic transfer potentiality. SNP-based analysis indicated similar clustering to that of core genome phylogeny. Among the Indian strains, the highest number of unique SNPs (112 SNPs) were shared by a node that involved three strains from Tamil Nadu. The node SNPs involved several peptidase genes like U32, M16 inactive domain protein, clp protease family protein, and M23 family protein and mostly represented non-synonymous (NS) substitutions. Vaccination has been followed in several parts of the world to prevent small ruminant brucellosis but not in India. Comparison of Indian strains with vaccine strains showed that M5 is genetically closer to most of the Indian strains than Rev.1 strain. The presence of most of the virulence genes among all Indian strains and conserved core genome compositions suggest the use of any circulating strain/genotypes for the development of a vaccine candidate for small ruminant brucellosis in India.
Collapse
Affiliation(s)
- Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Subbaiyan Anbazhagan
- Division of Bacteriology and Mycology, Indian Veterinary Research Institute, Bareilly, India
| | - Prasad Thomas
- Division of Bacteriology and Mycology, Indian Veterinary Research Institute, Bareilly, India
| | - Murugesan Ananda Chitra
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | | | - Ramaswamy Sridhar
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Gopal Dhinakar Raj
- Centre for Animal Health Studies, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| |
Collapse
|
17
|
Whole-Genome Sequencing for Tracing the Genetic Diversity of Brucella abortus and Brucella melitensis Isolated from Livestock in Egypt. Pathogens 2021; 10:pathogens10060759. [PMID: 34208761 PMCID: PMC8235727 DOI: 10.3390/pathogens10060759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 02/03/2023] Open
Abstract
Brucellosis is a highly contagious zoonosis that occurs worldwide. Whole-genome sequencing (WGS) has become a widely accepted molecular typing method for outbreak tracing and genomic epidemiology of brucellosis. Twenty-nine Brucella spp. (eight B. abortus biovar 1 and 21 B. melitensis biovar 3) were isolated from lymph nodes, milk, and fetal abomasal contents of infected cattle, buffaloes, sheep, and goats originating from nine districts in Egypt. The isolates were identified by microbiological methods and matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS). Differentiation and genotyping were confirmed using multiplex PCR. Illumina MiSeq® was used to sequence the 29 Brucella isolates. Using MLST typing, ST11 and ST1 were identified among B. melitensis and B. abortus, respectively. Brucella abortus and B. melitensis isolates were divided into two main clusters (clusters 1 and 2) containing two and nine distinct genotypes by core-genome SNP analysis, respectively. The genotypes were irregularly distributed over time and space in the study area. Both Egyptian B. abortus and B. melitensis isolates proved to be genomically unique upon comparison with publicly available sequencing from strains of neighboring Mediterranean, African, and Asian countries. The antimicrobial resistance mechanism caused by mutations in rpoB, gyrA, and gyrB genes associated with rifampicin and ciprofloxacin resistance were identified. To the best of our knowledge, this is the first study investigating the epidemiology of Brucella isolates from livestock belonging to different localities in Egypt based on whole genome analysis.
Collapse
|
18
|
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: 15.7] [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
|
19
|
González-Espinoza G, Arce-Gorvel V, Mémet S, Gorvel JP. Brucella: Reservoirs and Niches in Animals and Humans. Pathogens 2021; 10:pathogens10020186. [PMID: 33572264 PMCID: PMC7915599 DOI: 10.3390/pathogens10020186] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 01/18/2023] Open
Abstract
Brucella is an intracellular bacterium that causes abortion, reproduction failure in livestock and leads to a debilitating flu-like illness with serious chronic complications if untreated in humans. As a successful intracellular pathogen, Brucella has developed strategies to avoid recognition by the immune system of the host and promote its survival and replication. In vivo, Brucellae reside mostly within phagocytes and other cells including trophoblasts, where they establish a preferred replicative niche inside the endoplasmic reticulum. This process is central as it gives Brucella the ability to maintain replicating-surviving cycles for long periods of time, even at low bacterial numbers, in its cellular niches. In this review, we propose that Brucella takes advantage of the environment provided by the cellular niches in which it resides to generate reservoirs and disseminate to other organs. We will discuss how the favored cellular niches for Brucella infection in the host give rise to anatomical reservoirs that may lead to chronic infections or persistence in asymptomatic subjects, and which may be considered as a threat for further contamination. A special emphasis will be put on bone marrow, lymph nodes, reproductive and for the first time adipose tissues, as well as wildlife reservoirs.
Collapse
|
20
|
Rajendhran J. Genomic insights into Brucella. INFECTION GENETICS AND EVOLUTION 2020; 87:104635. [PMID: 33189905 DOI: 10.1016/j.meegid.2020.104635] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 01/21/2023]
Abstract
Brucellosis is a zoonotic disease caused by certain species of Brucella. Each species has its preferred host animal, though it can infect other animals too. For a longer period, only six classical species were recognized in the genus Brucella. No vaccine is available for human brucellosis. Therefore, human brucellosis can be controlled only by controlling brucellosis in animals. The genus is now expanding with the newly isolated atypical strains from various animals, including marine mammals. Presently, 12 species of Brucella have been recognized. The first genome of Brucella was released in 2002, and today, we have more than 1500 genomes of Brucella spp. isolated worldwide. Multiple genome sequences are available for the major zoonotic species, B. abortus, B. melitensis, and B. suis. The Brucella genome has two chromosomes with the approximate sizes of 2.1 and 1.2 Mbp. The genome of Brucella is highly conserved across all the species at the nucleotide level. One of the unanswered questions is what makes host preference in different species of Brucella. Here, I summarize the recent advancements in the Brucella genomics research.
Collapse
Affiliation(s)
- Jeyaprakash Rajendhran
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamil Nadu, India.
| |
Collapse
|
21
|
Ma Z, Li R, Hu R, Deng X, Xu Y, Zheng W, Yi J, Wang Y, Chen C. Brucella abortus BspJ Is a Nucleomodulin That Inhibits Macrophage Apoptosis and Promotes Intracellular Survival of Brucella. Front Microbiol 2020; 11:599205. [PMID: 33281799 PMCID: PMC7688787 DOI: 10.3389/fmicb.2020.599205] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/23/2020] [Indexed: 12/11/2022] Open
Abstract
To date, a variety of Brucella effector proteins have been found to mediate host cell secretion, autophagy, inflammation, and other signal pathways, but nuclear effector proteins have not yet been reported. We identified the first Brucella nucleomodulin, BspJ, and we screened out the BspJ interaction host proteins NME/NM23 nucleoside diphosphate kinase 2 (NME2) and creatine kinase B (CKB) through yeast two-hybrid and co-immunoprecipitation assays. These proteins are related to the host cell energy synthesis, metabolism, and apoptosis pathways. Brucella nucleomodulin BspJ will decrease the expression level of NME2 and CKB. In addition, BspJ gene deletion strains promoted the apoptosis of macrophages and reduced the intracellular survival of Brucella in host cells. In short, we found nucleomodulin BspJ may directly or indirectly regulate host cell apoptosis through the interaction with NME2 and CKB by mediating energy metabolism pathways in response to the intracellular circulation of Brucella infection, but the mechanism needs further study.
