1
|
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
|
2
|
Yu H, Gu X, Wang D, Wang Z. Brucella infection and Toll-like receptors. Front Cell Infect Microbiol 2024; 14:1342684. [PMID: 38533384 PMCID: PMC10963510 DOI: 10.3389/fcimb.2024.1342684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/17/2024] [Indexed: 03/28/2024] Open
Abstract
Brucella consists of gram-negative bacteria that have the ability to invade and replicate in professional and non-professional phagocytes, and its prolonged persistence in the host leads to brucellosis, a serious zoonosis. Toll-like receptors (TLRs) are the best-known sensors of microorganisms implicated in the regulation of innate and adaptive immunity. In particular, TLRs are transmembrane proteins with a typical structure of an extracellular leucine-rich repeat (LRR) region and an intracellular Toll/interleukin-1 receptor (TIR) domain. In this review, we discuss Brucella infection and the aspects of host immune responses induced by pathogens. Furthermore, we summarize the roles of TLRs in Brucella infection, with substantial emphasis on the molecular insights into its mechanisms of action.
Collapse
Affiliation(s)
- Hui Yu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Xinyi Gu
- The College of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Danfeng Wang
- The College of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| |
Collapse
|
3
|
Yang J, Wang Y, Hou Y, Sun M, Xia T, Wu X. Evasion of host defense by Brucella. CELL INSIGHT 2024; 3:100143. [PMID: 38250017 PMCID: PMC10797155 DOI: 10.1016/j.cellin.2023.100143] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024]
Abstract
Brucella , an adept intracellular pathogen, causes brucellosis, a zoonotic disease leading to significant global impacts on animal welfare and the economy. Regrettably, there is currently no approved and effective vaccine for human use. The ability of Brucella to evade host defenses is essential for establishing chronic infection and ensuring stable intracellular growth. Brucella employs various mechanisms to evade and undermine the innate and adaptive immune responses of the host through modulating the activation of pattern recognition receptors (PRRs), inflammatory responses, or the activation of immune cells like dendritic cells (DCs) to inhibit antigen presentation. Moreover, it regulates multiple cellular processes such as apoptosis, pyroptosis, and autophagy to establish persistent infection within host cells. This review summarizes the recently discovered mechanisms employed by Brucella to subvert host immune responses and research progress on vaccines, with the aim of advancing our understanding of brucellosis and facilitating the development of more effective vaccines and therapeutic approaches against Brucella .
Collapse
Affiliation(s)
- Jinke Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yue Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yuanpan Hou
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Mengyao Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Tian Xia
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Xin Wu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| |
Collapse
|
4
|
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
|
5
|
Murugan S, Nandi BR, Mazumdar V, Joshi K, Nandini P, Namani S, Jakka P, Radhakrishnan GK. Outer membrane protein 25 of Brucella suppresses TLR-mediated expression of proinflammatory cytokines through degradation of TLRs and adaptor proteins. J Biol Chem 2023; 299:105309. [PMID: 37778729 PMCID: PMC10641269 DOI: 10.1016/j.jbc.2023.105309] [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: 02/08/2023] [Revised: 08/30/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023] Open
Abstract
Toll-like receptors (TLRs) are essential components of innate immunity that serves as the first line of defense against the invaded microorganisms. However, successful infectious pathogens subvert TLR signaling to suppress the activation of innate and adaptive responses. Brucella species are infectious intracellular bacterial pathogens causing the worldwide zoonotic disease, brucellosis, that impacts economic growth of many countries. Brucella species are considered as stealthy bacterial pathogens as they efficiently evade or suppress host innate and adaptive immune responses for their chronic persistence. However, the bacterial effectors and their host targets for modulating the immune responses remain obscure. Brucella encodes various outer membrane proteins (Omps) that facilitate their invasion, intracellular replication, and immunomodulation. Outer membrane protein 25 (Omp25) of Brucella plays an important role in the immune modulation through suppression of proinflammatory cytokines. However, the mechanism and the signaling pathways that are targeted by Omp25 to attenuate the production of proinflammatory cytokines remain obscure. Here, we report that Omp25 and its variants, viz. Omp25b, Omp25c, and Omp25d, suppress production of proinflammatory cytokines that are mediated by various TLRs. Furthermore, we demonstrate that Omp25 and its variants promote enhanced ubiquitination and degradation of TLRs and their adaptor proteins to attenuate the expression of proinflammatory cytokines. Targeting multiple TLRs and adaptor proteins enables Omp25 to effectively suppress the expression of proinflammatory cytokines that are induced by diverse pathogen-associated molecular patterns. This can contribute to the defective adaptive immune response and the chronic persistence of Brucella in the host.
Collapse
Affiliation(s)
- Subathra Murugan
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana, India; Centre for Biotechnology, Institute of Science and Technology, Jawaharlal Nehru Technological University, Hyderabad, India
| | - Binita Roy Nandi
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana, India; Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Varadendra Mazumdar
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana, India; Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Kiranmai Joshi
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana, India; Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Prachita Nandini
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana, India; Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Swapna Namani
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana, India
| | - Padmaja Jakka
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana, India
| | - Girish K Radhakrishnan
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana, India.
| |
Collapse
|
6
|
Li C, Wang J, Sun W, Liu X, Wang J, Peng Q. The Brucella Effector BspI Suppresses Inflammation via Inhibition of IRE1 Kinase Activity during Brucella Infection. THE JOURNAL OF IMMUNOLOGY 2022; 209:488-497. [DOI: 10.4049/jimmunol.2200001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/26/2022] [Indexed: 01/04/2023]
Abstract
Abstract
Mammalian GTPase-activating proteins (GAPs) can inhibit innate immunity signaling in a spatiotemporal fashion; however, the role of bacterial GAPs in mediating innate immunity remains unknown. In this study, we show that BspI, a Brucella type IV secretion system (T4SS) effector protein, containing a GAP domain at the C terminus, negatively regulates proinflammatory responses and host protection to Brucella abotus infection in a mouse model. In macrophages, BspI inhibits the activation of inositol-requiring enzyme 1 (IRE1) kinase, but it does not inhibit activation of ATF6 and PERK. BspI suppresses induction of proinflammatory cytokines via inhibiting the activity of IRE1 kinase caused by VceC, a type IV secretion system effector protein that localizes to the endoplasmic reticulum. Ectopically expressed BspI interacts with IRE1 in HeLa cells. The inhibitory function of BspI depends on its GAP domain but not on interaction with small GTPase Ras-associated binding protein 1B (RAB1B). Collectively, these data support a model where BspI, in a GAP domain–dependent manner, inhibits activation of IRE1 to prevent proinflammatory cytokine responses.
Collapse
Affiliation(s)
- Chen Li
- *Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, China
| | - Jingyu Wang
- *Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, China
| | - Wanchun Sun
- *Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xiaofeng Liu
- †Tumor Hospital of Jilin Province, Changchun, China; and
| | - Jun Wang
- §Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Qisheng Peng
- *Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, China
| |
Collapse
|
7
|
Ihsan Rashan A, Mahdi Rheima A, Ghadhanfar Alwan M, Abed Jawad M, Mohammed HT, Gaber Abdel Razzaq M, Ahmed Al-Tawee A, Attia Thijail H, Ahjel S, Jalil Obaid A. Evaluating the Level of Serum IL-23 in Brucellosis Infection by ELISA and Investigating its Relationship in Cases with Failure to Respond to Treatment. ARCHIVES OF RAZI INSTITUTE 2022; 77:1275-1279. [PMID: 36618293 PMCID: PMC9759221 DOI: 10.22092/ari.2022.358877.2324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 05/31/2022] [Indexed: 01/10/2023]
Abstract
Brucella is belonging to the small immobile gram-negative spore-lacking cocco-bacilli bacteria family that grows in an aerobic environment, it is known as a zoonosis infection named brucellosis. This study was designed to investigate serum values of IL-23 in patient with brucellosis and investigate its relationship with cases with failure to respond to conventional medical therapy. A total of 372 individuals were divided into 2 groups (n=186) as follows: Group A comprising 186 infected participants with brucella (7-80 years-old), these people had not received antibiotics for at least 6 months ago. Group B including the healthy participants. All the participants in both groups were in the same age range. 5 ml blood samples were obtained from the participants intravenously (without anticoagulation substance). The serum level of IL-23 was investigated by ELISA diagnostic kit. The recorded data showed that the levels of IL-23 in the serum samples obtained from group A (143.64 Pg/ml) significantly (P<0.001) increased compared with this value in group B (23.14 Pg/ml). Based on the recorded data in the forms completed by all the participants at the day 0 of the experiment, 44 out of 186 individuals in group A, had experienced Brucellosis attack 2-3 times in spite of receiving medical prescriptions. A hypothesis about the possible immune system disorders in these participants lead us to did the re-sampling following drug administration. Results illustrated failure to respond to conventional medical therapy in patients with low level of serum IL-23.
