1
|
Zhang M, Qi L, Li J, Yuan N, Zhai Y, Hao M, Zhou D, Liu W, Jin Y, Wang A. SIRT2 inhibition enhances mitochondrial apoptosis in Brucella-infected bovine placental trophoblast cells. Vet Res 2025; 56:97. [PMID: 40317067 PMCID: PMC12049057 DOI: 10.1186/s13567-025-01518-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/11/2025] [Indexed: 05/04/2025] Open
Abstract
Brucella is a successful pathogen that employs a plethora of immune evasion mechanisms. This contributes to pathogenesis and persistence and limits the efficacy of available treatments. An increasing understanding of host‒pathogen interactions suggests that integrating host-directed strategies with existing anti-Brucella treatments could lead to more effective bacterial clearance and a reduction in drug-resistant strains. SIRT2 is a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase found in mammals. It can deacetylate various transcription factors and regulatory proteins, playing crucial roles in host‒pathogen interactions and pathogen infection-induced apoptosis. In this study, we investigated the role of SIRT2 in Brucella-induced cell apoptosis using bovine placental trophoblast cells. Our results indicate that B. abortus A19 infection upregulates SIRT2 protein expression and significantly induces mitochondrial apoptosis in these cells. Furthermore, inhibition of SIRT2 exacerbates B. abortus A19-induced mitochondrial apoptosis and markedly inhibits intracellular bacterial survival. These results prove the role of SIRT2 in Brucella pathogenesis and the mechanism of action.
Collapse
Affiliation(s)
- Mengyu Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang, 712100, China
| | - Lin Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang, 712100, China
| | - Junmei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang, 712100, China
| | - NingQiu Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang, 712100, China
| | - Yunyi Zhai
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang, 712100, China
| | - Mingyue Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang, 712100, China
| | - Dong Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang, 712100, China
| | - Wei Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang, 712100, China
| | - Yaping Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang, 712100, China
| | - Aihua Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang, 712100, China.
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang, 712100, China.
| |
Collapse
|
2
|
Li X, Wang B, Li X, He J, Shi Y, Wang R, Li D, Haitao D. Analysis and validation of serum biomarkers in brucellosis patients through proteomics and bioinformatics. Front Cell Infect Microbiol 2025; 14:1446339. [PMID: 39872944 PMCID: PMC11769985 DOI: 10.3389/fcimb.2024.1446339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/24/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction This study aims to utilize proteomics, bioinformatics, and machine learning algorithms to identify diagnostic biomarkers in the serum of patients with acute and chronic brucellosis. Methods Proteomic analysis was conducted on serum samples from patients with acute and chronic brucellosis, as well as from healthy controls. Differential expression analysis was performed to identify proteins with altered expression, while Weighted Gene Co-expression Network Analysis (WGCNA) was applied to detect co-expression modules associated with clinical features of brucellosis. Machine learning algorithms were subsequently used to identify the optimal combination of diagnostic biomarkers. Finally, ELISA was employed to validate the identified proteins. Results A total of 1,494 differentially expressed proteins were identified, revealing two co-expression modules significantly associated with the clinical characteristics of brucellosis. The Gaussian Mixture Model (GMM) algorithm identified six proteins that were concurrently present in both the differentially expressed and co-expression modules, demonstrating promising diagnostic potential. After ELISA validation, five proteins were ultimately selected. Discussion These five proteins are implicated in the innate immune processes of brucellosis, potentially associated with its pathogenic mechanisms and chronicity. Furthermore, we highlighted their potential as diagnostic biomarkers for brucellosis. This study further enhances our understanding of brucellosis at the protein level, paving the way for future research endeavors.