Collapse
Affiliation(s)
- Zhongchen Ma
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Control and Prevention of Animal Disease, Xinjiang Production & Construction Corps, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Ruirui Li
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Control and Prevention of Animal Disease, Xinjiang Production & Construction Corps, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Ruirui Hu
- College of Life Science, Shihezi University, Shihezi, China
| | - Xiaoyu Deng
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Control and Prevention of Animal Disease, Xinjiang Production & Construction Corps, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Yimei Xu
- Xinjiang Center for Disease Control and Prevention, Urumqi, China
| | - Wei Zheng
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Control and Prevention of Animal Disease, Xinjiang Production & Construction Corps, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Jihai Yi
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Control and Prevention of Animal Disease, Xinjiang Production & Construction Corps, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Yong Wang
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Control and Prevention of Animal Disease, Xinjiang Production & Construction Corps, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Chuangfu Chen
- International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Control and Prevention of Animal Disease, Xinjiang Production & Construction Corps, College of Animal Science and Technology, Shihezi University, Shihezi, China
| |
Collapse
|
22
|
Abstract
Brucella spp. are Gram negative intracellular bacteria responsible for brucellosis, a worldwide distributed zoonosis. A prominent aspect of the Brucella life cycle is its ability to invade, survive and multiply within host cells. Comprehensive approaches, such as proteomics, have aided in unravelling the molecular mechanisms underlying Brucella pathogenesis. Technological and methodological advancements such as increased instrument performance and multiplexed quantification have broadened the range of proteome studies, enabling new and improved analyses, providing deeper and more accurate proteome coverage. Indeed, proteomics has demonstrated its contribution to key research questions in Brucella biology, i.e., immunodominant proteins, host-cell interaction, stress response, antibiotic targets and resistance, protein secretion. Here, we review the proteomics of Brucella with a focus on more recent works and novel findings, ranging from reconfiguration of the intracellular bacterial proteome and studies on proteomic profiles of Brucella infected tissues, to the identification of Brucella extracellular proteins with putative roles in cell signaling and pathogenesis. In conclusion, proteomics has yielded copious new candidates and hypotheses that require future verification. It is expected that proteomics will continue to be an invaluable tool for Brucella and applications will further extend to the currently ill-explored aspects including, among others, protein processing and post-translational modification.
Collapse
|
23
|
Smith EP, Cotto-Rosario A, Borghesan E, Held K, Miller CN, Celli J. Epistatic Interplay between Type IV Secretion Effectors Engages the Small GTPase Rab2 in the Brucella Intracellular Cycle. mBio 2020; 11:e03350-19. [PMID: 32234817 PMCID: PMC7157780 DOI: 10.1128/mbio.03350-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/06/2020] [Indexed: 11/20/2022] Open
Abstract
Intracellular bacterial pathogens remodel cellular functions during their infectious cycle via the coordinated actions of effector molecules delivered through dedicated secretion systems. While the function of many individual effectors is known, how they interact to promote pathogenesis is rarely understood. The zoonotic bacterium Brucella abortus, the causative agent of brucellosis, delivers effector proteins via its VirB type IV secretion system (T4SS) which mediate biogenesis of the endoplasmic reticulum (ER)-derived replicative Brucella-containing vacuole (rBCV). Here, we show that T4SS effectors BspB and RicA display epistatic interactions in Brucella replication. Defects in rBCV biogenesis and Brucella replication caused by deletion of bspB were dependent on the host GTPase Rab2a and suppressed by the deletion of ricA, indicating a role of Rab2-binding effector RicA in these phenotypic defects. Rab2a requirements for rBCV biogenesis and Brucella intracellular replication were abolished upon deletion of both bspB and ricA, demonstrating that the functional interaction of these effectors engages Rab2-dependent transport in the Brucella intracellular cycle. Expression of RicA impaired host secretion and caused Golgi fragmentation. While BspB-mediated changes in ER-to-Golgi transport were independent of RicA and Rab2a, BspB-driven alterations in Golgi vesicular traffic also involved RicA and Rab2a, defining BspB and RicA's functional interplay at the Golgi interface. Altogether, these findings support a model where RicA modulation of Rab2a functions impairs Brucella replication but is compensated by BspB-mediated remodeling of Golgi apparatus-associated vesicular transport, revealing an epistatic interaction between these T4SS effectors.IMPORTANCE Bacterial pathogens with an intracellular lifestyle modulate many host cellular processes to promote their infectious cycle. They do so by delivering effector proteins into host cells via dedicated secretion systems that target specific host functions. While the roles of many individual effectors are known, how their modes of action are coordinated is rarely understood. Here, we show that the zoonotic bacterium Brucella abortus delivers the BspB effector that mitigates the negative effect on bacterial replication that the RicA effector exerts via modulation of the host small GTPase Rab2. These findings provide an example of functional integration between bacterial effectors that promotes proliferation of pathogens.
Collapse
Affiliation(s)
- Erin P Smith
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA
| | - Alexis Cotto-Rosario
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA
| | - Elizabeth Borghesan
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA
| | - Kiara Held
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA
| | - Cheryl N Miller
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA
| | - Jean Celli
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA
| |
Collapse
|
24
|
Abstract
In this article, we explore the unique adaptations of intracellular bacterial pathogens that manipulate conserved cellular pathways, organelles, and cargo to convert the phagosome into a pathogen-containing vacuole (PCV). The phagosome is a degradative organelle that rapidly acidifies as it delivers cargo to the lysosome to destroy microbes and cellular debris. However, to avoid this fate, intracellular bacterial pathogens hijack the key molecular modulators of intracellular traffic: small GTPases, phospholipids, SNAREs, and their associated effectors. Following uptake, pathogens that reside in the phagosome either remain associated with the endocytic pathway or rapidly diverge from the preprogrammed route to the lysosome. Both groups rely on effector-mediated mechanisms to meet the common challenges of intracellular life, such as nutrient acquisition, vacuole expansion, and evasion of the host immune response. Mycobacteria, Salmonella, and Coxiella serve as a lens through which we explore regulators of the canonical endocytic route and pathogens that seek to subvert it. On the other hand, pathogens such as Chlamydia, Legionella, and Brucella disconnect from the canonical endocytic route. This bifurcation is linked to extensive hijacking of the secretory pathway and repurposing of the PCV into specialized compartments that resemble organelles in the secretory network. Finally, each pathogen devises specific strategies to counteract host immune responses, such as autophagy, which aim to destroy these aberrant organelles. Collectively, each unique intracellular niche and the pathogens that construct them reflect the outcome of an aggressive and ongoing molecular arms race at the host-pathogen interface. Improving our understanding of these well-adapted pathogens can help us refine our knowledge of conserved cell biological processes.
Collapse
|
25
|
Dehio C, Tsolis RM. Type IV Effector Secretion and Subversion of Host Functions by Bartonella and Brucella Species. Curr Top Microbiol Immunol 2019. [PMID: 29536363 DOI: 10.1007/978-3-319-75241-9_11] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2023]
Abstract
Bartonella and Brucella species comprise closely related genera of the order Rhizobiales within the class α-proteobacteria. Both groups of bacteria are mammalian pathogens with a facultative intracellular lifestyle and are capable of causing chronic infections, but members of each genus have evolved broadly different infection and transmission strategies. While Brucella spp. transmit in general via the reproductive tract in their natural hosts, the Bartonella spp. have evolved to transmit via arthropod vectors. However, a shared feature of both groups of pathogens is their reliance on type IV secretion systems (T4SSs) to interact with cells in their mammalian hosts. The genomes of Bartonella spp. encode three types of T4SS, Trw, Vbh/TraG, and VirB/VirD4, whereas those of Brucella spp. uniformly contain a single T4SS of the VirB type. The VirB systems of Bartonella and Brucella are associated with distinct groups of effector proteins that collectively mediate interactions with host cells. This chapter discusses recent findings on the role of T4SS in the biology of Bartonella spp. and Brucella spp. with emphasis on effector repertoires, on recent advances in our understanding of their evolution, how individual effectors function at the molecular level, and on the consequences of these interactions for cellular and immune responses in the host.