Collapse
Affiliation(s)
- A Ihsan Rashan
- Department of Pharmacy, Al-Hadba University College, Mosul, Iraq
| | - A Mahdi Rheima
- College of technical engineering, The Islamic University, Najaf, Iraq,
Department of Optics Techniques, Dijlah University College, Al-Masafi Street, Al-Dora, Baghdad 00964, Iraq
| | - M Ghadhanfar Alwan
- Medical laboratory techniques Department, Medical (Technology) College, Al-Farahidi University, Baghdad, Iraq
| | | | - H. T Mohammed
- Anesthesia Techniques Department, Al-Mustaqbal University College, Babylon, Iraq
| | | | | | - H Attia Thijail
- College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | - S Ahjel
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - A Jalil Obaid
- Medical Laboratory Techniques Department, Hilla University College, Babylon, Iraq
| |
Collapse
|
8
|
Pellegrini JM, Gorvel JP, Mémet S. Immunosuppressive Mechanisms in Brucellosis in Light of Chronic Bacterial Diseases. Microorganisms 2022; 10:1260. [PMID: 35888979 PMCID: PMC9324529 DOI: 10.3390/microorganisms10071260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 01/27/2023] Open
Abstract
Brucellosis is considered one of the major zoonoses worldwide, constituting a critical livestock and human health concern with a huge socio-economic burden. Brucella genus, its etiologic agent, is composed of intracellular bacteria that have evolved a prodigious ability to elude and shape host immunity to establish chronic infection. Brucella's intracellular lifestyle and pathogen-associated molecular patterns, such as its specific lipopolysaccharide (LPS), are key factors for hiding and hampering recognition by the immune system. Here, we will review the current knowledge of evading and immunosuppressive mechanisms elicited by Brucella species to persist stealthily in their hosts, such as those triggered by their LPS and cyclic β-1,2-d-glucan or involved in neutrophil and monocyte avoidance, antigen presentation impairment, the modulation of T cell responses and immunometabolism. Attractive strategies exploited by other successful chronic pathogenic bacteria, including Mycobacteria, Salmonella, and Chlamydia, will be also discussed, with a special emphasis on the mechanisms operating in brucellosis, such as granuloma formation, pyroptosis, and manipulation of type I and III IFNs, B cells, innate lymphoid cells, and host lipids. A better understanding of these stratagems is essential to fighting bacterial chronic infections and designing innovative treatments and vaccines.
Collapse
|
9
|
Mazumdar V, Joshi K, Nandi BR, Namani S, Gupta VK, Radhakrishnan G. Host F-Box Protein 22 Enhances the Uptake of Brucella by Macrophages and Drives a Sustained Release of Proinflammatory Cytokines through Degradation of the Anti-Inflammatory Effector Proteins of Brucella. Infect Immun 2022; 90:e0006022. [PMID: 35420446 PMCID: PMC9119127 DOI: 10.1128/iai.00060-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 11/20/2022] Open
Abstract
Brucella species are intracellular bacterial pathogens, causing the worldwide zoonotic disease brucellosis. Brucella invades professional and nonprofessional phagocytic cells, followed by resisting intracellular killing and establishing a replication permissive niche. Brucella also modulates the innate and adaptive immune responses of the host for its chronic persistence. The complex intracellular cycle of Brucella depends in a major way on multiple host factors, but limited information is available on host and bacterial proteins that play an essential role in the invasion, intracellular replication, and modulation of host immune responses. By employing a small interfering RNA (siRNA) screening, we identified a role for the host protein FBXO22 in the Brucella-macrophage interaction. FBXO22 is the key element in the SCF E3 ubiquitination complex, where it determines the substrate specificity for ubiquitination and degradation of various host proteins. Downregulation of FBXO22 by siRNA or the CRISPR-Cas9 system resulted in diminished uptake of Brucella into macrophages, which was dependent on NF-κB-mediated regulation of phagocytic receptors. FBXO22 expression was upregulated in Brucella-infected macrophages, which resulted in induction of phagocytic receptors and enhanced production of proinflammatory cytokines through NF-κB. Furthermore, we found that FBXO22 recruits the effector proteins of Brucella, including the anti-inflammatory proteins TcpB and OMP25, for degradation through the SCF complex. We did not observe any role for another F-box-containing protein of the SCF complex, β-TrCP, in the Brucella-macrophage interaction. Our findings unravel novel functions of FBXO22 in host-pathogen interaction and its contribution to pathogenesis of infectious diseases.
Collapse
Affiliation(s)
- Varadendra Mazumdar
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Kiranmai Joshi
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Binita Roy Nandi
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Swapna Namani
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| | - Vivek Kumar Gupta
- ICAR-Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar, Bareilly, India
| | - Girish Radhakrishnan
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| |
Collapse
|
10
|
Li J, Zhang G, Zhi F, Zhai Y, Zhou D, Chen H, Lin P, Tang K, Liu W, Jin Y, Wang A. BtpB inhibits innate inflammatory responses in goat alveolar macrophages through the TLR/NF-κB pathway and NLRP3 inflammasome during Brucella infection. Microb Pathog 2022; 166:105536. [DOI: 10.1016/j.micpath.2022.105536] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/15/2022] [Accepted: 04/10/2022] [Indexed: 01/19/2023]
|
11
|
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
|
12
|
Nimma S, Gu W, Maruta N, Li Y, Pan M, Saikot FK, Lim BYJ, McGuinness HY, Zaoti ZF, Li S, Desa S, Manik MK, Nanson JD, Kobe B. Structural Evolution of TIR-Domain Signalosomes. Front Immunol 2021; 12:784484. [PMID: 34868065 PMCID: PMC8635717 DOI: 10.3389/fimmu.2021.784484] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/25/2021] [Indexed: 01/23/2023] Open
Abstract
TIR (Toll/interleukin-1 receptor/resistance protein) domains are cytoplasmic domains widely found in animals and plants, where they are essential components of the innate immune system. A key feature of TIR-domain function in signaling is weak and transient self-association and association with other TIR domains. An additional new role of TIR domains as catalytic enzymes has been established with the recent discovery of NAD+-nucleosidase activity by several TIR domains, mostly involved in cell-death pathways. Although self-association of TIR domains is necessary in both cases, the functional specificity of TIR domains is related in part to the nature of the TIR : TIR interactions in the respective signalosomes. Here, we review the well-studied TIR domain-containing proteins involved in eukaryotic immunity, focusing on the structures, interactions and their corresponding functional roles. Structurally, the signalosomes fall into two separate groups, the scaffold and enzyme TIR-domain assemblies, both of which feature open-ended complexes with two strands of TIR domains, but differ in the orientation of the two strands. We compare and contrast how TIR domains assemble and signal through distinct scaffolding and enzymatic roles, ultimately leading to distinct cellular innate-immunity and cell-death outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
13
|
Ma Z, Yu S, Cheng K, Miao Y, Xu Y, Hu R, Zheng W, Yi J, Zhang H, Li R, Li Z, Wang Y, Chen C. Nucleomodulin BspJ as an effector promotes the colonization of Brucella abortus in the host. J Vet Sci 2021; 23:e8. [PMID: 34841746 PMCID: PMC8799945 DOI: 10.4142/jvs.21224] [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: 08/09/2021] [Revised: 09/21/2021] [Accepted: 10/04/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Brucella infection induces brucellosis, a zoonotic disease. The intracellular circulation process and virulence of Brucella mainly depend on its type IV secretion system (T4SS) expressing secretory effectors. Secreted protein BspJ is a nucleomodulin of Brucella that invades the host cell nucleus. BspJ mediates host energy synthesis and apoptosis through interaction with proteins. However, the mechanism of BspJ as it affects the intracellular survival of Brucella remains to be clarified. OBJECTIVES To verify the functions of nucleomodulin BspJ in Brucella's intracellular infection cycles. METHODS Constructed Brucella abortus BspJ gene deletion strain (B. abortus ΔBspJ) and complement strain (B. abortus pBspJ) and studied their roles in the proliferation of Brucella both in vivo and in vitro. RESULTS BspJ gene deletion reduced the survival and intracellular proliferation of Brucella at the replicating Brucella-containing vacuoles (rBCV) stage. Compared with the parent strain, the colonization ability of the bacteria in mice was significantly reduced, causing less inflammatory infiltration and pathological damage. We also found that the knockout of BspJ altered the secretion of cytokines (interleukin [IL]-6, IL-1β, IL-10, tumor necrosis factor-α, interferon-γ) in host cells and in mice to affect the intracellular survival of Brucella. CONCLUSIONS BspJ is extremely important for the circulatory proliferation of Brucella in the host, and it may be involved in a previously unknown mechanism of Brucella's intracellular survival.
Collapse
Affiliation(s)
- Zhongchen Ma
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Shuifa Yu
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Kejian Cheng
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Yuhe Miao
- Fujian Sunvet Biological Technology Co., Ltd, Nanping 354100, Fujian, China
| | - Yimei Xu
- Xinjiang Center for Disease Control and Prevention, Urumqi 830002, Xinjiang, China
| | - Ruirui Hu
- College of Life Sciences, Shihezi University, Shihezi 832003, Xinjiang, China
| | - Wei Zheng
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Jihai Yi
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Huan Zhang
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Ruirui Li
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Zhiqiang Li
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan, China
| | - Yong Wang
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China.
| | - Chuangfu Chen
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China.
| |
Collapse
|
14
|
Undercover Agents of Infection: The Stealth Strategies of T4SS-Equipped Bacterial Pathogens. Toxins (Basel) 2021; 13:toxins13100713. [PMID: 34679006 PMCID: PMC8539587 DOI: 10.3390/toxins13100713] [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: 07/29/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 11/16/2022] Open
Abstract
Intracellular bacterial pathogens establish their replicative niches within membrane-encompassed compartments, called vacuoles. A subset of these bacteria uses a nanochannel called the type 4 secretion system (T4SS) to inject effector proteins that subvert the host cell machinery and drive the biogenesis of these compartments. These bacteria have also developed sophisticated ways of altering the innate immune sensing and response of their host cells, which allow them to cause long-lasting infections and chronic diseases. This review covers the mechanisms employed by intravacuolar pathogens to escape innate immune sensing and how Type 4-secreted bacterial effectors manipulate host cell mechanisms to allow the persistence of bacteria.