Collapse
Affiliation(s)
- Xiao Li
- Department of Inner Mongolia Clinical Medicine College, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Bo Wang
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Xiaocong Li
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Juan He
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Yue Shi
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Rui Wang
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Dongwei Li
- Department of Inner Mongolia Clinical Medicine College, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Ding Haitao
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Hohhot, Inner Mongolia, China
| |
Collapse
|
3
|
Qin Y, Zhou G, Jiao F, Cheng C, Meng C, Wang L, Wu S, Fan C, Li J, Zhou B, Chu Y, Jiao H. Brucella mediates autophagy, inflammation, and apoptosis to escape host killing. Front Cell Infect Microbiol 2024; 14:1408407. [PMID: 39507949 PMCID: PMC11537862 DOI: 10.3389/fcimb.2024.1408407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/26/2024] [Indexed: 11/08/2024] Open
Abstract
Brucellosis is a serious zoonosis caused by Brucella spp. infection, which not only seriously jeopardizes the health of humans and mammals, but also causes huge economic losses to the livestock industry. Brucella is a Gram-negative intracellular bacterium that relies primarily on its virulence factors and a variety of evolved survival strategies to replicate and proliferate within cells. Currently, the mechanisms of autophagy, inflammation, and apoptosis in Brucella-infected hosts are not fully understood and require further research and discussion. This review focuses on the relationship between Brucella and autophagy, inflammation, and apoptosis to provide the scientific basis for revealing the pathogenesis of Brucella.
Collapse
Affiliation(s)
- Yaqiong Qin
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Gengxu Zhou
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Fengyuan Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Chuan Cheng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Chi Meng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Lingjie Wang
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Shengping Wu
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Cailiang Fan
- The College of Veterinary Medicine, Southwest University, Chongqing, China
- Animal Epidemic Prevention and Control Center of Rongchang, Chongqing, China
| | - Jixiang Li
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Bo Zhou
- Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Yuefeng Chu
- 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, Gansu, China
| | - Hanwei Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
4
|
Li Z, Wang S, Han J, Shi C, Xi L, Cui Y, Zhang H. Expression of cytokine and Apoptosis-Associated genes in mice bone Marrow-Derived Macrophages stimulated with Brucella recombinant type IV secretion effectors. Cytokine 2024; 182:156711. [PMID: 39094437 DOI: 10.1016/j.cyto.2024.156711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/23/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Brucellosis is an economically important infectious caused by most commonly by Brucella. Detection of infected animals at the early stage is important for controlling the disease. The diagnostic antigens, usually protein antigens, have attracted much interest. However, the accurate mechanism of immune response is still unknown. The secretory effectors (BPE005, BPE275, and BPE123) of the type IV secretion system (T4SS) were involved in the intracellular circulation process of Brucella and the immune responses of the host. METHODS Genes encoding three B. abortus effector proteins (BPE005, BPE275, and BPE123) of T4SS were cloned and the recombinant proteins were expressed and purified. The purified recombinant proteins were named rBPE005, rBPE275 and rBPE123. Then, the expressions of Th1- and Th2-related cytokine genes were analyzed in mice bone marrow-derived macrophages (BMDMs) after stimulation with rBPE005, rBPE275, and rBPE123. Furthermore, four apoptosis-associated genes (Caspase-3, Caspase-8, Bax, and Bcl-2) were also detected to explore the damage of the proteins to the cells. RESULTS Expressions of all Th1- and Th2-related cytokine genes were induced with three proteins, and different cytokine expression patterns induced by each protein depend on the stimulation time and dose of protein. However, expressions of apoptosis-related genes did not change. CONCLUSION These results showed that the secreted antigens of Brucella induced an immune reaction via the production of Th1- and Th2-type cytokines in BMDMs without exerting any damage on the cells.
Collapse
Affiliation(s)
- Zhiqiang Li
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Shuli Wang
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Jincheng Han
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Chuanxin Shi
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Li Xi
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Yanyan Cui
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China; College of Smart Animal Husbandry, Shangqiu Normal University, Shangqiu 476000, Henan Provence, China
| | - Hui Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang Province, China.
| |
Collapse
|
5
|
Wang Y, Li H, Xu Z, Yi J, Li W, Meng C, Zhang H, Deng X, Ma Z, Wang Y, Chen C. Exosomes released by Brucella-infected macrophages inhibit the intracellular survival of Brucella by promoting the polarization of M1 macrophages. Microb Biotechnol 2023; 16:1524-1535. [PMID: 37212362 PMCID: PMC10281354 DOI: 10.1111/1751-7915.14274] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/23/2023] Open
Abstract
Exosomes, membrane vesicles released extracellularly from cells, contain nucleic acids, proteins, lipids and other components, allowing the transfer of material information between cells. Recent studies reported the role of exosomes in pathogenic microbial infection and host immune mechanisms. Brucella-invasive bodies can survive in host cells for a long time and cause chronic infection, which causes tissue damage. Whether exosomes are involved in host anti-Brucella congenital immune responses has not been reported. Here, we extracted and identified exosomes secreted by Brucella melitensis M5 (Exo-M5)-infected macrophages, and performed in vivo and in vitro studies to examine the effects of exosomes carrying antigen on the polarization of macrophages and immune activation. Exo-M5 promoted the polarization of M1 macrophages, which induced the significant secretion of M1 cytokines (tumour necrosis factor-α and interferon-γ) through NF-κB signalling pathways and inhibited the secretion of M2 cytokines (IL-10), thereby inhibiting the intracellular survival of Brucella. Exo-M5 activated innate immunity and promoted the release of IgG2a antibodies that protected mice from Brucella infection and reduced the parasitaemia of Brucella in the spleen. Furthermore, Exo-M5 contained Brucella antigen components, including Omp31 and OmpA. These results demonstrated that exosomes have an important role in immune responses against Brucella, which might help elucidate the mechanisms of host immunity against Brucella infection and aid the search for Brucella biomarkers and the development of new vaccine candidates.