Collapse
Affiliation(s)
| | - Renée M Tsolis
- Medical Microbiology and Immunology, University of California at Davis, Davis, CA, 95616, USA.
| |
Collapse
|
26
|
Abstract
Bacteria of the genus Brucella colonize a wide variety of mammalian hosts, in which their infectious cycle and ability to cause disease predominantly rely on an intracellular lifestyle within phagocytes. Upon entry into host cells, Brucella organisms undergo a complex, multistage intracellular cycle in which they sequentially traffic through, and exploit functions of, the endocytic, secretory, and autophagic compartments via type IV secretion system (T4SS)-mediated delivery of bacterial effectors. These effectors modulate an array of host functions and machineries to first promote conversion of the initial endosome-like Brucella-containing vacuole (eBCV) into a replication-permissive organelle derived from the host endoplasmic reticulum (rBCV) and then to an autophagy-related vacuole (aBCV) that mediates bacterial egress. Here we detail and discuss our current knowledge of cellular and molecular events of the Brucella intracellular cycle. We discuss the importance of the endosomal stage in determining T4SS competency, the roles of autophagy in rBCV biogenesis and aBCV formation, and T4SS-driven mechanisms of modulation of host secretory traffic in rBCV biogenesis and bacterial egress.
Collapse
|
27
|
Herrou J, Willett JW, Fiebig A, Varesio LM, Czyż DM, Cheng JX, Ultee E, Briegel A, Bigelow L, Babnigg G, Kim Y, Crosson S. Periplasmic protein EipA determines envelope stress resistance and virulence in Brucella abortus. Mol Microbiol 2019; 111:637-661. [PMID: 30536925 DOI: 10.1111/mmi.14178] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2018] [Indexed: 12/17/2022]
Abstract
Molecular components of the Brucella abortus cell envelope play a major role in its ability to infect, colonize and survive inside mammalian host cells. In this study, we have defined a role for a conserved gene of unknown function in B. abortus envelope stress resistance and infection. Expression of this gene, which we name eipA, is directly activated by the essential cell cycle regulator, CtrA. eipA encodes a soluble periplasmic protein that adopts an unusual eight-stranded β-barrel fold. Deletion of eipA attenuates replication and survival in macrophage and mouse infection models, and results in sensitivity to treatments that compromise the cell envelope integrity. Transposon disruption of genes required for LPS O-polysaccharide biosynthesis is synthetically lethal with eipA deletion. This genetic connection between O-polysaccharide and eipA is corroborated by our discovery that eipA is essential in Brucella ovis, a naturally rough species that harbors mutations in several genes required for O-polysaccharide production. Conditional depletion of eipA expression in B. ovis results in a cell chaining phenotype, providing evidence that eipA directly or indirectly influences cell division in Brucella. We conclude that EipA is a molecular determinant of Brucella virulence that functions to maintain cell envelope integrity and influences cell division.
Collapse
Affiliation(s)
- Julien Herrou
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Jonathan W Willett
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Aretha Fiebig
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Lydia M Varesio
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Daniel M Czyż
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Jason X Cheng
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Eveline Ultee
- Department of Biology, Universiteit Leiden, Leiden, Netherlands
| | - Ariane Briegel
- Department of Biology, Universiteit Leiden, Leiden, Netherlands
| | - Lance Bigelow
- Biosciences Division, Argonne National Laboratory, Argonne, IL, USA
| | - Gyorgy Babnigg
- Biosciences Division, Argonne National Laboratory, Argonne, IL, USA
| | - Youngchang Kim
- Biosciences Division, Argonne National Laboratory, Argonne, IL, USA
| | - Sean Crosson
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
28
|
Li P, Tian M, Hu H, Yin Y, Guan X, Ding C, Wang S, Yu S. Lable-free based comparative proteomic analysis of secretory proteins of rough Brucella mutants. J Proteomics 2019; 195:66-75. [PMID: 30659936 DOI: 10.1016/j.jprot.2019.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/07/2019] [Accepted: 01/13/2019] [Indexed: 01/18/2023]
Abstract
Brucella rough mutants are reported to induce infected macrophage death, which is type IV secretion system (T4SS) dependent. T4SS and its secretory proteins play a major role in host-bacteria interactions, but the crucial secretory proteins to promote macrophage death during Brucella rough mutant infection have not been characterized. In this study, we found that T4SS components played no role for macrophage death induced by Brucella rough mutant infection, but some T4SS effectors did. Proteomics of secretory proteins from Brucella rough mutants ΔrfbE and ΔrfbEΔvirB123 was analyzed by liquid chromatography/tandem mass spectrometry and 861 unique proteins were identified, among which 37 were differential secretory proteins. Gene ontology and pathway analysis showed that differential secretory proteins involved in cellular process and metabolic process, distributed in the cell and membrane, possessed molecular function of catalytic activity and binding, and were associated with ribosome, NOD-like receptor signaling pathway, two-component system and bacterial secretion system. Cell death analysis showed that T4SS effector VceC, and two differential secretory proteins OmpW family protein (BAB1_1579) and protein BAB1_1185 were associated with Brucella cytotoxicity. This study provides new insights into the molecular mechanisms associated with Brucella cytotoxicity and valuable information for screening vaccine candidates for Brucella. SIGNIFICANCE: Brucella rough mutants induce infected macrophage death, which is T4SS dependent. In the present report, a comparative proteomics analysis revealed 37 differential secretory proteins between Brucella rough mutants ΔrfbE and ΔrfbEΔvirB123. Further study demonstrated OmpW family protein (BAB1_1579) and uncharacterized protein BAB1_1185, two differential secretory proteins, were associated with Brucella cytotoxicity. This study provides novel information of the secretory proteins from the Brucella rough mutants and their effects on the Brucella cytotoxicity.
Collapse
Affiliation(s)
- Peng Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, PR China
| | - Mingxing Tian
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, PR China
| | - Hai Hu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, PR China
| | - Yi Yin
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, PR China
| | - Xiang Guan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, PR China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, PR China
| | - Shaohui Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, PR China
| | - Shengqing Yu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, PR China.
| |
Collapse
|
29
|
Martinez E, Siadous FA, Bonazzi M. Tiny architects: biogenesis of intracellular replicative niches by bacterial pathogens. FEMS Microbiol Rev 2018; 42:425-447. [PMID: 29596635 DOI: 10.1093/femsre/fuy013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/26/2018] [Indexed: 11/13/2022] Open
Abstract
Co-evolution of bacterial pathogens with their hosts led to the emergence of a stunning variety of strategies aiming at the evasion of host defences, colonisation of host cells and tissues and, ultimately, the establishment of a successful infection. Pathogenic bacteria are typically classified as extracellular and intracellular; however, intracellular lifestyle comes in many different flavours: some microbes rapidly escape to the cytosol whereas other microbes remain within vacuolar compartments and harness membrane trafficking pathways to generate their host-derived, pathogen-specific replicative niche. Here we review the current knowledge on a variety of vacuolar lifestyles, the effector proteins used by bacteria as tools to take control of the host cell and the main membrane trafficking signalling pathways targeted by vacuolar pathogens as source of membranes and nutrients. Finally, we will also discuss how host cells have developed countermeasures to sense the biogenesis of the aberrant organelles harbouring bacteria. Understanding the dialogue between bacterial and eukaryotic proteins is the key to unravel the molecular mechanisms of infection and in turn, this may lead to the identification of new targets for the development of new antimicrobials.