Collapse
|
15
|
Mirzaei R, Sholeh M, Jalalifar S, Zafari E, Kazemi S, Rasouli-Saravani A, Karampoor S, Yousefimashouf R. Immunometabolism in human brucellosis: An emerging field of investigation. Microb Pathog 2021; 158:105115. [PMID: 34332069 DOI: 10.1016/j.micpath.2021.105115] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 01/16/2023]
Abstract
In recent years, extreme attention has been focused on the role of immunometabolism in the regulation of immune cell responses in healthy individuals during infection, autoimmunity, and cancer. In the infection biology area, it has been shown that there is a close relationship between the immune system and the host metabolic changes. Brucella species is an intracellular coccobacillus that infects humans and mammals, which led to brucellosis. Brucella species with host-specific evolutionary mechanisms allow it to hide from or manipulate cellular immunity and achieve intracellular persistence. Intracellular bacterial pathogens such as Brucella species also employ host cell resources to replicate and persist inside the host. Targeting these host systems is one promising strategy for developing novel antimicrobials to tackle intracellular infections. This study will summarize the role of metabolic reprogramming in immune cells and their relationship to brucellosis.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Mohammad Sholeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saba Jalalifar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Zafari
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sima Kazemi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ashkan Rasouli-Saravani
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
16
|
Rajpoot S, Wary KK, Ibbott R, Liu D, Saqib U, Thurston TLM, Baig MS. TIRAP in the Mechanism of Inflammation. Front Immunol 2021; 12:697588. [PMID: 34305934 PMCID: PMC8297548 DOI: 10.3389/fimmu.2021.697588] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022] Open
Abstract
The Toll-interleukin-1 Receptor (TIR) domain-containing adaptor protein (TIRAP) represents a key intracellular signalling molecule regulating diverse immune responses. Its capacity to function as an adaptor molecule has been widely investigated in relation to Toll-like Receptor (TLR)-mediated innate immune signalling. Since the discovery of TIRAP in 2001, initial studies were mainly focused on its role as an adaptor protein that couples Myeloid differentiation factor 88 (MyD88) with TLRs, to activate MyD88-dependent TLRs signalling. Subsequent studies delineated TIRAP’s role as a transducer of signalling events through its interaction with non-TLR signalling mediators. Indeed, the ability of TIRAP to interact with an array of intracellular signalling mediators suggests its central role in various immune responses. Therefore, continued studies that elucidate the molecular basis of various TIRAP-protein interactions and how they affect the signalling magnitude, should provide key information on the inflammatory disease mechanisms. This review summarizes the TIRAP recruitment to activated receptors and discusses the mechanism of interactions in relation to the signalling that precede acute and chronic inflammatory diseases. Furthermore, we highlighted the significance of TIRAP-TIR domain containing binding sites for several intracellular inflammatory signalling molecules. Collectively, we discuss the importance of the TIR domain in TIRAP as a key interface involved in protein interactions which could hence serve as a therapeutic target to dampen the extent of acute and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Sajjan Rajpoot
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Kishore K Wary
- Department of Pharmacology and Regenerative Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| | - Rachel Ibbott
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, United States.,School of Graduate Studies, Rutgers Biomedical and Health Sciences, Newark, NJ, United States.,Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Uzma Saqib
- Discipline of Chemistry, Indian Institute of Technology Indore (IITI), Indore, India
| | - Teresa L M Thurston
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| |
Collapse
|
17
|
Yuan S, Fang Y, Tang M, Hu Z, Rao C, Chen J, Xia Y, Zhang M, Yan J, Tang B, He X, Xie J, Mao X, Li Q. Tauroursodeoxycholic acid prevents Burkholderia pseudomallei-induced endoplasmic reticulum stress and is protective during melioidosis in mice. BMC Microbiol 2021; 21:137. [PMID: 33947331 PMCID: PMC8094575 DOI: 10.1186/s12866-021-02199-x] [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: 10/12/2020] [Accepted: 04/19/2021] [Indexed: 11/24/2022] Open
Abstract
Background Burkholderia pseudomallei, a facultative intracellular bacterium, is the aetiological agent of melioidosis that is responsible for up to 40% sepsis-related mortality in epidemic areas. However, no effective vaccine is available currently, and the drug resistance is also a major problem in the treatment of melioidosis. Therefore, finding new clinical treatment strategies in melioidosis is extremely urgent. Results We demonstrated that tauroursodeoxycholic acid (TUDCA), a clinically available endoplasmic reticulum (ER) stress inhibitor, can promote B. pseudomallei clearance both in vivo and in vitro. In this study, we investigated the effects of TUDCA on the survival of melioidosis mice, and found that treatment with TUDCA significantly decreased intracellular survival of B. pseudomallei. Mechanistically, we found that B. pseudomallei induced apoptosis and activated IRE1 and PERK signaling ways of ER stress in RAW264.7 macrophages. TUDCA treatment could reduce B. pseudomallei-induced ER stress in vitro, and TUDCA is protective in vivo. Conclusion Taken together, our study has demonstrated that B. pseudomallei infection results in ER stress-induced apoptosis, and TUDCA enhances the clearance of B. pseudomallei by inhibiting ER stress-induced apoptosis both in vivo and in vitro, suggesting that TUDCA could be used as a potentially alternative treatment for melioidosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-021-02199-x.
Collapse
Affiliation(s)
- Siqi Yuan
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, 400715, China
| | - Yao Fang
- Department of Respiratory, General Hospital of Center Theater Command, Wuhan, 400070, China
| | - Mengling Tang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, 400715, China
| | - Zhiqiang Hu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Chenglong Rao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jiangao Chen
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,Department of General Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yupei Xia
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Meijuan Zhang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jingmin Yan
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Bin Tang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaoyi He
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Beibei, Chongqing, 400715, China
| | - Xuhu Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qian Li
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
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
|
Ma Z, Yu S, Cheng K, Miao Y, Xu Y, Hu R, Zheng W, Yi J, Zhang H, Li R, Li Z, Wang Y, Chen C. Nucleomodulin BspJ as an effector promotes the colonization of Brucella abortus in the host. J Vet Sci 2021. [DOI: 10.4142/jvs.2021.22.e94] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Zhongchen Ma
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Shuifa Yu
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Kejian Cheng
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Yuhe Miao
- Fujian Sunvet Biological Technology Co., Ltd, Nanping 354100, Fujian, China
| | - Yimei Xu
- Xinjiang Center for Disease Control and Prevention, Urumqi 830002, Xinjiang, China
| | - Ruirui Hu
- College of Life Sciences, Shihezi University, Shihezi 832003, Xinjiang, China
| | - Wei Zheng
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Jihai Yi
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Huan Zhang
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Ruirui Li
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Zhiqiang Li
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan, China
| | - Yong Wang
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| | - Chuangfu Chen
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, 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 832003, Xinjiang, China
| |
Collapse
|
20
|
Xiong D, Song L, Geng S, Jiao Y, Zhou X, Song H, Kang X, Zhou Y, Xu X, Sun J, Pan Z, Jiao X. Salmonella Coiled-Coil- and TIR-Containing TcpS Evades the Innate Immune System and Subdues Inflammation. Cell Rep 2020; 28:804-818.e7. [PMID: 31315056 DOI: 10.1016/j.celrep.2019.06.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/23/2019] [Accepted: 06/13/2019] [Indexed: 01/12/2023] Open
Abstract
Toll-like receptors (TLRs) activate innate immunity via interactions between their Toll/interleukin-1 (IL-1) receptor (TIR) domain and downstream adaptor proteins. Here we report that Salmonella Enteritidis produces a secreted protein (TcpS) that contains both a TIR domain and a coiled-coil domain. TcpS blocks MyD88- and TRIF-mediated TLR signaling, inhibits inflammatory responses, and promotes bacterial survival. Early-stage immune evasion by TcpS results in severe tissue damage in the late stage of infection and contributes to Salmonella virulence. TcpS-derived peptides inhibit nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) activation and reduce lipopolysaccharide (LPS)-elicited systemic inflammation. Therapeutic peptide administration alleviates weight loss of mice infected with H1N1 influenza. Importantly, maximal TcpS-mediated TLR inhibition requires the critical TIR-TcpS residues Y191 and I284, as well as TcpS homodimerization via its N-terminal coiled-coil domain. Our study unveils a mechanism in which TcpS suppresses innate immunity via both its homodimerization and interaction with MyD88. TcpS is also a potential therapeutic agent for inflammation-associated diseases.