Collapse
Affiliation(s)
- Yueli Wang
- College of Animal Science and TechnologyShihezi UniversityShiheziChina
| | - Honghuan Li
- College of Animal Science and TechnologyShihezi UniversityShiheziChina
| | - Zhenyu Xu
- College of Animal Science and TechnologyShihezi UniversityShiheziChina
| | - Jihai Yi
- College of Animal Science and TechnologyShihezi UniversityShiheziChina
| | - Wei Li
- Xinjiang Center for Animal Disease Control and PreventionUrumqiChina
| | - Chuang Meng
- Jiangsu Key Laboratory of ZoonosisYangzhou UniversityYangzhouChina
| | - Huan Zhang
- College of Animal Science and TechnologyShihezi UniversityShiheziChina
| | - Xiaoyu Deng
- College of Animal Science and TechnologyShihezi UniversityShiheziChina
| | - Zhongchen Ma
- College of Animal Science and TechnologyShihezi UniversityShiheziChina
| | - Yong Wang
- College of Animal Science and TechnologyShihezi UniversityShiheziChina
| | - Chuangfu Chen
- College of Animal Science and TechnologyShihezi UniversityShiheziChina
| |
Collapse
|
6
|
Guo X, Zeng H, Li M, Xiao Y, Gu G, Song Z, Shuai X, Guo J, Huang Q, Zhou B, Chu Y, Jiao H. The mechanism of chronic intracellular infection with Brucella spp. Front Cell Infect Microbiol 2023; 13:1129172. [PMID: 37143745 PMCID: PMC10151771 DOI: 10.3389/fcimb.2023.1129172] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Globally, brucellosis is a widespread zoonotic disease. It is prevalent in more than 170 countries and regions. It mostly damages an animal's reproductive system and causes extreme economic losses to the animal husbandry industry. Once inside cells, Brucella resides in a vacuole, designated the BCV, which interacts with components of the endocytic and secretory pathways to ensure bacterial survival. Numerous studies conducted recently have revealed that Brucella's ability to cause a chronic infection depends on how it interacts with the host. This paper describes the immune system, apoptosis, and metabolic control of host cells as part of the mechanism of Brucella survival in host cells. Brucella contributes to both the body's non-specific and specific immunity during chronic infection, and it can aid in its survival by causing the body's immune system to become suppressed. In addition, Brucella regulates apoptosis to avoid being detected by the host immune system. The BvrR/BvrS, VjbR, BlxR, and BPE123 proteins enable Brucella to fine-tune its metabolism while also ensuring its survival and replication and improving its ability to adapt to the intracellular environment.