Collapse
Affiliation(s)
- Eric Martinez
- IRIM, University of Montpellier, CNRS, 34293 Montpellier, France
| | | | - Matteo Bonazzi
- IRIM, University of Montpellier, CNRS, 34293 Montpellier, France
| |
Collapse
|
30
|
Tian M, Lian Z, Bao Y, Bao S, Yin Y, Li P, Ding C, Wang S, Li T, Qi J, Wang X, Yu S. Identification of a novel, small, conserved hypothetical protein involved inBrucella abortusvirulence by modifying the expression of multiple genes. Transbound Emerg Dis 2018; 66:349-362. [DOI: 10.1111/tbed.13028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/05/2018] [Accepted: 09/21/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Mingxing Tian
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences (CAAS) Shanghai China
| | - Zhengmin Lian
- China College of Veterinary Medicine Gansu Agricultural University LanzhouChina
| | - Yanqing Bao
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences (CAAS) Shanghai China
| | - Shijun Bao
- China College of Veterinary Medicine Gansu Agricultural University LanzhouChina
| | - Yi Yin
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences (CAAS) Shanghai China
| | - Peng Li
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences (CAAS) Shanghai China
| | - Chan Ding
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences (CAAS) Shanghai China
| | - Shaohui Wang
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences (CAAS) Shanghai China
| | - Tao Li
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences (CAAS) Shanghai China
| | - Jingjing Qi
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences (CAAS) Shanghai China
| | - Xiaolan Wang
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences (CAAS) Shanghai China
| | - Shengqing Yu
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences (CAAS) Shanghai China
| |
Collapse
|
31
|
Patrick KL, Wojcechowskyj JA, Bell SL, Riba MN, Jing T, Talmage S, Xu P, Cabello AL, Xu J, Shales M, Jimenez-Morales D, Ficht TA, de Figueiredo P, Samuel JE, Li P, Krogan NJ, Watson RO. Quantitative Yeast Genetic Interaction Profiling of Bacterial Effector Proteins Uncovers a Role for the Human Retromer in Salmonella Infection. Cell Syst 2018; 7:323-338.e6. [PMID: 30077634 PMCID: PMC6160342 DOI: 10.1016/j.cels.2018.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/21/2018] [Accepted: 06/21/2018] [Indexed: 11/26/2022]
Abstract
Intracellular bacterial pathogens secrete a repertoire of effector proteins into host cells that are required to hijack cellular pathways and cause disease. Despite decades of research, the molecular functions of most bacterial effectors remain unclear. To address this gap, we generated quantitative genetic interaction profiles between 36 validated and putative effectors from three evolutionarily divergent human bacterial pathogens and 4,190 yeast deletion strains. Correlating effector-generated profiles with those of yeast mutants, we recapitulated known biology for several effectors with remarkable specificity and predicted previously unknown functions for others. Biochemical and functional validation in human cells revealed a role for an uncharacterized component of the Salmonella SPI-2 translocon, SseC, in regulating maintenance of the Salmonella vacuole through interactions with components of the host retromer complex. These results exhibit the power of genetic interaction profiling to discover and dissect complex biology at the host-pathogen interface.
Collapse
Affiliation(s)
- Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA
| | - Jason A Wojcechowskyj
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA 94158, USA; J. David Gladstone Institute, San Francisco, CA 94158, USA
| | - Samantha L Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA
| | - Morgan N Riba
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA
| | - Tao Jing
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Sara Talmage
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA
| | - Pengbiao Xu
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Ana L Cabello
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA; Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA; Norman Borlaug Center, Texas A&M University, College Station, TX 77843, USA
| | - Jiewei Xu
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA 94158, USA
| | - Michael Shales
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA 94158, USA
| | - David Jimenez-Morales
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA 94158, USA; J. David Gladstone Institute, San Francisco, CA 94158, USA
| | - Thomas A Ficht
- Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA
| | - Paul de Figueiredo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA; Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA; Norman Borlaug Center, Texas A&M University, College Station, TX 77843, USA
| | - James E Samuel
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA 94158, USA; J. David Gladstone Institute, San Francisco, CA 94158, USA.
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77802, USA.
| |
Collapse
|
32
|
Mitochondrial fragmentation affects neither the sensitivity to TNFα-induced apoptosis of Brucella-infected cells nor the intracellular replication of the bacteria. Sci Rep 2018; 8:5173. [PMID: 29581535 PMCID: PMC5979954 DOI: 10.1038/s41598-018-23483-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 03/13/2018] [Indexed: 01/18/2023] Open
Abstract
Mitochondria are complex organelles that participate in many cellular functions, ranging from ATP production to immune responses against viruses and bacteria. This integration of a plethora of functions within a single organelle makes mitochondria a very attractive target to manipulate for intracellular pathogens. We characterised the crosstalk that exists between Brucella abortus, the causative agent of brucellosis, and the mitochondria of infected cells. Brucella replicates in a compartment derived from the endoplasmic reticulum (ER) and modulates ER functionality by activating the unfolded protein response. However, the impact of Brucella on the mitochondrial population of infected cells still requires a systematic study. We observed physical contacts between Brucella containing vacuoles and mitochondria. We also found that B. abortus replication is independent of mitochondrial oxidative phosphorylation and that mitochondrial reactive oxygen species do not participate to the control of B. abortus infection in vitro. We demonstrated that B. abortus and B. melitensis induce a drastic mitochondrial fragmentation at 48 hours post-infection in different cell types, including myeloid and non-myeloid cells. This fragmentation is DRP1-independent and might be caused by a deficit of mitochondrial fusion. However, mitochondrial fragmentation does not change neither Brucella replication efficiency, nor the susceptibility of infected cells to TNFα-induced apoptosis.
Collapse
|
33
|
Weber MM, Faris R. Subversion of the Endocytic and Secretory Pathways by Bacterial Effector Proteins. Front Cell Dev Biol 2018; 6:1. [PMID: 29417046 PMCID: PMC5787570 DOI: 10.3389/fcell.2018.00001] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 01/09/2018] [Indexed: 12/22/2022] Open
Abstract
Intracellular bacteria have developed numerous strategies to hijack host vesicular trafficking pathways to form their unique replicative niches. To promote intracellular replication, the bacteria must interact with host organelles and modulate host signaling pathways to acquire nutrients and membrane for the growing parasitophorous vacuole all while suppressing activation of the immune response. To facilitate host cell subversion, bacterial pathogens use specialized secretion systems to deliver bacterial virulence factors, termed effectors, into the host cell that mimic, agonize, and/or antagonize the function of host proteins. In this review we will discuss how bacterial effector proteins from Coxiella burnetii, Brucella abortus, Salmonella enterica serovar Typhimurium, Legionella pneumophila, Chlamydia trachomatis, and Orientia tsutsugamushi manipulate the endocytic and secretory pathways. Understanding how bacterial effector proteins manipulate host processes not only gives us keen insight into bacterial pathogenesis, but also enhances our understanding of how eukaryotic membrane trafficking is regulated.