Collapse
Affiliation(s)
- Dan Xiong
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Li Song
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Shizhong Geng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yang Jiao
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xiaohui Zhou
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06269, USA
| | - Hongqin Song
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xilong Kang
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yi Zhou
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xiulong Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Jun Sun
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
21
|
Gómez LA, Alvarez FI, Molina RE, Soto-Shara R, Daza-Castro C, Flores MR, León Y, Oñate AA. A Zinc-Dependent Metalloproteinase of Brucella abortus Is Required in the Intracellular Adaptation of Macrophages. Front Microbiol 2020; 11:1586. [PMID: 32765455 PMCID: PMC7379133 DOI: 10.3389/fmicb.2020.01586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/17/2020] [Indexed: 01/19/2023] Open
Abstract
Brucella abortus is a pathogen that survives in macrophages. Several virulence factors participate in this process, including the open reading frame (ORF) BAB1_0270 codifying for a zinc-dependent metalloproteinase (ZnMP). Here, its contribution in the intracellular adaptation of B. abortus was analyzed by infecting RAW264.7 macrophages with the mutant B. abortus Δ270 strain. Results showed that this ZnMP did not participated in either the adherence or the initial intracellular traffic of B. abortus in macrophages. Nevertheless, its deletion significantly increased the co-localization of B. abortus Δ270 with phagolysosomal cathepsin D and reduced its co-localization with calnexin present in endoplasmic reticulum (RE)-derived vesicles. Although B. abortus Δ270 showed an upregulated expression of genes involved in virulence (vjbR, hutC, bvrR, virB1), it was insufficient to reach a successful intracellular replication within macrophages. Furthermore, its attenuation favored in macrophages infected the production of high levels of cytokines (TNF-α and IL-6) and co-stimulatory proteins (CD80 and CD86), signals required in T cell activation. Finally, its deletion significantly reduced the ability of B. abortus Δ270 to adapt, grow and express several virulence factors under acidic conditions. Based on these results, and considering that this ZnMP has homology with ImmA/IrrE proteases, we discuss its role in the virulence of this pathogen, concluding that ZnMP is required in the intracellular adaptation of B. abortus 2308 during the infection of macrophages.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Angel A. Oñate
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| |
Collapse
|
22
|
The TIR-domain containing effectors BtpA and BtpB from Brucella abortus impact NAD metabolism. PLoS Pathog 2020; 16:e1007979. [PMID: 32298382 PMCID: PMC7188309 DOI: 10.1371/journal.ppat.1007979] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 04/28/2020] [Accepted: 03/26/2020] [Indexed: 12/20/2022] Open
Abstract
Brucella species are facultative intracellular Gram-negative bacteria relevant to animal and human health. Their ability to establish an intracellular niche and subvert host cell pathways to their advantage depends on the delivery of bacterial effector proteins through a type IV secretion system. Brucella Toll/Interleukin-1 Receptor (TIR)-domain-containing proteins BtpA (also known as TcpB) and BtpB are among such effectors. Although divergent in primary sequence, they interfere with Toll-like receptor (TLR) signaling to inhibit the innate immune responses. However, the molecular mechanisms implicated still remain unclear. To gain insight into the functions of BtpA and BtpB, we expressed them in the budding yeast Saccharomyces cerevisiae as a eukaryotic cell model. We found that both effectors were cytotoxic and that their respective TIR domains were necessary and sufficient for yeast growth inhibition. Growth arrest was concomitant with actin depolymerization, endocytic block and a general decrease in kinase activity in the cell, suggesting a failure in energetic metabolism. Indeed, levels of ATP and NAD+ were low in yeast cells expressing BtpA and BtpB TIR domains, consistent with the recently described enzymatic activity of some TIR domains as NAD+ hydrolases. In human epithelial cells, both BtpA and BtpB expression reduced intracellular total NAD levels. In infected cells, both BtpA and BtpB contributed to reduction of total NAD, indicating that their NAD+ hydrolase functions are active intracellularly during infection. Overall, combining the yeast model together with mammalian cells and infection studies our results show that BtpA and BtpB modulate energy metabolism in host cells through NAD+ hydrolysis, assigning a novel role for these TIR domain-containing effectors in Brucella pathogenesis. Brucella is a genus of zoonotic bacteria that cause severe disease in a variety of mammals, ranging from farm animals (as bovines, swine and ovine) to marine mammals. Transmission to humans, often by ingestion of non-treated dairy products, leads to serious systemic infection. Brucella abortus invades host cells and replicates intracellularly. Such behavior relies on the injection of bacterial proteins into the host cytoplasm via specialized secretion systems. Our work focuses on the study of two of these factors, BtpA and BtpB, previously described to contain Toll/Interleukin-1 Receptor (TIR)-domains that modulate innate immunity. We use here two biological models: the yeast Saccharomyces cerevisiae and human cell lines. We found that the TIR domains of both Brucella proteins were necessary and sufficient to collapse energy metabolism in yeast by depleting ATP and NAD+. This result was translatable to higher cells and consistent with the recently described NADase activity of some TIR domains both in mammalian and bacterial proteins. Importantly, we demonstrate that Brucella down-regulates total NAD levels in host cells by using both BtpA and BtpB effectors. Our results show that NAD+ is targeted by Brucella during infection, which may constitute a novel mechanism for its pathogenicity.
Collapse
|
23
|
Avila-Calderón ED, Flores-Romo L, Sharon W, Donis-Maturano L, Becerril-García MA, Arreola MGA, Reynoso BA, Güemes FS, Contreras-Rodríguez A. Dendritic cells and Brucella spp. interaction: the sentinel host and the stealthy pathogen. Folia Microbiol (Praha) 2020; 65:1-16. [PMID: 30783994 PMCID: PMC7224029 DOI: 10.1007/s12223-019-00691-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/06/2019] [Indexed: 01/18/2023]
Abstract
As dendritic cells (DCs) are among the first cells to encounter antigens, these cells trigger both innate and T cell responses, and are the most potent antigen-presenting cells. Brucella spp., which is an intracellular facultative and stealthy pathogen, is able to evade the bactericidal activities of professional phagocytes. Several studies have demonstrated that Brucella can survive and replicate intracellularly, thereby provoking impaired maturation of DCs. Therefore, the interaction between DCs and Brucella becomes an interesting model to study the immune response. In this review, we first will describe the most common techniques for DCs differentiation in vitro as well as general features of brucellosis. Then, the interaction of DCs and Brucella, including pathogen recognition, molecular mechanisms of bacterial pathogenesis, and intracellular trafficking of Brucella to subvert innate response, will be reviewed. Finally, we will debate diversity in immunological DC response and the controversial role of DC activation against Brucella infection.
Collapse
Affiliation(s)
- Eric Daniel Avila-Calderón
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, CINVESTAV-IPN, Av. IPN No 2508, Zacatenco, C.P 07330, Mexico city, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico
| | - Leopoldo Flores-Romo
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, CINVESTAV-IPN, Av. IPN No 2508, Zacatenco, C.P 07330, Mexico city, Mexico
| | - Witonsky Sharon
- Center for Molecular Medicine and Infectious Diseases/Center for One Health, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061-0442, USA
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061-0442, USA
| | - Luis Donis-Maturano
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana 3918, Zona Playitas, 22860, Ensenada, Baja California, Mexico
| | - Miguel Angel Becerril-García
- Departamento de Microbiología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Avenida Francisco I Madero y Dr. Aguirre Pequeño S/N Mitras Centro, 64460, Monterrey, Nuevo León, Mexico
| | - Ma Guadalupe Aguilera Arreola
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico
| | - Beatriz Arellano Reynoso
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico city, Mexico
| | - Francisco Suarez Güemes
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico city, Mexico
| | - Araceli Contreras-Rodríguez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico.
| |
Collapse
|
24
|
Ding J, Liu Q. Toll-like receptor 4: A promising therapeutic target for pneumonia caused by Gram-negative bacteria. J Cell Mol Med 2019; 23:5868-5875. [PMID: 31350813 PMCID: PMC6714139 DOI: 10.1111/jcmm.14529] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/11/2019] [Accepted: 06/15/2019] [Indexed: 02/06/2023] Open
Abstract
Gram‐negative bacteria (GNB) emerge as important pathogens causing pulmonary infection, which can develop into sepsis due to bacterial resistance to antibiotics. GNB pneumonia poses a huge social and economic burden all over the world. During GNB infection in the lung, Toll‐like receptor 4 (TLR4) can form a complex with MD2 and CD14 after recognizing lipopolysaccharide of GNB, initiate the MyD88‐ and TRIF‐dependent signalling pathways and stimulate host non‐specific immune response. In this review, we summarize recent progress in our understanding of the role of TLR4 in GNB pneumonia. The latest experimental results, especially in TLR4 knockout animals, suggest a promising potential of targeting TLR4 signalling pathway for the treatment of GNB pneumonia. Furthermore, we highlight the benefits of Traditional Chinese Medicine as novel candidates for the therapy of GNB pneumonia due to the modulation of TLR4 signalling pathway. Finally, we discuss the promise and challenge in the development of TLR4‐based drugs for GNB pneumonia.
Collapse
Affiliation(s)
- Junying Ding
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China.,Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Qingquan Liu
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China.,Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Institute of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
25
|
Udgata A, Dolasia K, Ghosh S, Mukhopadhyay S. Dribbling through the host defence: targeting the TLRs by pathogens. Crit Rev Microbiol 2019; 45:354-368. [DOI: 10.1080/1040841x.2019.1608904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Atul Udgata
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Manipal Academy of Higher Education, Manipal, India
| | - Komal Dolasia
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Manipal Academy of Higher Education, Manipal, India
| | - Sudip Ghosh
- Molecular Biology Division, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| |
Collapse
|
26
|
Głowacka P, Żakowska D, Naylor K, Niemcewicz M, Bielawska-Drózd A. Brucella - Virulence Factors, Pathogenesis and Treatment. Pol J Microbiol 2019; 67:151-161. [PMID: 30015453 PMCID: PMC7256693 DOI: 10.21307/pjm-2018-029] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2018] [Indexed: 12/27/2022] Open
Abstract
Brucellae are Gram-negative, small rods infecting mammals and capable of causing disease called brucellosis. The infection results in abortion and sterility in domestic animals (sheeps, pigs, rams etc). Especially dangerous for humans are: Brucella melitensis, Brucella suis, Brucella abortus, and Brucella canis that trigger unspecific symptoms (flu-like manifestation). Brucella rods are introduced via host cells, by inhalation, skin abrasions, ingestion or mucosal membranes. The most important feature of Brucella is the ability to survive and multiply within both phagocytic and non-phagocytic cells. Brucella does not produce classical virulence factors: exotoxin, cytolisins, exoenzymes, plasmids, fimbria, and drug resistant forms. Major virulence factors are: lipopolysaccharide (LPS), T4SS secretion system and BvrR/BvrS system, which allow interaction with host cell surface, formation of an early, late BCV (Brucella Containing Vacuole) and interaction with endoplasmic reticulum (ER) when the bacteria multiply. The treatment of brucellosis is based on two-drug therapy, the most common combinations of antibiotics are: doxycycline with rifampicin or fluoroquinolones with rifampicin. Currently, also other methods are used to disrupt Brucella intracellular replication (tauroursodeoxycholic acid or ginseng saponin fraction A).
Collapse
Affiliation(s)
- Patrycja Głowacka
- Biological Threats Identification and Countermeasure Center of the General Karol Kaczkowski Military Institute of Hygiene and Epidemiology,Puławy,Poland
| | - Dorota Żakowska
- Biological Threats Identification and Countermeasure Center of the General Karol Kaczkowski Military Institute of Hygiene and Epidemiology,Puławy,Poland
| | - Katarzyna Naylor
- Lublin Medical University, Department of Didactics and Medical Simulation,Lublin,Poland
| | - Marcin Niemcewicz
- Biological Threats Identification and Countermeasure Center of the General Karol Kaczkowski Military Institute of Hygiene and Epidemiology,Puławy,Poland
| | - Agata Bielawska-Drózd
- Biological Threats Identification and Countermeasure Center of the General Karol Kaczkowski Military Institute of Hygiene and Epidemiology,Puławy,Poland
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Amjadi O, Rafiei A, Mardani M, Zafari P, Zarifian A. A review of the immunopathogenesis of Brucellosis. Infect Dis (Lond) 2019; 51:321-333. [PMID: 30773082 DOI: 10.1080/23744235.2019.1568545] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Brucellosis, caused by the intracellular pathogens Brucella, is one of the major zoonotic infections. Considering the economic burden, its prevalence has been a health concern especially in endemic regions. Brucella is able to survive and replicate within host cells by expressing different virulence factors and using various strategies to avoid the host's immune response. This leads to progression of the disease from an acute phase to chronic brucellosis. Exploration of genetic variations has confirmed the expected influence of gene polymorphisms on susceptibility and resistance to brucellosis of humans. Since there is no approved human vaccine and treatment is uncertain with risk of relapse, it is important to increase knowledge about pathogenesis, diagnosis and treatment of brucellosis in order to manage and control this infection, especially in endemic regions.