Collapse
Affiliation(s)
- Xiaoyi Guo
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Hui Zeng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Mengjuan Li
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yu Xiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Guojing Gu
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenhui Song
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xuehong Shuai
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jianhua Guo
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Qingzhou Huang
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Bo Zhou
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
- *Correspondence: Bo Zhou, ; Yuefeng Chu, ; Hanwei Jiao,
| | - Yuefeng Chu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- *Correspondence: Bo Zhou, ; Yuefeng Chu, ; Hanwei Jiao,
| | - Hanwei Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
- The Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
- *Correspondence: Bo Zhou, ; Yuefeng Chu, ; Hanwei Jiao,
| |
Collapse
|
7
|
Wang Y, Xi J, Wu P, Zhang H, Deng X, Wang Y, Ma Z, Yi J, Chen C. Small ubiquitin-related modifier 2 affects the intracellular survival of Brucella abortus 2308 by regulating activation of the NF-κB pathway. Innate Immun 2020; 27:81-88. [PMID: 33241964 PMCID: PMC7780357 DOI: 10.1177/1753425920972171] [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] [Indexed: 11/26/2022] Open
Abstract
Brucella is a genus of Gram-negative intracellular pathogens that cause animal and human diseases. Brucella survival and replication inside immune cells is critical for the establishment of chronic infections. Protein modifications by small ubiquitin-related modifier proteins and the NF-κB pathway are involved in many cellular activities, playing major roles in regulating protein function that is essential for pathogenic bacteria during infection. However, the relationship between them in the intracellular survival of Brucella is still largely unknown. We demonstrated that Brucella abortus 2308 infection can activate the expression of small ubiquitin-related modifier-2 proteins in a time-dependent manner. We found the production of Th1 cytokines (IFN-γ and TNF-α) and the transcription of NF-κB/p65 were promoted by overexpression and inhibited by interference of small ubiquitin-related modifier-2. In addition, we showed that small ubiquitin-related modifier-2 can inhibit intracellular survival of Brucella abortus 2308 by regulating activation of the NF-κB pathway. Taken together, this work shows that small ubiquitin-related modifier-2 modification of NF-κB2/p65 is essential for the survival of Brucella abortus 2308 inside macrophages. This work may help to unravel the pathogenic mechanisms of Brucella infections.
Collapse
Affiliation(s)
- Yueli Wang
- College of Animal Science and Technology, Shihezi University, China
| | - Jing Xi
- College of Animal Science and Technology, Shihezi University, China
| | - Peng Wu
- College of Life Science, Shihezi University, China
| | - Huan Zhang
- College of Animal Science and Technology, Shihezi University, China
| | - Xiaoyu Deng
- College of Animal Science and Technology, Shihezi University, China
| | - Yong Wang
- College of Animal Science and Technology, Shihezi University, China
| | - Zhongchen Ma
- College of Animal Science and Technology, Shihezi University, China
| | - Jihai Yi
- College of Animal Science and Technology, Shihezi University, China
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, China
| |
Collapse
|
8
|
Xiong Q, Yang M, Li P, Wu C. Bacteria Exploit Autophagy For Their Own Benefit. Infect Drug Resist 2019; 12:3205-3215. [PMID: 31632106 PMCID: PMC6792943 DOI: 10.2147/idr.s220376] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/25/2019] [Indexed: 01/18/2023] Open
Abstract
Autophagy is a lysosomal degradation pathway to clear long-lived proteins, protein aggregates, and damaged organelles. Certain microorganisms can be eliminated by an autophagic degradation process termed xenophagy. However, many pathogens deploy highly evolved mechanisms to evade autophagic degradation. What is more, series of pathogens have developed different strategies to exploit autophagy to ensure their survival. These bacteria could induce autophagy and/or prevent autophagosomes fusion with lysosomes through secreted effector proteins or utilizing host components, thereby maintaining the localization of the bacteria within the autophagosomes where they replicate. Here, we review the current knowledge of the mechanisms developed by the bacteria to benefit from autophagy for their survival.
Collapse
Affiliation(s)
- Qiuhong Xiong
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, People's Republic of China
| | - Min Yang
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, People's Republic of China
| | - Ping Li
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, People's Republic of China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, People's Republic of China
| |
Collapse
|
9
|
Deletion of the Type IV Secretion System Effector VceA Promotes Autophagy and Inhibits Apoptosis in Brucella-Infected Human Trophoblast Cells. Curr Microbiol 2019; 76:510-519. [PMID: 30805699 DOI: 10.1007/s00284-019-01651-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/11/2019] [Indexed: 01/18/2023]
Abstract
Brucellosis is the most common zoonotic disease that caused by intracellular parasitic bacteria Brucella. The survival and replication of Brucella in the host depend on the type IV secretion system (T4SS). The T4SS system of Brucella has many components and secreted proteins. But the mechanism helped Brucella to evade the host defense is still not clear. The objective of the present study was to investigate the effects of VceA on autophagy and apoptosis in Brucella-infected embryonic trophoblast cells. We constructed the VceA mutant strain (2308ΔVceA) and complementary strain (2308ΔVceA-C) of Brucella abortus 2308 (S2308). The human trophoblast cells (HPT-8 cells) and mice were infected by S2308, 2308ΔVceA and 2308ΔVceA-C. The cell autophagy and apoptosis were detected. The Atg5, LC3-II and Bcl-2 mRNA expression were significantly increased in 2308ΔVceA group than the S2308 group, and mRNA expression of P62 and Caspase-3 were significantly decreased than the S2308 group. Western blotting, qPCR and flow cytometry analysis showed that 2308ΔVceA promoted autophagy and inhibited apoptosis. Mouse immunohistochemistry experiments showed that P62 protein was scattered coloring and Cytochrome C protein was scarcely in 2308ΔVceA group at the myometrium. These results indicated that 2308ΔVceA promoted autophagy and inhibited apoptosis in HPT-8 cells during Brucella infection.