Collapse
Affiliation(s)
- Mary M Weber
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Robert Faris
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
34
|
Liu X, Zhou M, Yang Y, Wu J, Peng Q. Overexpression of Cu-Zn SOD in Brucella abortus suppresses bacterial intracellular replication via down-regulation of Sar1 activity. Oncotarget 2018. [PMID: 29515756 PMCID: PMC5839387 DOI: 10.18632/oncotarget.24073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Brucella Cu-Zn superoxide dismutase (Cu-Zn SOD) is a periplasmic protein, and immunization of mice with recombinant Cu-Zn SOD protein confers protection against Brucella abortus infection. However, the role of Cu-Zn SOD during the process of Brucella infection remains unknown. Here, we report that Cu-Zn SOD is secreted into culture medium and is translocated into host cells independent of type IV secretion systems (T4SS). Furthermore, co-immunoprecipitation and immunofluorescence studies reveal that Brucella abortus Cu-Zn SOD interacts with the small GTPase Sar1. Overexpression of Cu-Zn SOD in Brucella abortus inhibits bacterial intracellular growth by abolishing Sar1 activity in a manner independent of reactive oxygen species (ROS) production.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Tumor Hospital of Jilin Province, Changchun 130021, China
| | - Mi Zhou
- Changchun Medical College, Changchun 130031, China
| | - Yanling Yang
- Institute of Special Wild Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Jing Wu
- School of Nursing, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Qisheng Peng
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun 130062, China
| |
Collapse
|
35
|
Brucella abortus Promotes a Fibrotic Phenotype in Hepatic Stellate Cells, with Concomitant Activation of the Autophagy Pathway. Infect Immun 2017; 86:IAI.00522-17. [PMID: 28993461 DOI: 10.1128/iai.00522-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/01/2017] [Indexed: 01/18/2023] Open
Abstract
The liver is frequently affected in patients with active brucellosis. The present study demonstrates that Brucella abortus infection induces the activation of the autophagic pathway in hepatic stellate cells to create a microenvironment that promotes a profibrogenic phenotype through the induction of transforming growth factor-β1 (TGF-β1), collagen deposition, and inhibition of matrix metalloproteinase-9 (MMP-9) secretion. Autophagy was revealed by upregulation of the LC3II/LC3I ratio and Beclin-1 expression as well as inhibition of p62 expression in infected cells. The above-described findings were dependent on the type IV secretion system (VirB) and the secreted BPE005 protein, which were partially corroborated using the pharmacological inhibitors wortmannin, a phosphatidyl inositol 3-kinase inhibitor, and leupeptin plus E64 (inhibitors of lysosomal proteases). Activation of the autophagic pathway in hepatic stellate cells during Brucella infection could have an important contribution to attenuating inflammatory hepatic injury by inducing fibrosis. However, with time, B. abortus infection induced Beclin-1 cleavage with concomitant cleavage of caspase-3, indicating the onset of apoptosis of LX-2 cells, as was confirmed by the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay and Hoechst staining. These results demonstrate that the cross talk of LX-2 cells and B. abortus induces autophagy and fibrosis with concomitant apoptosis of LX-2 cells, which may explain some potential mechanisms of liver damage observed in human brucellosis.
Collapse
|
36
|
Miller CN, Smith EP, Cundiff JA, Knodler LA, Bailey Blackburn J, Lupashin V, Celli J. A Brucella Type IV Effector Targets the COG Tethering Complex to Remodel Host Secretory Traffic and Promote Intracellular Replication. Cell Host Microbe 2017; 22:317-329.e7. [PMID: 28844886 PMCID: PMC5599354 DOI: 10.1016/j.chom.2017.07.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 06/16/2017] [Accepted: 07/27/2017] [Indexed: 10/19/2022]
Abstract
Many intracellular pathogens exploit host secretory trafficking to support their intracellular cycle, but knowledge of these pathogenic processes is limited. The bacterium Brucella abortus uses a type IV secretion system (VirB T4SS) to generate a replication-permissive Brucella-containing vacuole (rBCV) derived from the host ER, a process that requires host early secretory trafficking. Here we show that the VirB T4SS effector BspB contributes to rBCV biogenesis and Brucella replication by interacting with the conserved oligomeric Golgi (COG) tethering complex, a major coordinator of Golgi vesicular trafficking, thus remodeling Golgi membrane traffic and redirecting Golgi-derived vesicles to the BCV. Altogether, these findings demonstrate that Brucella modulates COG-dependent trafficking via delivery of a T4SS effector to promote rBCV biogenesis and intracellular proliferation, providing mechanistic insight into how bacterial exploitation of host secretory functions promotes pathogenesis.
Collapse
Affiliation(s)
- Cheryl N Miller
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA 99164, USA
| | - Erin P Smith
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA 99164, USA
| | - Jennifer A Cundiff
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA 99164, USA
| | - Leigh A Knodler
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA 99164, USA
| | - Jessica Bailey Blackburn
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Vladimir Lupashin
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jean Celli
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
37
|
Roset MS, Alefantis TG, DelVecchio VG, Briones G. Iron-dependent reconfiguration of the proteome underlies the intracellular lifestyle of Brucella abortus. Sci Rep 2017; 7:10637. [PMID: 28878308 PMCID: PMC5587712 DOI: 10.1038/s41598-017-11283-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 08/21/2017] [Indexed: 12/24/2022] Open
Abstract
Brucella ssp. is a facultative intracellular pathogen that causes brucellosis, a worldwide zoonosis that affects a wide range of mammals including humans. A critical step for the establishment of a successful Brucella infection is its ability to survive within macrophages. To further understand the mechanisms that Brucella utilizes to adapt to an intracellular lifestyle, a differential proteomic study was performed for the identification of intracellular modulated proteins. Our results demonstrated that at 48 hours post-infection Brucella adjusts its metabolism in order to survive intracellularly by modulating central carbon metabolism. Remarkably, low iron concentration is likely the dominant trigger for reprogramming the protein expression profile. Up-regulation of proteins dedicated to reduce the concentration of reactive oxygen species, protein chaperones that prevent misfolding of proteins, and proteases that degrade toxic protein aggregates, suggest that Brucella protects itself from damage likely due to oxidative burst. This proteomic analysis of B. abortus provides novel insights into the mechanisms utilized by Brucella to establish an intracellular persistent infection and will aid in the development of new control strategies and novel targets for antimicrobial therapy.
Collapse
Affiliation(s)
- M S Roset
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de General San Martín, IIB-INTECH-CONICET, San Martín 1650, Buenos Aires, Argentina.
| | - T G Alefantis
- Vital Probes Inc., 1820 N. E.27th Drive, Wilton Manors, Florida, USA.,Sanofi Pasteur, 1 Discovery Drive, Swiftwater, PA, USA
| | - V G DelVecchio
- Vital Probes Inc., 1820 N. E.27th Drive, Wilton Manors, Florida, USA
| | - G Briones
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de General San Martín, IIB-INTECH-CONICET, San Martín 1650, Buenos Aires, Argentina.
| |
Collapse
|
38
|
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: 1.1] [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
|
39
|
The Enterococcus faecalis virulence factor ElrA interacts with the human Four-and-a-Half LIM Domains Protein 2. Sci Rep 2017; 7:4581. [PMID: 28676674 PMCID: PMC5496941 DOI: 10.1038/s41598-017-04875-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/22/2017] [Indexed: 12/22/2022] Open
Abstract
The commensal bacterium Enterococcus faecalis is a common cause of nosocomial infections worldwide. The increasing prevalence of multi-antibiotic resistant E. faecalis strains reinforces this public health concern. Despite numerous studies highlighting several pathology-related genetic traits, the molecular mechanisms of E. faecalis virulence remain poorly understood. In this work, we studied 23 bacterial proteins that could be considered as virulence factors or involved in the Enterococcus interaction with the host. We systematically tested their interactions with human proteins using the Human ORFeome library, a set of 12,212 human ORFs, in yeast. Among the thousands of tested interactions, one involving the E. faecalis virulence factor ElrA and the human protein FHL2 was evidenced by yeast two-hybrid and biochemically confirmed. Further molecular characterizations allowed defining an FHL2-interacting domain (FID) of ElrA. Deletion of the FID led to an attenuated in vivo phenotype of the mutated strain clearly indicating that this interaction is likely to contribute to the multifactorial virulence of this opportunistic pathogen. Altogether, our results show that FHL2 is the first host cellular protein directly targeted by an E. faecalis virulence factor and that this interaction is involved in Enterococcus pathogenicity.