Collapse
Affiliation(s)
- Omolbanin Amjadi
- a Student Research Committee, Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran
| | - Alireza Rafiei
- b Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran
| | - Masoud Mardani
- c Infectious Diseases and Tropical Medicine Research Center , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Parisa Zafari
- a Student Research Committee, Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran.,b Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran
| | - Ahmadreza Zarifian
- d Infectious Disease Research Group, Student Research Committee, Medical School , Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
30
|
Characterization of circulating miRNA signature in water buffaloes (Bubalus bubalis) during Brucella abortus infection and evaluation as potential biomarkers for non-invasive diagnosis in vaginal fluid. Sci Rep 2019; 9:1945. [PMID: 30760784 PMCID: PMC6374377 DOI: 10.1038/s41598-018-38365-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/15/2018] [Indexed: 12/12/2022] Open
Abstract
Brucellosis is an infectious disease caused by bacteria from the Brucella genus that can be transmitted to humans through contact with infected animals or contaminated animal products. Brucellosis also causes financial losses in animal production. Ruminants are highly susceptible to brucellosis, and the causative agent water buffaloes (Bubalus bubalis) is Brucella abortus. Circulating microRNAs (miRNAs) are cropping up as promising biomarkers for several infectious diseases. The goals of this study were to characterize the serum miRNA signature associated with brucellosis in water buffaloes and investigate the miRNAs’ potential use as biomarkers in vaginal fluids. Next Generation Sequencing was used to assess miRNA expression profiles in Brucella-positive and Brucella-negative blood sera; dysregulated miRNAs in blood serum and vaginal fluids were validated using RT-qPCR. ROC curves were generated to evaluate the diagnostic value of miRNAs for Brucella. GO and KEGG pathway enrichment analyses were exploited to investigate the biological functions of dysregulated miRNAs. The results showed that 20 miRNAs were modulated, of which, 12 were upregulated and 8 were downregulated. These findings were corroborated by RT-qPCR, and ROC curves indicated that the miRNAs can serve as potential biomarkers for Brucella. GO and KEGG pathway analyses pointed out that some of these miRNAs are related to immune response and apoptosis. These results provided an overview of miRNA expression profiles and highlighted potential biomarkers for Brucella infection in water buffaloes. We also demonstrated the potential of vaginal fluids in studies involving microRNA detection. Further functional and mechanistic studies of these miRNAs may improve our understanding of the biological processes involved in Brucella infection and host immune response.
Collapse
|
31
|
Rossi UA, Hasenauer FC, Caffaro ME, Raschia MA, Maurizio E, Cortez HS, Neumann RD, Poli MA, Rossetti CA. Association of an IRF3 putative functional uORF variant with resistance to Brucella infection: A candidate gene based analysis of InDel polymorphisms in goats. Cytokine 2018; 115:109-115. [PMID: 30477986 DOI: 10.1016/j.cyto.2018.11.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 11/06/2018] [Accepted: 11/19/2018] [Indexed: 12/22/2022]
Abstract
Brucellosis is an important zoonotic disease caused by infection with Brucella spp. It generates major economic losses in livestock production worldwide. Goats are the principal hosts of B. melitensis, the main infection agent of caprine and human brucellosis. The selection of resistance-related genes is considered one of the best long-term means to improve control to bacterial infection in domestic ruminants. We performed a candidate gene association study to test if six short insertion/deletion polymorphisms (InDels) at bacterial-infection related genes influence the resistance to Brucella infection in female creole goats. InDels (IRF3-540: rs660531540, FKBP5-294: rs448529294, TIRAP-561: rs657494561, PTPRT-588: rs667380588, KALRN-989: rs667660989 and RAB5a-016: rs661537016) were resolved by PCR-capillary electrophoresis in samples from 64 cases and 64 controls for brucellosis. Allelic frequencies were significantly different between cases and controls at IRF3-540 and KALRN-989 (p = 0.001 and 0.005). Indeed, the minor alleles (a and k) at InDels IRF3-540 and KALRN-989 were more frequent among controls than cases, providing evidence that these alleles confer protection against Brucella infection. Moreover, IRF3-540 a-containing genotypes (Aa and aa) were associated with absence of Brucella-specific antibodies in goats (p = 0.003; OR = 3.52; 95% CI = 1.55-7.96), and more specifically, a-allele was associated with resistance to Brucella infection in a dose-dependent manner. Also, we observed that the IRF3-540 deletion (a-allele) extends a conserved upstream ORF by 75 nucleotides to the main ORF, and thus it may decrease gene expression by reducing translation efficiency from the main ORF. These results suggest a potential functional role of IRF3-540 deletion in genetic resistance to Brucella infection in goats.
Collapse
Affiliation(s)
- Ursula A Rossi
- Instituto Nacional de Tecnología Agropecuaria, CICVyA, Instituto de Patobiología, Nicolás Repetto y de Los Reseros s/n, Hurlingham, Buenos Aires B1686, Argentina
| | - Flavia C Hasenauer
- Instituto Nacional de Tecnología Agropecuaria, CICVyA, Instituto de Patobiología, Nicolás Repetto y de Los Reseros s/n, Hurlingham, Buenos Aires B1686, Argentina; CONICET, Buenos Aires, Argentina
| | - María E Caffaro
- Inst. de Genética ''Ewald A. Favret'', Nicolás Repetto y de Los Reseros s/n, Hurlingham, Buenos Aires B1686, Argentina
| | - Maria A Raschia
- Inst. de Genética ''Ewald A. Favret'', Nicolás Repetto y de Los Reseros s/n, Hurlingham, Buenos Aires B1686, Argentina
| | - Estefania Maurizio
- Instituto Nacional de Tecnología Agropecuaria, CICVyA, Instituto de Patobiología, Nicolás Repetto y de Los Reseros s/n, Hurlingham, Buenos Aires B1686, Argentina
| | - Hector S Cortez
- Instituto Nacional de Tecnología Agropecuaria, IIACS, Area de Salud Animal, RN 68 (km 172) Cerrillos, Salta, Argentina
| | - Roberto D Neumann
- Instituto Nacional de Tecnología Agropecuaria, IIACS, Area de Salud Animal, RN 68 (km 172) Cerrillos, Salta, Argentina
| | - Mario A Poli
- Inst. de Genética ''Ewald A. Favret'', Nicolás Repetto y de Los Reseros s/n, Hurlingham, Buenos Aires B1686, Argentina
| | - Carlos A Rossetti
- Instituto Nacional de Tecnología Agropecuaria, CICVyA, Instituto de Patobiología, Nicolás Repetto y de Los Reseros s/n, Hurlingham, Buenos Aires B1686, Argentina.
| |
Collapse
|
32
|
Saqib U, Baig MS. Scaffolding role of TcpB in disrupting TLR4‐Mal interactions: Three to tango. J Cell Biochem 2018; 120:3455-3458. [DOI: 10.1002/jcb.27619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Uzma Saqib
- Discipline of Chemistry, School of Basic Sciences, Indian Institute of Technology, Indore Indore Madhya Pradesh India
| | - Mirza S Baig
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore (IITI) Madhya Pradesh India
| |
Collapse
|
33
|
Boggiatto PM, Fitzsimmons D, Bayles DO, Alt D, Vrentas CE, Olsen SC. Coincidence cloning recovery of Brucella melitensis RNA from goat tissues: advancing the in vivo analysis of pathogen gene expression in brucellosis. BMC Mol Biol 2018; 19:10. [PMID: 30068312 PMCID: PMC6071331 DOI: 10.1186/s12867-018-0111-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 07/24/2018] [Indexed: 11/10/2022] Open
Abstract
Background Brucella melitensis bacteria cause persistent, intracellular infections in small ruminants as well as in humans, leading to significant morbidity and economic loss worldwide. The majority of experiments on the transcriptional responses of Brucella to conditions inside the host have been performed following invasion of cultured mammalian cells, and do not address gene expression patterns during long-term infection. Results Here, we examine the application of the previously developed coincidence cloning methodology to recover and characterize B. melitensis RNA from the supramammary lymph node of experimentally-infected goats. Using coincidence cloning, we successfully recovered Brucella RNA from supramammary lymph nodes of B. melitensis-infected goats at both short-term (4 weeks) and long-term (38 weeks) infection time points. Amplified nucleic acid levels were sufficient for analysis of Brucella gene expression patterns by RNA-sequencing, providing evidence of metabolic activity in both the short-term and the long-term samples. We developed a workflow for the use of sequence polymorphism analysis to confirm recovery of the inoculated strain in the recovered reads, and utilized clustering analysis to demonstrate a distinct transcriptional profile present in samples recovered in long-term infection. In this first look at B. melitensis gene expression patterns in vivo, the subset of Brucella genes that was highly upregulated in long-term as compared to short-term infection included genes linked to roles in murine infection, such as genes involved in proline utilization and signal transduction. Finally, we demonstrated the challenges of qPCR validation of samples with very low ratios of pathogen:host RNA, as is the case during in vivo brucellosis, and alternatively characterized intermediate products of the coincidence cloning reaction. Conclusions Overall, this study provides the first example of recovery plus characterization of B. melitensis RNA from in vivo lymph node infection, and demonstrates that the coincidence cloning technique is a useful tool for characterizing in vivo transcriptional changes in Brucella species. Genes upregulated in long-term infection in this data set, including many genes not previously demonstrated to be virulence factors in mice or macrophage experiments, are candidates of future interest for potential roles in Brucella persistence in natural host systems. Electronic supplementary material The online version of this article (10.1186/s12867-018-0111-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paola M Boggiatto
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, 50010, USA
| | - Daniel Fitzsimmons
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, 50010, USA
| | - Darrell O Bayles
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, 50010, USA
| | - David Alt
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, 50010, USA
| | - Catherine E Vrentas
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, 50010, USA.