Collapse
|
10
|
Cerqueira DM, Gomes MTR, Silva ALN, Rungue M, Assis NRG, Guimarães ES, Morais SB, Broz P, Zamboni DS, Oliveira SC. Guanylate-binding protein 5 licenses caspase-11 for Gasdermin-D mediated host resistance to Brucella abortus infection. PLoS Pathog 2018; 14:e1007519. [PMID: 30589883 PMCID: PMC6326519 DOI: 10.1371/journal.ppat.1007519] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/09/2019] [Accepted: 12/10/2018] [Indexed: 01/18/2023] Open
Abstract
Innate immune response against Brucella abortus involves activation of Toll-like receptors (TLRs) and NOD-like receptors (NLRs). Among the NLRs involved in the recognition of B. abortus are NLRP3 and AIM2. Here, we demonstrate that B. abortus triggers non-canonical inflammasome activation dependent on caspase-11 and gasdermin-D (GSDMD). Additionally, we identify that Brucella-LPS is the ligand for caspase-11 activation. Interestingly, we determine that B. abortus is able to trigger pyroptosis leading to pore formation and cell death, and this process is dependent on caspase-11 and GSDMD but independently of caspase-1 protease activity and NLRP3. Mice lacking either caspase-11 or GSDMD were significantly more susceptible to infection with B. abortus than caspase-1 knockout or wild-type animals. Additionally, guanylate-binding proteins (GBPs) present in mouse chromosome 3 participate in the recognition of LPS by caspase-11 contributing to non-canonical inflammasome activation as observed by the response of Gbpchr3-/- BMDMs to bacterial stimulation. We further determined by siRNA knockdown that among the GBPs contained in mouse chromosome 3, GBP5 is the most important for Brucella LPS to be recognized by caspase-11 triggering IL-1β secretion and LDH release. Additionally, we observed a reduction in neutrophil, dendritic cell and macrophage influx in spleens of Casp11-/- and Gsdmd-/- compared to wild-type mice, indicating that caspase-11 and GSDMD are implicated in the recruitment and activation of immune cells during Brucella infection. Finally, depletion of neutrophils renders wild-type mice more susceptible to Brucella infection. Taken together, these data suggest that caspase-11/GSDMD-dependent pyroptosis triggered by B. abortus is important to infection restriction in vivo and contributes to immune cell recruitment and activation. Brucella abortus is the causative agent of brucellosis, a zoonotic disease that affects both humans and cattle. In humans, it is characterized by undulant fever and chronic symptoms as arthritis, endocarditis, and meningitis, while in cattle it causes abortion and infertility. Due to its difficult diagnosis and treatment, it leads to severe economic losses and human suffering. Recently, a novel non-canonical inflammasome pathway was described that involves sensing of bacterial LPS by an intracellular receptor termed caspase-11 and leads to pyroptosis and non-canonical NLRP3 inflammasome activation. Here, we show that B. abortus or its purified LPS is able to activate the non-canonical inflammasome. In this process, activated caspase-11 leads to GSDMD-dependent pyroptosis. Moreover, this pathway was dependent of IFN-induced GBP proteins, mainly GBP5. To analyze the role of caspase-1, caspase-11 and GSDMD in controlling B. abortus infection, we infected knockout (KO) mice for these molecules and we observed that caspase-11 and GSDMD KO animals were more susceptible to infection compared to wild-type animals. Casp11-/- and Gsdmd-/- animals also recruited less immune cells in mouse spleens compared to wild-type animals in response to B. abortus. Thus, caspase-11 and GSDMD are major components of the innate immune system to restrict B. abortus in vivo. This pathway of bacterial sensing by the host immune system is important to future development of drugs and vaccines that may contribute to the control of brucellosis worldwide.