Collapse
|
40
|
Kleinman CL, Sycz G, Bonomi HR, Rodríguez RM, Zorreguieta A, Sieira R. ChIP-seq analysis of the LuxR-type regulator VjbR reveals novel insights into the Brucella virulence gene expression network. Nucleic Acids Res 2017; 45:5757-5769. [PMID: 28334833 PMCID: PMC5449634 DOI: 10.1093/nar/gkx165] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 01/31/2023] Open
Abstract
LuxR-type transcription factors control diverse physiological functions necessary for bacterial adaptation to environmental changes. In the intracellular pathogen Brucella, the LuxR homolog VjbR has been shown to regulate the expression of virulence factors acting at early stages of the intracellular infection and, directly or indirectly, hundreds of additional genes. However, the precise determination of VjbR direct targets has so far proved elusive. Here, we performed chromatin immunoprecipitation of VjbR followed by next-generation sequencing (ChIP-seq). We detected a large amount of VjbR-binding sites distributed across the Brucella genome and determined a markedly asymmetric binding consensus motif, an unusual feature among LuxR-type regulators. RNA-seq analysis performed under conditions mimicking the eukaryotic intracellular environment revealed that, among all loci associated to VjbR-binding, this regulator directly modulated the expression of only a subset of genes encoding functions consistent with an intracellular adaptation strategy for survival during the initial stages of the host cell infection. Other VjbR-binding events, however, showed to be dissociated from transcription and may require different environmental signals to produce a transcriptional output. Taken together, our results bring new insights into the extent and functionality of LuxR-type-related transcriptional networks.
Collapse
Affiliation(s)
| | - Gabriela Sycz
- Fundación Instituto Leloir, Ciudad de Buenos Aires C1405BWE, Argentina
| | - Hernán R. Bonomi
- Fundación Instituto Leloir, Ciudad de Buenos Aires C1405BWE, Argentina
| | | | | | - Rodrigo Sieira
- Fundación Instituto Leloir, Ciudad de Buenos Aires C1405BWE, Argentina
| |
Collapse
|
41
|
Abstract
Intracellular bacterial pathogens have evolved to exploit the protected niche provided within the boundaries of a eukaryotic host cell. Upon entering a host cell, some bacteria can evade the adaptive immune response of its host and replicate in a relatively nutrient-rich environment devoid of competition from other host flora. Growth within a host cell is not without their hazards, however. Many pathogens enter their hosts through receptor-mediated endocytosis or phagocytosis, two intracellular trafficking pathways that terminate in a highly degradative organelle, the phagolysosome. This usually deadly compartment is maintained at a low pH and contains degradative enzymes and reactive oxygen species, resulting in an environment to which few bacterial species are adapted. Some intracellular pathogens, such as Shigella, Listeria, Francisella, and Rickettsia, escape the phagosome to replicate within the cytosol of the host cell. Bacteria that remain within a vacuole either alter the trafficking of their initial phagosomal compartment or adapt to survive within the harsh environment it will soon become. In this chapter, we focus on the mechanisms by which different vacuolar pathogens either evade lysosomal fusion, as in the case of Mycobacterium and Chlamydia, or allow interaction with lysosomes to varying degrees, such as Brucella and Coxiella, and their specific adaptations to inhabit a replicative niche.
Collapse
|
42
|
Marchesini MI, Morrone Seijo SM, Guaimas FF, Comerci DJ. A T4SS Effector Targets Host Cell Alpha-Enolase Contributing to Brucella abortus Intracellular Lifestyle. Front Cell Infect Microbiol 2016; 6:153. [PMID: 27900285 PMCID: PMC5110553 DOI: 10.3389/fcimb.2016.00153] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/01/2016] [Indexed: 01/18/2023] Open
Abstract
Brucella abortus, the causative agent of bovine brucellosis, invades and replicates within cells inside a membrane-bound compartment known as the Brucella containing vacuole (BCV). After trafficking along the endocytic and secretory pathways, BCVs mature into endoplasmic reticulum-derived compartments permissive for bacterial replication. Brucella Type IV Secretion System (VirB) is a major virulence factor essential for the biogenesis of the replicative organelle. Upon infection, Brucella uses the VirB system to translocate effector proteins from the BCV into the host cell cytoplasm. Although the functions of many translocated proteins remain unknown, some of them have been demonstrated to modulate host cell signaling pathways to favor intracellular survival and replication. BPE123 (BAB2_0123) is a B. abortus VirB-translocated effector protein recently identified by our group whose function is yet unknown. In an attempt to identify host cell proteins interacting with BPE123, a pull-down assay was performed and human alpha-enolase (ENO-1) was identified by LC/MS-MS as a potential interaction partner of BPE123. These results were confirmed by immunoprecipitation assays. In bone-marrow derived macrophages infected with B. abortus, ENO-1 associates to BCVs in a BPE123-dependent manner, indicating that interaction with translocated BPE123 is also occurring during the intracellular phase of the bacterium. Furthermore, ENO-1 depletion by siRNA impaired B. abortus intracellular replication in HeLa cells, confirming a role for α-enolase during the infection process. Indeed, ENO-1 activity levels were enhanced upon B. abortus infection of THP-1 macrophagic cells, and this activation is highly dependent on BPE123. Taken together, these results suggest that interaction between BPE123 and host cell ENO-1 contributes to the intracellular lifestyle of B. abortus.
Collapse
Affiliation(s)
- María I Marchesini
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde," Instituto Tecnológico de Chascomús, CONICET, Universidad Nacional de San Martín Buenos Aires, Argentina
| | - Susana M Morrone Seijo
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde," Instituto Tecnológico de Chascomús, CONICET, Universidad Nacional de San Martín Buenos Aires, Argentina
| | - Francisco F Guaimas
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde," Instituto Tecnológico de Chascomús, CONICET, Universidad Nacional de San Martín Buenos Aires, Argentina
| | - Diego J Comerci
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde," Instituto Tecnológico de Chascomús, CONICET, Universidad Nacional de San MartínBuenos Aires, Argentina; Comisión Nacional de Energía Atómica, Grupo Pecuario, Centro Atómico EzeizaBuenos Aires, Argentina
| |
Collapse
|
43
|
Abstract
Small GTPases of the Rab protein family control intracellular vesicular trafficking to allow their communication and maintenance. It is a common strategy for intracellular bacteria to exploit these pathways to shape their respective niches for survival. The subversion of Rabs for the generation of an intracellular environment favoring the pathogen has been described almost exclusively for intracellular bacteria that reside within bacterial containing vacuoles (BCVs). However, less is known about Rab subversion for bacteria that rupture the BCV to reach the host cytoplasm. Here, we provide recent examples of Rab targeting by both groups of intracellular bacteria with a special focus on Shigella, the causative agent of bacillary dysentery. Shigella recruits Rab11, the hallmark of the perinuclear recycling compartment to in situ formed macropinosomes at the entry foci via the bacterial effector IpgD. This leads to efficient BCV rupture and cytosolic escape. We discuss the concept of diverted recycling through host Rab GTPases that emerges as a novel pathogen strategy.