| | - Steven C Olsen
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, 50010, USA
| |
Collapse
|
34
|
Nanson JD, Rahaman MH, Ve T, Kobe B. Regulation of signaling by cooperative assembly formation in mammalian innate immunity signalosomes by molecular mimics. Semin Cell Dev Biol 2018; 99:96-114. [PMID: 29738879 DOI: 10.1016/j.semcdb.2018.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 03/18/2018] [Accepted: 05/04/2018] [Indexed: 12/16/2022]
Abstract
Innate immunity pathways constitute the first line of defense against infections and cellular damage. An emerging concept in these pathways is that signaling involves the formation of finite (e.g. rings in NLRs) or open-ended higher-order assemblies (e.g. filamentous assemblies by members of the death-fold family and TIR domains). This signaling by cooperative assembly formation (SCAF) mechanism allows rapid and strongly amplified responses to minute amounts of stimulus. While the characterization of the molecular mechanisms of SCAF has seen rapid progress, little is known about its regulation. One emerging theme involves proteins produced both in host cells and by pathogens that appear to mimic the signaling components. Recently characterized examples involve the capping of the filamentous assemblies formed by caspase-1 CARDs by the CARD-only protein INCA, and those formed by caspase-8 by the DED-containing protein MC159. By contrast, the CARD-only protein ICEBERG and the DED-containing protein cFLIP incorporate into signaling filaments and presumably interfere with proximity based activation of caspases. We review selected examples of SCAF in innate immunity pathways and focus on the current knowledge on signaling component mimics produced by mammalian and pathogen cells and what is known about their mechanisms of action.
Collapse
Affiliation(s)
- Jeffrey D Nanson
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Md Habibur Rahaman
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Thomas Ve
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia; Institute for Glycomics, Griffith University, Southport, QLD, 4222, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
35
|
Jakka P, Bhargavi B, Namani S, Murugan S, Splitter G, Radhakrishnan G. Cytoplasmic Linker Protein CLIP170 Negatively Regulates TLR4 Signaling by Targeting the TLR Adaptor Protein TIRAP. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:704-714. [PMID: 29222167 PMCID: PMC5760445 DOI: 10.4049/jimmunol.1601559] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/05/2017] [Indexed: 02/06/2023]
Abstract
Cytoplasmic linker protein 170 (CLIP170) is a CAP-Gly domain-containing protein that is associated with the plus end of growing microtubules and implicated in various cellular processes, including the regulation of microtubule dynamics, cell migration, and intracellular transport. Our studies revealed a previously unrecognized property and role of CLIP170. We identified CLIP170 as one of the interacting partners of Brucella effector protein TcpB that negatively regulates TLR2 and TLR4 signaling. In this study, we demonstrate that CLIP170 interacts with the TLR2 and TLR4 adaptor protein TIRAP. Furthermore, our studies revealed that CLIP170 induces ubiquitination and subsequent degradation of TIRAP to negatively regulate TLR4-mediated proinflammatory responses. Overexpression of CLIP170 in mouse macrophages suppressed the LPS-induced expression of IL-6 and TNF-α whereas silencing of endogenous CLIP170 potentiated the levels of proinflammatory cytokines. In vivo silencing of CLIP170 in C57BL/6 mice by CLIP170-specific small interfering RNA enhanced LPS-induced IL-6 and TNF-α expression. Furthermore, we found that LPS modulates the expression of CLIP170 in mouse macrophages. Overall, our experimental data suggest that CLIP170 serves as an intrinsic negative regulator of TLR4 signaling that targets TIRAP.
Collapse
Affiliation(s)
- Padmaja Jakka
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500049, India
- Graduate Studies, Manipal University, Manipal, Karnataka 576104, India; and
| | - Bindu Bhargavi
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500049, India
| | - Swapna Namani
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500049, India
| | - Subathra Murugan
- National Institute of Animal Biotechnology, Hyderabad, Telangana 500049, India
| | - Gary Splitter
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53706
| | | |
Collapse
|
36
|
Nadella V, Mohanty A, Sharma L, Yellaboina S, Mollenkopf HJ, Mazumdar VB, Palaparthi R, Mylavarapu MB, Maurya R, Kurukuti S, Rudel T, Prakash H. Inhibitors of Apoptosis Protein Antagonists (Smac Mimetic Compounds) Control Polarization of Macrophages during Microbial Challenge and Sterile Inflammatory Responses. Front Immunol 2018; 8:1792. [PMID: 29375545 PMCID: PMC5767188 DOI: 10.3389/fimmu.2017.01792] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/30/2017] [Indexed: 12/11/2022] Open
Abstract
Apoptosis is a physiological cell death process essential for development, tissue homeostasis, and for immune defense of multicellular animals. Inhibitors of apoptosis proteins (IAPs) regulate apoptosis in response to various cellular assaults. Using both genetic and pharmacological approaches we demonstrate here that the IAPs not only support opportunistic survival of intracellular human pathogens like Chlamydia pneumoniae but also control plasticity of iNOS+ M1 macrophage during the course of infection and render them refractory for immune stimulation. Treatment of Th1 primed macrophages with birinapant (IAP-specific antagonist) inhibited NO generation and relevant proteins involved in innate immune signaling. Accordingly, birinapant promoted hypoxia, angiogenesis, and tumor-induced M2 polarization of iNOS+ M1 macrophages. Interestingly, birinapant-driven changes in immune signaling were accompanied with changes in the expression of various proteins involved in the metabolism, and thus revealing the new role of IAPs in immune metabolic reprogramming in committed macrophages. Taken together, our study reveals the significance of IAP targeting approaches (Smac mimetic compounds) for the management of infectious and inflammatory diseases relying on macrophage plasticity.
Collapse
Affiliation(s)
- Vinod Nadella
- Laboratory of Translational Medicine, School of Life Sciences, University of Hyderabad, Telangana, India
| | - Aparna Mohanty
- Laboratory of Translational Medicine, School of Life Sciences, University of Hyderabad, Telangana, India
| | - Lalita Sharma
- Laboratory of Translational Medicine, School of Life Sciences, University of Hyderabad, Telangana, India
| | - Sailu Yellaboina
- YU-IOB Centre for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore, India
| | - Hans-Joachim Mollenkopf
- Core Facility Genomics and Microarray, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Varadendra Balaji Mazumdar
- Laboratory of Translational Medicine, School of Life Sciences, University of Hyderabad, Telangana, India
| | | | | | - Radheshyam Maurya
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Sreenivasulu Kurukuti
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Thomas Rudel
- Biocentre, Department of Microbiology, University of Würzburg, Würzburg, Germany
| | - Hridayesh Prakash
- Laboratory of Translational Medicine, School of Life Sciences, University of Hyderabad, Telangana, India
| |
Collapse
|
37
|
Jakka P, Namani S, Murugan S, Rai N, Radhakrishnan G. The Brucella effector protein TcpB induces degradation of inflammatory caspases and thereby subverts non-canonical inflammasome activation in macrophages. J Biol Chem 2017; 292:20613-20627. [PMID: 29061850 DOI: 10.1074/jbc.m117.815878] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/08/2017] [Indexed: 12/20/2022] Open
Abstract
The inflammasome contains intracellular receptors that recognize various pathogen-associated molecular patterns and play crucial roles in innate immune responses to invading pathogens. Non-canonical inflammasome activation is mediated by caspase-4/11, which recognizes intracellular LPS and promotes pyroptosis and secretion of proinflammatory cytokines. Brucella species are infectious intracellular pathogens that replicate in professional and non-professional phagocytic cells and subvert immune responses for chronic persistence in the host. The Brucella effector protein TcpB suppresses Toll-like receptor 2 (TLR2)- and TLR4-mediated innate immune responses by targeted degradation of the Toll/interleukin-1 receptor (TIR) domain-containing adaptor protein. TcpB is a cell-permeable protein with multiple functions, and its intracellular targets other than TIR domain-containing adaptor protein remain unclear. Here, we report that TcpB induces ubiquitination and degradation of the inflammatory caspases 1, 4, and 11. Furthermore, in both mouse and human macrophages, TcpB attenuated LPS-induced non-canonical inflammasome activation and suppressed pyroptosis and secretion of IL-1α and IL-1β induced by intracellular LPS delivery. The intact TIR domain was essential for TcpB to subvert the non-canonical inflammasome activation as a TcpB(G158A) mutant failed to suppress pyroptotic cell death and inflammatory responses. Brucella-infected macrophages exhibited minimal pyroptosis but secreted IL-1β, which was suppressed by TcpB. We also demonstrated that TcpB protein can efficiently attenuate Salmonella enterica serovar Typhimurium-induced pyroptosis and proinflammatory cytokine secretion in macrophages. Because TcpB suppresses both TLR4- and caspase-4/11-mediated inflammation, TcpB might be a candidate target for developing drugs against LPS-induced septicemia.
Collapse
Affiliation(s)
- Padmaja Jakka
- From the Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana 500049, India and.,Graduate Studies, Manipal University, Manipal, Karnataka 576104, India
| | - Swapna Namani
- From the Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana 500049, India and
| | - Subathra Murugan
- From the Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana 500049, India and
| | - Nivedita Rai
- From the Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana 500049, India and
| | - Girish Radhakrishnan
- From the Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology, Hyderabad, Telangana 500049, India and
| |
Collapse
|
38
|
Abstract
Many bacterial pathogens can cause acute infections that are cleared with the onset of adaptive immunity, but a subset of these pathogens can establish persistent, and sometimes lifelong, infections. While bacteria that cause chronic infections are phylogenetically diverse, they share common features in their interactions with the host that enable a protracted period of colonization. This article will compare the persistence strategies of two chronic pathogens from the Proteobacteria, Brucella abortus and Salmonella enterica serovar Typhi, to consider how these two pathogens, which are very different at the genomic level, can utilize common strategies to evade immune clearance to cause chronic intracellular infections of the mononuclear phagocyte system.