Collapse
Affiliation(s)
- Daiane M Cerqueira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marco Túlio R Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alexandre L N Silva
- Departamento de Biologia Celular, Universidade de São Paulo-Ribeirão Preto, Brazil
| | - Marcella Rungue
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Natan R G Assis
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Erika S Guimarães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Suellen B Morais
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Petr Broz
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Dario S Zamboni
- Departamento de Biologia Celular, Universidade de São Paulo-Ribeirão Preto, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
| |
Collapse
|
11
|
Han G, Liu K, Li L, Li X, Zhao P. The effects of hyperbaric oxygen therapy on neuropathic pain via mitophagy in microglia. Mol Pain 2018; 13:1744806917710862. [PMID: 28580811 PMCID: PMC5464519 DOI: 10.1177/1744806917710862] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Hyperbaric oxygen (HBO) therapy has been suggested to palliate neuropathic pain, but the mechanisms involved are not well understood. This study explored the involvement of microglial mitophagy via HBO relative to neuropathic pain therapy. Materials and methods A total of 80 male Sprague Dawley rats were randomly divided into two groups: a normal group (n = 40) and a mitophagy inhibitor group (n = 40) in which the mitophagy inhibitor cyclosporin A (CsA) was administrated prior to chronic constriction injury (CCI). Groups (n = 10 rats per group) consisted of the following: control (C), sham operation (S), sciatic nerve with chronic constriction injury (CCI), and a CCI plus HBO treatment (CCI + HBO). Pain-related behaviors were evaluated using mechanical withdraw tendency and thermal withdraw latency analysis. Mitochondrial membrane potential was measured, and Western blot was employed to assess expression of NIX and BNIP3. Immunofluorescence changes in neuron protein (NESTIN) and mitochondria inner or outer layer proteins (TIM23, TOM20) were examined. Results HBO significantly ameliorated pain-related behaviors, which were downregulated by mitophagy inhibitors (P < 0.05). Mitochondrial membrane potential indexes were decreased after HBO therapy, but were reversed in the mitophagy inhibitor group (P < 0.05). HBO upregulated NIX and BNIP3 expression, which did not occur in the CCI group (P < 0.05). However, expression was reduced when mitophagy inhibitors were administered. Immunofluorescence examination showed that mitophagy in microglia was induced by CCI, which was upregulated after HBO treatment. This phenomenon was not observed in the mitophagy inhibitor group. Conclusions HBO therapy palliated CCI-induced neuropathic pain in rats by upregulating microglial mitophagy. These results could serve as guidelines to improve neuropathic pain therapy using HBO to maximize therapeutic efficiency.
Collapse
Affiliation(s)
- Guang Han
- Department of Anesthesiology, Shengjing Hospital, China Medical University
| | - Kun Liu
- Department of Anesthesiology, Shengjing Hospital, China Medical University
| | - Lu Li
- Department of Anesthesiology, Shengjing Hospital, China Medical University
| | - Xingyue Li
- Department of Anesthesiology, Shengjing Hospital, China Medical University
| | | |
Collapse
|
12
|
The relationship between caspase-1 related inflammasome expression and serum inflammatory cytokine levels during acute brucellosis. North Clin Istanb 2018; 6:117-123. [PMID: 31297476 PMCID: PMC6593912 DOI: 10.14744/nci.2018.96992] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/22/2018] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Brucellosis is a zoonotic disease caused by Brucella in domestic and wild animals. It also causes systemic diseases with the involvement of different parts of the human body. An efficient innate immune response is crucial to cure brucellosis with optimum antibiotic treatment. The inflammasomes are innate immune system receptors and sensors that regulate the activation of cysteine-dependent aspartate specific protease-1 (caspase-1) and caspase-1-induced cell death process known as pyroptosis. The aim of the present study was to investigate the expression levels of CASPASE-1 and associated inflammasomes AIM2, NLRP3, and NLRC4 to analyze their relationship with the inflammatory cytokine interleukin (IL)-1β, IL-18, and interferon-gamma (IFN-γ) in peripheral blood samples of patients with acute brucellosis with healthy controls. METHODS Peripheral blood samples were obtained from 20 healthy volunteers and 20 patients with acute brucellosis. RNA and serum samples were isolated to examine the expression levels of AIM2, NLRP3, NLRC4, and CASPASE-1 by real-time polymerase chain reaction, and IL-1β, IL-18, and IFN-γ were measured by enzyme-linked immunosorbent assay. RESULTS In the acute brucellosis group, AIM2 and NLRC4 expressions were significantly higher than in healthy volunteers. A significant increase on caspase-1 expression in patients with acute brucellosis was not observed. Serum IL-18 and IFN-γ levels were significantly higher in patients with acute brucellosis than in healthy controls. CONCLUSION Caspase-1-related inflammasomes are sufficiently activated to induce the secretion of cytokines, such as IFN-γ and IL-18, to induce cellular immune response. Caspase-1 activation level should be investigated at different periods of disease in a group with high number of patients to understand the role of pyroptosis and caspase-1 in brucellosis.