Collapse
Affiliation(s)
- Noelia López-Montero
- a Institut Pasteur, Research unit "Dynamics of host-pathogen interactions," Paris , France
| | - Jost Enninga
- a Institut Pasteur, Research unit "Dynamics of host-pathogen interactions," Paris , France
| |
Collapse
|
44
|
Miller C, Celli J. Avoidance and Subversion of Eukaryotic Homeostatic Autophagy Mechanisms by Bacterial Pathogens. J Mol Biol 2016; 428:3387-98. [PMID: 27456933 PMCID: PMC5010449 DOI: 10.1016/j.jmb.2016.07.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 07/07/2016] [Accepted: 07/08/2016] [Indexed: 12/31/2022]
Abstract
Autophagy is a conserved lysosomal recycling process, which maintains cellular homeostasis during stress and starvation conditions by degrading and recycling proteins, lipids, and carbohydrates, ultimately increasing nutrient availability in eukaryotes. An additional function of autophagy, termed xenophagy, is to detect, capture, and destroy invading microorganisms, such as viruses, bacteria, and protozoa, providing autophagy with a role in innate immunity. Many intracellular pathogens have, however, developed mechanisms to avoid xenophagy and have evolved strategies to take advantage of select autophagic processes to undergo their intracellular life cycle. This review article will discuss the molecular mechanisms used by the intracellular bacterial pathogens Francisella spp. and Brucella spp. to manipulate components of the autophagic pathway, promoting cytosolic growth in the case of Francisella spp. and facilitating cellular egress and cell-to-cell spread in the case of Brucella spp. These examples highlight how successful, highly infectious bacterial pathogens avoid or subvert host autophagy mechanisms normally employed to maintain eukaryotic homeostasis.
Collapse
Affiliation(s)
- Cheryl Miller
- Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, 99164-7090 WA, USA.
| | - Jean Celli
- Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, 99164-7090 WA, USA.
| |
Collapse
|
45
|
Liu N, Sun C, Cui G, Wei P, Yang L, Sun W, Wang S, Wang L, Peng Q. The Rab1 in host cells modulates Brucella intracellular survival and binds to Brucella DnaK protein. Arch Microbiol 2016; 198:923-31. [PMID: 27286866 DOI: 10.1007/s00203-016-1246-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/18/2016] [Accepted: 05/20/2016] [Indexed: 01/18/2023]
Abstract
The intracellular pathogen Brucella abortus (B. abortus) survives and replicates inside host cells within the Brucella-containing vacuole, in which membrane contains a small GTPase Rab1. Here, we reported that Rab1 mediates B. abortus intracellular growth. Furthermore, B. abortus DnaK was identified to interact with Rab1 using GST pull-down and mass spectrometry analysis. This interaction was confirmed by co-immunoprecipitation and immunofluorescence. Through DnaK-CyaA fusion protein translocation assay and immunofluorescence confocal microscopy, the B. abortus DnaK was proved to be a virB-dependent translocated substrate.
Collapse
Affiliation(s)
- Ning Liu
- Central Laboratory, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Changjiang Sun
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Guimei Cui
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, China
| | - Pan Wei
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, China
| | - Li Yang
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, China
| | - Wanchun Sun
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, China
| | - Shuangxi Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University, Qilu Hospital, Jinan City, 250012, Shandong, China
| | - Lin Wang
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, China.
| | - Qisheng Peng
- Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, China.
| |
Collapse
|
46
|
Yin S, Li M, Rao X, Yao X, Zhong Q, Wang M, Wang J, Peng Y, Tang J, Hu F, Zhao Y. Subtilisin-like protease-1 secreted through type IV secretion system contributes to high virulence of Streptococcus suis 2. Sci Rep 2016; 6:27369. [PMID: 27270879 PMCID: PMC4897608 DOI: 10.1038/srep27369] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 05/18/2016] [Indexed: 11/24/2022] Open
Abstract
Streptococcus suis serotype 2 is an emerging zoonotic pathogen that triggered two outbreaks of streptococcal toxic shock syndrome (STSS) in China. Our previous research demonstrated that a type IV secretion system (T4SS) harbored in the 89K pathogenicity island contributes to the pathogenicity of S. suis 2. In the present study, a shotgun proteomics approach was employed to identify the effectors secreted by T4SS in S. suis 2, and surface-associated subtilisin-like protease-1 (SspA-1) was identified as a potential virulence effector. Western blot analysis and pull-down assay revealed that SspA-1 secretion depends on T4SS. Knockout mutations affecting sspA-1 attenuated S. suis 2 and impaired the pathogen’s ability to trigger inflammatory response in mice. And purified SspA-1 induced the secretion of IL-6, TNF-α, and IL-12p70 in THP-1 cells directly. SspA-1 is the first T4SS virulence effector reported in Gram-positive bacteria. Overall, these findings allow us to gain further insights into the pathogenesis of T4SS and STSS.
Collapse
Affiliation(s)
- Supeng Yin
- Department of Microbiology, Third Military Medical University, Chongqing, China
| | - Ming Li
- Department of Microbiology, Third Military Medical University, Chongqing, China
| | - Xiancai Rao
- Department of Microbiology, Third Military Medical University, Chongqing, China
| | - Xinyue Yao
- Department of Microbiology, Third Military Medical University, Chongqing, China
| | - Qiu Zhong
- Department of Microbiology, Third Military Medical University, Chongqing, China
| | - Min Wang
- Department of Microbiology, Third Military Medical University, Chongqing, China
| | - Jing Wang
- Department of Microbiology, Third Military Medical University, Chongqing, China
| | - Yizhi Peng
- Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiaqi Tang
- PLA Research Institute of Clinical Laboratory Medicine, Nanjing general hospital of Nanjing Military command, Nanjing 210002, China
| | - Fuquan Hu
- Department of Microbiology, Third Military Medical University, Chongqing, China
| | - Yan Zhao
- Department of Microbiology, Third Military Medical University, Chongqing, China
| |
Collapse
|
47
|
Gonzalez-Rivera C, Bhatty M, Christie PJ. Mechanism and Function of Type IV Secretion During Infection of the Human Host. Microbiol Spectr 2016; 4:10.1128/microbiolspec.VMBF-0024-2015. [PMID: 27337453 PMCID: PMC4920089 DOI: 10.1128/microbiolspec.vmbf-0024-2015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Indexed: 02/07/2023] Open
Abstract
Bacterial pathogens employ type IV secretion systems (T4SSs) for various purposes to aid in survival and proliferation in eukaryotic hosts. One large T4SS subfamily, the conjugation systems, confers a selective advantage to the invading pathogen in clinical settings through dissemination of antibiotic resistance genes and virulence traits. Besides their intrinsic importance as principle contributors to the emergence of multiply drug-resistant "superbugs," detailed studies of these highly tractable systems have generated important new insights into the mode of action and architectures of paradigmatic T4SSs as a foundation for future efforts aimed at suppressing T4SS machine function. Over the past decade, extensive work on the second large T4SS subfamily, the effector translocators, has identified a myriad of mechanisms employed by pathogens to subvert, subdue, or bypass cellular processes and signaling pathways of the host cell. An overarching theme in the evolution of many effectors is that of molecular mimicry. These effectors carry domains similar to those of eukaryotic proteins and exert their effects through stealthy interdigitation of cellular pathways, often with the outcome not of inducing irreversible cell damage but rather of reversibly modulating cellular functions. This article summarizes the major developments for the actively studied pathogens with an emphasis on the structural and functional diversity of the T4SSs and the emerging common themes surrounding effector function in the human host.