Collapse
|
39
|
Imbert PR, Louche A, Luizet JB, Grandjean T, Bigot S, Wood TE, Gagné S, Blanco A, Wunderley L, Terradot L, Woodman P, Garvis S, Filloux A, Guery B, Salcedo SP. A Pseudomonas aeruginosa TIR effector mediates immune evasion by targeting UBAP1 and TLR adaptors. EMBO J 2017; 36:1869-1887. [PMID: 28483816 PMCID: PMC5494471 DOI: 10.15252/embj.201695343] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 03/29/2017] [Accepted: 04/05/2017] [Indexed: 12/11/2022] Open
Abstract
Bacterial pathogens often subvert the innate immune system to establish a successful infection. The direct inhibition of downstream components of innate immune pathways is particularly well documented but how bacteria interfere with receptor proximal events is far less well understood. Here, we describe a Toll/interleukin 1 receptor (TIR) domain‐containing protein (PumA) of the multi‐drug resistant Pseudomonas aeruginosa PA7 strain. We found that PumA is essential for virulence and inhibits NF‐κB, a property transferable to non‐PumA strain PA14, suggesting no additional factors are needed for PumA function. The TIR domain is able to interact with the Toll‐like receptor (TLR) adaptors TIRAP and MyD88, as well as the ubiquitin‐associated protein 1 (UBAP1), a component of the endosomal‐sorting complex required for transport I (ESCRT‐I). These interactions are not spatially exclusive as we show UBAP1 can associate with MyD88, enhancing its plasma membrane localization. Combined targeting of UBAP1 and TLR adaptors by PumA impedes both cytokine and TLR receptor signalling, highlighting a novel strategy for innate immune evasion.
Collapse
Affiliation(s)
- Paul Rc Imbert
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Arthur Louche
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Jean-Baptiste Luizet
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Teddy Grandjean
- EA 7366 Recherche Translationelle Relations Hôte-Pathogènes, Faculté de Médecine Pôle Recherche, Université Lille 2, Lille, France
| | - Sarah Bigot
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Thomas E Wood
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Stéphanie Gagné
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Amandine Blanco
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Lydia Wunderley
- School of Biological Sciences, Faculty of Biology Medicine and Health University of Manchester Manchester Academic Health Science Centre, Manchester, UK†
| | - Laurent Terradot
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| | - Philip Woodman
- School of Biological Sciences, Faculty of Biology Medicine and Health University of Manchester Manchester Academic Health Science Centre, Manchester, UK†
| | - Steve Garvis
- Laboratoire de Biologie et Modelisation, Ecole Normal Supérieur, UMR5239, Lyon, France
| | - Alain Filloux
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Benoit Guery
- EA 7366 Recherche Translationelle Relations Hôte-Pathogènes, Faculté de Médecine Pôle Recherche, Université Lille 2, Lille, France
| | - Suzana P Salcedo
- Laboratory of Molecular Microbiology and Structural Biochemistry, Centre National de la Recherche Scientifique, University of Lyon, Lyon, France
| |
Collapse
|
40
|
Rodríguez AM, Delpino MV, Miraglia MC, Costa Franco MM, Barrionuevo P, Dennis VA, Oliveira SC, Giambartolomei GH. Brucella abortus-activated microglia induce neuronal death through primary phagocytosis. Glia 2017; 65:1137-1151. [PMID: 28398652 DOI: 10.1002/glia.23149] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/13/2017] [Accepted: 03/22/2017] [Indexed: 01/18/2023]
Affiliation(s)
- Ana M. Rodríguez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires; Buenos Aires Argentina
| | - M. Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires; Buenos Aires Argentina
| | - M. Cruz Miraglia
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires; Buenos Aires Argentina
| | - Miriam M. Costa Franco
- Department of Biochemistry and Immunology; Institute of Biological Sciences, Federal University of Minas Gerais; Belo Horizonte-Minas Gerais Brazil
| | - Paula Barrionuevo
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina); Buenos Aires Argentina
| | - Vida A. Dennis
- Center for Nano Biotechnology Research and Department of Biological Sciences; Alabama State University; Montgomery AL
| | - Sergio C. Oliveira
- Department of Biochemistry and Immunology; Institute of Biological Sciences, Federal University of Minas Gerais; Belo Horizonte-Minas Gerais Brazil
| | - Guillermo H. Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires; Buenos Aires Argentina
| |
Collapse
|
41
|
Odendall C, Kagan JC. Activation and pathogenic manipulation of the sensors of the innate immune system. Microbes Infect 2017; 19:229-237. [PMID: 28093320 PMCID: PMC6697111 DOI: 10.1016/j.micinf.2017.01.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/06/2017] [Indexed: 12/13/2022]
Abstract
The innate immune system detects the presence of microbes through different families of pattern-recognition receptors (PRRs). PRRs detect pathogens of all origins and trigger signaling events that activate innate and adaptive immunity. These events need to be tightly regulated in order to ensure optimal activation when required, and minimal signaling in the absence of microbial encounters. This regulation is achieved, at least in part, through the precise subcellular positioning of receptors and downstream signaling proteins. Consequently, mislocalization of these proteins inhibits innate immune pathways, and pathogens have evolved to alter host protein localization as a strategy to evade immune detection. This review describes the importance of subcellular localization of various PRR families and their adaptors, and highlights pathogenic immune evasion strategies that operate by altering immune protein localization.
Collapse
Affiliation(s)
- Charlotte Odendall
- Department of Infectious Diseases, King's College London, London SE1 9RT, UK
| | - Jonathan C Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
42
|
Brucella TIR-like protein TcpB/Btp1 specifically targets the host adaptor protein MAL/TIRAP to promote infection. Biochem Biophys Res Commun 2016; 477:509-14. [PMID: 27311859 DOI: 10.1016/j.bbrc.2016.06.064] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 06/13/2016] [Indexed: 02/06/2023]
Abstract
Brucella spp. are known to avoid host immune recognition and weaken the immune response to infection. Brucella like accomplish this by employing two clever strategies, called the stealth strategy and hijacking strategy. The TIR domain-containing protein (TcpB/Btp1) of Brucella melitensis is thought to be involved in inhibiting host NF-κB activation by binding to adaptors downstream of Toll-like receptors. However, of the five TIR domain-containing adaptors conserved in mammals, whether MyD88 or MAL, even other three adaptors, are specifically targeted by TcpB has not been identified. Here, we confirmed the effect of TcpB on B.melitensis virulence in mice and found that TcpB selectively targets MAL. By using siRNA against MAL, we found that TcpB from B.melitensis is involved in intracellular survival and that MAL affects intracellular replication of B.melitensis. Our results confirm that TcpB specifically targets MAL/TIRAP to disrupt downstream signaling pathways and promote intra-host survival of Brucella spp.
Collapse
|
43
|
Ke Y, Li W, Wang Y, Yang M, Guo J, Zhan S, Du X, Wang Z, Yang M, Li J, Li W, Chen Z. Inhibition of TLR4 signaling by Brucella TIR-containing protein TcpB-derived decoy peptides. Int J Med Microbiol 2016; 306:391-400. [PMID: 27289452 DOI: 10.1016/j.ijmm.2016.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 05/04/2016] [Accepted: 05/08/2016] [Indexed: 02/06/2023] Open
Abstract
Brucella spp. avoid host immune recognition and thus, weaken the immune response to infection. The Toll/interleukin-1 receptor (TIR) domain-containing protein (TcpB/Btp1) of Brucella spp. is thought to be involved in blocking host innate immune responses by binding to adaptors downstream of Toll-like receptors. In this study, based on the observation that TcpB binds to the host target proteins, MAL, through the TIR domain, we examined decoy peptides from TcpB TIR domains and found that TB-8 and TB-9 substantially inhibit lipopolysaccharide (LPS)-induced signaling in vitro and in vivo. Both these peptides share a common loop, the DD loop, indicating a novel structural region mediating TIR interactions. The inhibition of LPS signaling by TB-8 and TB-9 shows no preference to MyD88-dependent cytokines, such as TNF-α and IL-1β or TRIF-dependent cytokines including IFN-β and IL-6. Furthermore, these two peptides rescue the virulence of Brucella ΔtcpB mutants at the cellular level, indicating key roles of the DD loop in Brucella pathogenesis. In conclusion, identification of inhibitors from the bacterial TIR domains is helpful not only for illustrating interacting mechanisms between TIR domains and bacterial pathogenesis, but also for developing novel signaling inhibitors and therapeutics for human inflammatory diseases.