Collapse
|
13
|
Mitochondrial fragmentation affects neither the sensitivity to TNFα-induced apoptosis of Brucella-infected cells nor the intracellular replication of the bacteria. Sci Rep 2018; 8:5173. [PMID: 29581535 PMCID: PMC5979954 DOI: 10.1038/s41598-018-23483-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 03/13/2018] [Indexed: 01/18/2023] Open
Abstract
Mitochondria are complex organelles that participate in many cellular functions, ranging from ATP production to immune responses against viruses and bacteria. This integration of a plethora of functions within a single organelle makes mitochondria a very attractive target to manipulate for intracellular pathogens. We characterised the crosstalk that exists between Brucella abortus, the causative agent of brucellosis, and the mitochondria of infected cells. Brucella replicates in a compartment derived from the endoplasmic reticulum (ER) and modulates ER functionality by activating the unfolded protein response. However, the impact of Brucella on the mitochondrial population of infected cells still requires a systematic study. We observed physical contacts between Brucella containing vacuoles and mitochondria. We also found that B. abortus replication is independent of mitochondrial oxidative phosphorylation and that mitochondrial reactive oxygen species do not participate to the control of B. abortus infection in vitro. We demonstrated that B. abortus and B. melitensis induce a drastic mitochondrial fragmentation at 48 hours post-infection in different cell types, including myeloid and non-myeloid cells. This fragmentation is DRP1-independent and might be caused by a deficit of mitochondrial fusion. However, mitochondrial fragmentation does not change neither Brucella replication efficiency, nor the susceptibility of infected cells to TNFα-induced apoptosis.
Collapse
|
14
|
Leptospira interrogans infection leads to IL-1β and IL-18 secretion from a human macrophage cell line through reactive oxygen species and cathepsin B mediated-NLRP3 inflammasome activation. Microbes Infect 2018; 20:254-260. [PMID: 29432801 DOI: 10.1016/j.micinf.2018.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/29/2017] [Accepted: 01/26/2018] [Indexed: 12/17/2022]
Abstract
Leptospirosis is a worldwide zoonosis caused by spirochetes from the genus Leptospira. Although there is a large diversity of clinical signs and symptoms, a severe inflammatory response is common to all leptospirosis patients. The mechanism of IL-1β secretion during Leptospira infection has been previously studied in mouse macrophages. However, the outcome of Leptospira infection is very different in human and murine macrophages, and the mechanisms responsible for IL-1β secretion in human macrophages had not been investigated. This study therefore examines the effects of Leptospira interrogans infection on inflammasome activation and proinflammatory cytokine expression in human macrophages. Increased mRNA and protein expression of NLRP3 was observed by real time RT-PCR and flow cytometry at 1 h after co-cultivation. Enzyme-linked immunosorbent assay (ELISA) determination showed that IL-1β and IL-18 are released in the culture supernatants at 1 h after cultivation. The inhibition assay showed that glybenclamide (a K+ efflux inhibitor that blocks NLRP3 inflammasome activation) and N-benzyloxycarbony-Val-Ala-Asp (O-methyl)-fluoromethylketone (Z-VAD-FMK; a caspase-1 inhibitor) and NLRP3 depletion with siRNAs reduced the levels of IL-1β and IL-18 release. Moreover, the levels of IL-1β and IL-18 production decreased in CA-074 (a cathepsin B inhibitor) and NAC (an anti-oxidant) pretreated human macrophages, compared to untreated controls. This study suggests that L. interrogans infection leads to reactive oxygen species (ROS)- and cathepsin B-dependent NLRP3 inflammasome activation, which subsequently mediates caspase-1 activation and IL-1β and IL-18 release.