Collapse
Affiliation(s)
- Christian Gonzalez-Rivera
- Department of Microbiology and Molecular Genetics, University of Texas Medical School at Houston, 6431 Fannin St, Houston, Texas 77030, Phone: 713-500-5440 (P. J. Christie); 713-500-5441 (C. Gonzalez-Rivera, M. Bhatty)
| | - Minny Bhatty
- Department of Microbiology and Molecular Genetics, University of Texas Medical School at Houston, 6431 Fannin St, Houston, Texas 77030, Phone: 713-500-5440 (P. J. Christie); 713-500-5441 (C. Gonzalez-Rivera, M. Bhatty)
| | - Peter J. Christie
- Department of Microbiology and Molecular Genetics, University of Texas Medical School at Houston, 6431 Fannin St, Houston, Texas 77030, Phone: 713-500-5440 (P. J. Christie); 713-500-5441 (C. Gonzalez-Rivera, M. Bhatty)
| |
Collapse
|
48
|
Di Russo Case E, Smith JA, Ficht TA, Samuel JE, de Figueiredo P. Space: A Final Frontier for Vacuolar Pathogens. Traffic 2016; 17:461-74. [PMID: 26842840 PMCID: PMC6048968 DOI: 10.1111/tra.12382] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 12/12/2022]
Abstract
There is a fundamental gap in our understanding of how a eukaryotic cell apportions the limited space within its cell membrane. Upon infection, a cell competes with intracellular pathogens for control of this same precious resource. The struggle between pathogen and host provides us with an opportunity to uncover the mechanisms regulating subcellular space by understanding how pathogens modulate vesicular traffic and membrane fusion events to create a specialized compartment for replication. By comparing several important intracellular pathogens, we review the molecular mechanisms and trafficking pathways that drive two space allocation strategies, the formation of tight and spacious pathogen-containing vacuoles. Additionally, we discuss the potential advantages of each pathogenic lifestyle, the broader implications these lifestyles might have for cellular biology and outline exciting opportunities for future investigation.
Collapse
Affiliation(s)
- Elizabeth Di Russo Case
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, USA
| | - Judith A. Smith
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Thomas A. Ficht
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - James E. Samuel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, USA
| | - Paul de Figueiredo
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
- Norman Borlaug Center, Texas A&M University, College Station, TX, USA
| |
Collapse
|
49
|
Del Giudice MG, Döhmer PH, Spera JM, Laporte FT, Marchesini MI, Czibener C, Ugalde JE. VirJ Is a Brucella Virulence Factor Involved in the Secretion of Type IV Secreted Substrates. J Biol Chem 2016; 291:12383-93. [PMID: 27059960 DOI: 10.1074/jbc.m116.730994] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Indexed: 11/06/2022] Open
Abstract
The VirB secretion apparatus in Brucella belongs to the type IV secretion systems present in many pathogenic bacteria and is absolutely necessary for the efficient evasion of the Brucella-containing vacuole from the phagocytic route in professional phagocytes. This system is responsible for the secretion of a plethora of effector proteins that alter the biology of the host cell and promote the intracellular replication process. Although many VirB substrates have been identified in Brucella, we still know very little about the secretion mechanism that mediates their translocation across the two membranes and the periplasmic space. In this manuscript, we describe the identification of a gene, virJ, that codes for a protein with periplasmic localization that is involved in the intracellular replication process and virulence in mice. Our analysis revealed that this protein is necessary for the secretion of at least two VirB substrates that have a periplasmic intermediate and that it directly interacts with them. We additionally show that VirJ also associates with the apparatus per se and that its absence affects the assembly of the complex. We hypothesize that VirJ is part of a secretion platform composed of the translocon and several secretion substrates and that it probably coordinates the proper assembly of this macromolecular complex.
Collapse
Affiliation(s)
- Mariela Giselda Del Giudice
- From the Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto de Investigaciones Biotecnológicas (IIB-INTECH), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de San Martín, San Martín 1650, Buenos Aires, Argentina
| | - Peter Hans Döhmer
- From the Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto de Investigaciones Biotecnológicas (IIB-INTECH), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de San Martín, San Martín 1650, Buenos Aires, Argentina
| | - Juan Manuel Spera
- From the Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto de Investigaciones Biotecnológicas (IIB-INTECH), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de San Martín, San Martín 1650, Buenos Aires, Argentina
| | - Fernando Tomás Laporte
- From the Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto de Investigaciones Biotecnológicas (IIB-INTECH), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de San Martín, San Martín 1650, Buenos Aires, Argentina
| | - María Inés Marchesini
- From the Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto de Investigaciones Biotecnológicas (IIB-INTECH), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de San Martín, San Martín 1650, Buenos Aires, Argentina
| | - Cecilia Czibener
- From the Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto de Investigaciones Biotecnológicas (IIB-INTECH), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de San Martín, San Martín 1650, Buenos Aires, Argentina
| | - Juan Esteban Ugalde
- From the Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto de Investigaciones Biotecnológicas (IIB-INTECH), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de San Martín, San Martín 1650, Buenos Aires, Argentina
| |
Collapse
|
50
|
Wang X, Lin P, Li Y, Xiang C, Yin Y, Chen Z, Du Y, Zhou D, Jin Y, Wang A. Brucella suis Vaccine Strain 2 Induces Endoplasmic Reticulum Stress that Affects Intracellular Replication in Goat Trophoblast Cells In vitro. Front Cell Infect Microbiol 2016; 6:19. [PMID: 26904517 PMCID: PMC4746994 DOI: 10.3389/fcimb.2016.00019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/25/2016] [Indexed: 01/16/2023] Open
Abstract
Brucella has been reported to impair placental trophoblasts, a cellular target where Brucella efficiently replicates in association with the endoplasmic reticulum (ER), and ultimately trigger abortion in pregnant animals. However, the precise effects of Brucella on trophoblast cells remain unclear. Here, we describe the infection and replication of Brucella suis vaccine strain 2 (B.suis.S2) in goat trophoblast cells (GTCs) and the cellular and molecular responses induced in vitro. Our studies demonstrated that B.suis.S2 was able to infect and proliferate to high titers, hamper the proliferation of GTCs and induce apoptosis due to ER stress. Tunicamycin (Tm), a pharmacological chaperone that strongly mounts ER stress-induced apoptosis, inhibited B.suis.S2 replication in GTCs. In addition, 4 phenyl butyric acid (4-PBA), a pharmacological chaperone that alleviates ER stress-induced apoptosis, significantly enhanced B.suis.S2 replication in GTCs. The Unfolded Protein Response (UPR) chaperone molecule GRP78 also promoted B.suis.S2 proliferation in GTCs by inhibiting ER stress-induced apoptosis. We also discovered that the IRE1 pathway, but not the PERK or ATF6 pathway, was activated in the process. However, decreasing the expression of phosphoIRE1α and IRE1α proteins with Irestatin 9389 (IRE1 antagonist) in GTCs did not affect the proliferation of B.suis.S2. Although GTC implantation was not affected upon B.suis.S2 infection, progesterone secretion was suppressed, and prolactin and estrogen secretion increased; these effects were accompanied by changes in the expression of genes encoding key steroidogenic enzymes. This study systematically explored the mechanisms of abortion in Brucella infection from the viewpoint of pathogen invasion, ER stress and reproductive endocrinology. Our findings may provide new insight for understanding the mechanisms involved in goat abortions caused by Brucella infection.
Collapse
Affiliation(s)
- Xiangguo Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F UniversityYangling, China; College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Pengfei Lin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F UniversityYangling, China; College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Yang Li
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F UniversityYangling, China; College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Caixia Xiang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F UniversityYangling, China; College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Yanlong Yin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F UniversityYangling, China; College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Zhi Chen
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F UniversityYangling, China; College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Yue Du
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F UniversityYangling, China; College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Dong Zhou
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F UniversityYangling, China; College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Yaping Jin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F UniversityYangling, China; College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Aihua Wang
- College of Veterinary Medicine, Northwest A&F University Yangling, China
| |
Collapse
|