Collapse
Affiliation(s)
- Yuehua Ke
- Institute of Disease Control and Prevention, AMMS, Beijing 100071, People's Republic of China
| | - Wenna Li
- Institute of Disease Control and Prevention, AMMS, Beijing 100071, People's Republic of China; School of Public Health, Jilin University, Changchun 130021, People's Republic of China
| | - Yufei Wang
- General Hospital of People's Armed Police Forces, Beijing 100039, People's Republic of China
| | - Mingjuan Yang
- Institute of Disease Control and Prevention, AMMS, Beijing 100071, People's Republic of China
| | - Jinpeng Guo
- Institute of Disease Control and Prevention, AMMS, Beijing 100071, People's Republic of China
| | - Shaoxia Zhan
- Institute of Disease Control and Prevention, AMMS, Beijing 100071, People's Republic of China
| | - Xinying Du
- Institute of Disease Control and Prevention, AMMS, Beijing 100071, People's Republic of China
| | - Zhoujia Wang
- Institute of Disease Control and Prevention, AMMS, Beijing 100071, People's Republic of China
| | - Min Yang
- Construction Engineering Research Institute, Xi'an 71000, Shanxi Province, People's Republic of China
| | - Juan Li
- School of Public Health, Jilin University, Changchun 130021, People's Republic of China.
| | - Wenfeng Li
- Department of Orthopedics, The First Affiliated Hospital of General Hospital of People's Liberation Army, Beijing 100048, People's Republic of China.
| | - Zeliang Chen
- Institute of Disease Control and Prevention, AMMS, Beijing 100071, People's Republic of China; College of Medicine, Shihezi University, Shihezi, People's Republic of China; Research Center of Molecular Biology, Inner Mongolia Medical University, Hohhot, People's Republic of China.
| |
Collapse
|
44
|
A Comparative Analysis of the Mechanism of Toll-Like Receptor-Disruption by TIR-Containing Protein C from Uropathogenic Escherichia coli. Pathogens 2016; 5:pathogens5010025. [PMID: 26938564 PMCID: PMC4810146 DOI: 10.3390/pathogens5010025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 02/23/2016] [Accepted: 02/25/2016] [Indexed: 11/17/2022] Open
Abstract
The TIR-containing protein C (TcpC) of uropathogenic Escherichia coli strains is a powerful virulence factor by impairing the signaling cascade of Toll-like receptors (TLRs). Several other bacterial pathogens like Salmonella, Yersinia, Staphylococcus aureus but also non-pathogens express similar proteins. We discuss here the pathogenic potential of TcpC and its interaction with TLRs and TLR-adapter proteins on the molecular level and compare its activity with the activity of other bacterial TIR-containing proteins. Finally, we analyze and compare the structure of bacterial TIR-domains with the TIR-domains of TLRs and TLR-adapters.
Collapse
|
45
|
Abstract
Brucellosis, caused by bacteria of the genus Brucella, is an important zoonotic infection that causes reproductive disease in domestic animals and chronic debilitating disease in humans. An intriguing aspect of Brucella infection is the ability of these bacteria to evade the host immune response, leading to pathogen persistence. Conversely, in the reproductive tract of infected animals, this stealthy pathogen is able to cause an acute severe inflammatory response. In this review, we discuss the different mechanisms used by Brucella to cause disease, with emphasis on its virulence factors and the dichotomy between chronic persistence and reproductive disease.
Collapse
Affiliation(s)
| | - Renee M Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616; ,
| |
Collapse
|
46
|
McGuire VA, Arthur JSC. Subverting Toll-Like Receptor Signaling by Bacterial Pathogens. Front Immunol 2015; 6:607. [PMID: 26648936 PMCID: PMC4664646 DOI: 10.3389/fimmu.2015.00607] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/13/2015] [Indexed: 12/26/2022] Open
Abstract
Pathogenic bacteria are detected by pattern-recognition receptors (PRRs) expressed on innate immune cells, which activate intracellular signal transduction pathways to elicit an immune response. Toll-like receptors are, perhaps, the most studied of the PRRs and can activate the mitogen-activated protein kinase (MAPK) and Nuclear Factor-κB (NF-κB) pathways. These pathways are critical for mounting an effective immune response. In order to evade detection and promote virulence, many pathogens subvert the host immune response by targeting components of these signal transduction pathways. This mini-review highlights the diverse mechanisms that bacterial pathogens have evolved to manipulate the innate immune response, with a particular focus on those that target MAPK and NF-κB signaling pathways. Understanding the elaborate strategies that pathogens employ to subvert the immune response not only highlights the importance of these proteins in mounting effective immune responses, but may also identify novel approaches for treatment or prevention of infection.
Collapse
Affiliation(s)
- Victoria A McGuire
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee , Dundee , UK
| | - J Simon C Arthur
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee , Dundee , UK
| |
Collapse
|
47
|
Ke Y, Wang Y, Li W, Chen Z. Type IV secretion system of Brucella spp. and its effectors. Front Cell Infect Microbiol 2015; 5:72. [PMID: 26528442 PMCID: PMC4602199 DOI: 10.3389/fcimb.2015.00072] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 09/28/2015] [Indexed: 11/13/2022] Open
Abstract
Brucella spp. are intracellular bacterial pathogens that cause infection in domestic and wild animals. They are often used as model organisms to study intracellular bacterial infections. Brucella VirB T4SS is a key virulence factor that plays important roles in mediating intracellular survival and manipulating host immune response to infection. In this review, we discuss the roles of Brucella VirB T4SS and 15 effectors that are proposed to be crucial for Brucella pathogenesis. VirB T4SS regulates the inflammation response and manipulates vesicle trafficking inside host cells. VirB T4SS also plays crucial roles in the inhibition of the host immune response and intracellular survival during infection. Here, we list the key molecular events in the intracellular life cycle of Brucella that are potentially targeted by the VirB T4SS effectors. Elucidating the functions of these effectors will help clarify the molecular role of T4SS during infection. Furthermore, studying the effectors secreted by Brucella spp. might provide insights into the mechanisms used by the bacteria to hijack the host signaling pathways and aid in the development of better vaccines and therapies against brucellosis.
Collapse
Affiliation(s)
- Yuehua Ke
- Institute of Disease Control and Prevention, AMMS Beijing, China
| | - Yufei Wang
- Department of Laboratory Medicine, General Hospital of Chinese People's Armed Police Forces Beijing, China
| | - Wengfeng Li
- Department of Orthopedics, The First Affiliated Hospital of General Hospital of People's Liberation Army Beijing, China
| | - Zeliang Chen
- Institute of Disease Control and Prevention, AMMS Beijing, China
| |
Collapse
|
48
|
Gupta CL, Akhtar S, Sayyed U, Pathak N, Bajpai P. In silicoanalysis of human Toll-like receptor 7 ligand binding domain. Biotechnol Appl Biochem 2015; 63:441-50. [DOI: 10.1002/bab.1377] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 03/23/2015] [Indexed: 12/28/2022]
Affiliation(s)
| | - Salman Akhtar
- Department of Bioengineering; Integral University; Lucknow India
| | - Uzma Sayyed
- Department of Biosciences; Integral University; Lucknow India
| | - Neelam Pathak
- Department of Biosciences; Integral University; Lucknow India
| | - Preeti Bajpai
- Department of Biosciences; Integral University; Lucknow India
| |
Collapse
|
49
|
Sellge G, Kufer TA. PRR-signaling pathways: Learning from microbial tactics. Semin Immunol 2015; 27:75-84. [PMID: 25911384 DOI: 10.1016/j.smim.2015.03.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 03/13/2015] [Indexed: 12/13/2022]
Abstract
Recognition of bacterial pathogens by the mammalian host relies on the induction of early innate immune responses initiated by the activation of pattern-recognition receptors (PRRs) upon sensing of their cognate microbe-associated-patterns (MAMPs). Successful pathogens have evolved to intercept PRR activation and signaling at multiple steps. The molecular dissection of the underlying mechanisms revealed many of the basic mechanisms used by the immune system. Here we provide an overview of the different strategies used by bacterial pathogens and commensals to subvert and reprogram PPR-mediated innate immune responses. A particular attention is given to recent discoveries highlighting novel molecular details of the host inflammatory response in mammalian cells and current advances in our understanding of the interaction of commensals with PRR-mediated responses.
Collapse
Affiliation(s)
- Gernot Sellge
- Department of Medicine III, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Thomas A Kufer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, Fruwirthstr. 12, 70599 Stuttgart, Germany.
| |
Collapse
|
50
|
de Figueiredo P, Ficht TA, Rice-Ficht A, Rossetti CA, Adams LG. Pathogenesis and immunobiology of brucellosis: review of Brucella-host interactions. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1505-17. [PMID: 25892682 DOI: 10.1016/j.ajpath.2015.03.003] [Citation(s) in RCA: 283] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 02/10/2015] [Accepted: 03/02/2015] [Indexed: 01/18/2023]
Abstract
This review of Brucella-host interactions and immunobiology discusses recent discoveries as the basis for pathogenesis-informed rationales to prevent or treat brucellosis. Brucella spp., as animal pathogens, cause human brucellosis, a zoonosis that results in worldwide economic losses, human morbidity, and poverty. Although Brucella spp. infect humans as an incidental host, 500,000 new human infections occur annually, and no patient-friendly treatments or approved human vaccines are reported. Brucellae display strong tissue tropism for lymphoreticular and reproductive systems with an intracellular lifestyle that limits exposure to innate and adaptive immune responses, sequesters the organism from the effects of antibiotics, and drives clinical disease manifestations and pathology. Stealthy brucellae exploit strategies to establish infection, including i) evasion of intracellular destruction by restricting fusion of type IV secretion system-dependent Brucella-containing vacuoles with lysosomal compartments, ii) inhibition of apoptosis of infected mononuclear cells, and iii) prevention of dendritic cell maturation, antigen presentation, and activation of naive T cells, pathogenesis lessons that may be informative for other intracellular pathogens. Data sets of next-generation sequences of Brucella and host time-series global expression fused with proteomics and metabolomics data from in vitro and in vivo experiments now inform interactive cellular pathways and gene regulatory networks enabling full-scale systems biology analysis. The newly identified effector proteins of Brucella may represent targets for improved, safer brucellosis vaccines and therapeutics.
Collapse
Affiliation(s)
- Paul de Figueiredo
- Department of Veterinary Pathobiology, Texas A&M University and Texas AgriLife Research, College Station, Texas; Norman Borlaug Center, Texas A&M University, College Station, Texas; Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas
| | - Thomas A Ficht
- Department of Veterinary Pathobiology, Texas A&M University and Texas AgriLife Research, College Station, Texas
| | - Allison Rice-Ficht
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - Carlos A Rossetti
- Institute of Pathobiology, CICVyA-CNIA, National Institute of Animal Agriculture Technology (INTA), Buenos Aires, Argentina
| | - L Garry Adams
- Department of Veterinary Pathobiology, Texas A&M University and Texas AgriLife Research, College Station, Texas.
| |
Collapse
|