Collapse
|
15
|
Brucella abortus Induces a Warburg Shift in Host Metabolism That Is Linked to Enhanced Intracellular Survival of the Pathogen. J Bacteriol 2017; 199:JB.00227-17. [PMID: 28559292 DOI: 10.1128/jb.00227-17] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/18/2017] [Indexed: 01/29/2023] Open
Abstract
Intracellular bacterial pathogens exploit host cell resources to replicate and survive inside the host. Targeting these host systems is one promising approach to developing novel antimicrobials to treat intracellular infections. We show that human macrophage-like cells infected with Brucella abortus undergo a metabolic shift characterized by attenuated tricarboxylic acid cycle metabolism, reduced amino acid consumption, altered mitochondrial localization, and increased lactate production. This shift to an aerobic glycolytic state resembles the Warburg effect, a change in energy production that is well described in cancer cells and also occurs in activated inflammatory cells. B. abortus efficiently uses lactic acid as its sole carbon and energy source and requires the ability to metabolize lactate for normal survival in human macrophage-like cells. We demonstrate that chemical inhibitors of host glycolysis and lactate production do not affect in vitro growth of B. abortus in axenic culture but decrease its survival in the intracellular niche. Our data support a model in which infection shifts host metabolism to a Warburg-like state, and B. abortus uses this change in metabolism to promote intracellular survival. Pharmacological perturbation of these features of host cell metabolism may be a useful strategy to inhibit infection by intracellular pathogens.IMPORTANCEBrucella spp. are intracellular bacterial pathogens that cause disease in a range of mammals, including livestock. Transmission from livestock to humans is common and can lead to chronic human disease. Human macrophage-like cells infected with Brucella abortus undergo a Warburg-like metabolic shift to an aerobic glycolytic state where the host cells produce lactic acid and have reduced amino acid catabolism. We provide evidence that the pathogen can exploit this change in host metabolism to support growth and survival in the intracellular niche. Drugs that inhibit this shift in host cell metabolism inhibit intracellular replication and decrease the survival of B. abortus in an in vitro infection model; these drugs may be broadly useful therapeutics for intracellular infections.
Collapse
|
16
|
Li T, Huang M, Xiao H, Zhang G, Ding J, Wu P, Zhang H, Sheng J, Chen C. Selection and characterization of specific nanobody against bovine virus diarrhea virus (BVDV) E2 protein. PLoS One 2017; 12:e0178469. [PMID: 28582444 PMCID: PMC5459339 DOI: 10.1371/journal.pone.0178469] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 05/13/2017] [Indexed: 01/15/2023] Open
Abstract
Bovine viral diarrhea-mucosal disease (BVD-MD) is caused by bovine viral diarrhea virus (BVDV), and results in abortion, stillbirth, and fetal malformation in cows. Here, we constructed the phage display vector pCANTAB 5E-VHH and then transformed it into Escherichia coli TG1-competent cells, to construct an initial anti-BVDV nanobody gene library. We obtained a BVDV-E2 antigen epitope bait protein by prokaryotic expression using the nucleotide sequence of the E2 gene of the BVDV-NADL strain published in GenBank. Phage display was used to screen the anti-BVDV nanobody gene library. We successfully constructed a high quality phage display nanobody library, with an initial library capacity of 4.32×105. After the rescue of helper phage, the titer of the phage display nanobody library was 1.3×1011. The BVDV-E2 protein was then expressed in Escherichia coli (DE3), and a 49.5 kDa band was observed with SDS-PAGE analysis that was consistent with the expected nanobody size. Thus, we were able to isolate one nanobody that exhibits high affinity and specificity against BVDV using phage display techniques. This isolated nanobody was then used in Enzyme Linked Immunosorbent Assay and qRT-PCR, and ELISA analyses of BVDV infection of MDBK cells indicated that the nanobodies exhibited good antiviral effect.
Collapse
Affiliation(s)
- Tiansen Li
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Meiling Huang
- Shihezi University Library, Shihezi University, Shihezi, Xinjiang, China
| | - Hongran Xiao
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Guoqi Zhang
- College of Life Science, Shihezi University, Shihezi, Xinjiang, China
| | - Jinhua Ding
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Peng Wu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Hui Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Jinliang Sheng
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|