1
|
Prabhakar YK, Skariah S, Shanmugam G, Shome R. Molecular epidemiology, immunobiology, genomics and proteomics insights into bovine brucellosis. Vet Microbiol 2025; 305:110505. [PMID: 40233684 DOI: 10.1016/j.vetmic.2025.110505] [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: 11/06/2024] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
Brucella species are intracellular Gram-negative bacteria that cause brucellosis, a global zoonosis that impacts cattle productivity and public health. Both cattle and buffaloes are susceptible to bovine brucellosis, which can lead to severe degenerative changes in uterine mucosa of non-pregnant animals, including ulcerative endometritis and fibrosis. Vasculitis, localized coagulative necrosis, and ulceration of the uterine mucosa have all been reported in pregnant animals. Male testicles get inflamed due to Brucella, which results in infertility. This review article covers the molecular epidemiology, pathophysiology, immunobiology, genomics, and proteomics of Brucella, with an emphasis on novel discoveries and more recent research, especially on bovine brucellosis. The integration of molecular pathology and sero-prevalence data provide the insights into epidemiology, transmission dynamics, and genetic diversity of bovine brucellosis. The immunobiological response studies of brucellosis have provided insights into the tactics employed by Brucella to infect host cells and elude immune responses. Proteomics was utilized to find biomarkers for both acute and chronic brucellosis, which resulted in the identification of proteins with differential expression linked to immune response, inflammation, and extracellular matrix modulation. The genetic diversity, virulence factors, and evolution of Brucella strains were mostly investigated using genomics. The genomic makeup and architecture of Brucella isolates were examined using whole-genome sequencing, which revealed genetic markers linked to pathogenicity and drug resistance. This review provides possible treatment targets, diagnostic biomarkers, and vaccine candidates, contributing to molecular understanding of bovine brucellosis.
Collapse
Affiliation(s)
- Y K Prabhakar
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Yelahanka, Bengaluru 560 064, India
| | - Somy Skariah
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Yelahanka, Bengaluru 560 064, India
| | - G Shanmugam
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Yelahanka, Bengaluru 560 064, India
| | - Rajeswari Shome
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Yelahanka, Bengaluru 560 064, India.
| |
Collapse
|
2
|
Hu R, Zhang Q, Wang W, Ren W, Yao M, Xu Y, Zhang H, Sheng J, Wang Y, Chen C, Ma Z. Brucella inactivated vaccine elicits immunity against B. melitensis infection in mice and guinea pigs. Biomed Pharmacother 2025; 187:118077. [PMID: 40280033 DOI: 10.1016/j.biopha.2025.118077] [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: 02/10/2025] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Brucellosis is a zoonotic disease that poses a significant threat to both animal husbandry and public health. Currently available vaccines for brucellosis are all live attenuated forms, which carry the risk of potential infectivity and provide a relatively limited range of protection. In contrast, inactivated vaccines are perceived to exhibit poor protective efficacy and fail to elicit effective cellular immunity. This study aimed to comprehensively evaluate the efficacy of the Brucella inactivated vaccine (CF) with the objective of developing a safer and more effective candidate for brucellosis vaccination. METHODS Firstly, we evaluated the safety of CF in mice. Subsequently, we immunized mice with CF using various doses and methods, determining the optimal immunization dose and method through challenge testing. We evaluated the vaccine's immunogenicity by detecting the cellular and humoral immune levels induced by CF in mice, and assessed the vaccine's protective effect based on the post challenge organ bacterial load. Additionally, we evaluated the protective effects of dual doses and secondary immunization in guinea pigs. RESULTS The results indicate that CF is safe and non-toxic; it induced significant increases in specific IgG antibody levels against Brucella during the early stages and markedly enhanced the T cell immune response, thereby promoting a Th1-biased immune response in mice. Following the challenge, CF demonstrated protective efficacy comparable to that of the S2 vaccine against B. melitensis biovar 3 infection in mice. CF-immunized guinea pigs were able to resist infection by B. melitensis M28 and B. melitensis biovar 3. CONCLUSIONS In summary, CF significantly induces both humoral and cellular immunity in mice. This study reports for the first time that a safe and effective inactivated Brucella vaccine (CF) can induce cellular immune responses and effectively prevent animal brucellosis.
Collapse
Affiliation(s)
- Ruirui Hu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China; College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Qianyi Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Wenjia Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Wenhao Ren
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Mengxin Yao
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Yimei Xu
- Center for Disease Control and Prevention of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830002, China.
| | - Hui Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Jinliang Sheng
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Yong Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Zhongchen Ma
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| |
Collapse
|
3
|
Caudill MT, Stoyanof ST, Caswell CC. Quorum sensing LuxR proteins VjbR and BabR jointly regulate Brucella abortus survival during infection. J Bacteriol 2025; 207:e0052724. [PMID: 40013834 PMCID: PMC11925318 DOI: 10.1128/jb.00527-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/02/2025] [Indexed: 02/28/2025] Open
Abstract
Brucella abortus maintains an N-acetyl homoserine lactone quorum sensing system that consists of two LuxR proteins, VjbR and BabR, as well as two signals, dodecanoyl (C12 AHL) and 3-oxododecanoyl (3-OXO-C12 AHL) homoserine lactone. This system regulates major virulence factors that influence the bacteria's survival during infection. We generated the first strain that lacks both LuxR proteins and found a synergistic interaction for survival in the chronic infection C57BL/6 mouse model. Transcriptomic analyses of the ∆vjbR∆babR double-deletion strain, as well as the cognate single-deletion strains, in a rich medium with vehicle control or supplemented with an AHL signal revealed large-scale genetic dysregulation in all conditions. Moreover, the double mutant maintained a limited response to AHL, even in the absence of the LuxR proteins. We additionally found that quorum sensing regulates the denitrification pathway but found no in vitro differences in the ability of quorum sensing deletion strains to clear nitric oxide stress or grow under anoxic denitrifying conditions. Finally, we confirmed that BabR autoregulates its own expression, and that VjbR mildly represses BabR expression. Altogether, these experiments help further characterize the Brucella quorum sensing systems and indicate that further attention should be given to the joint interactions between VjbR and BabR in controlling virulence.IMPORTANCEBrucella abortus is a zoonotic bacterial pathogen that uses its quorum sensing to survive within hosts. This study further characterizes that system and indicates important future lines of inquiry. We found that both quorum sensing proteins, VjbR and BabR, coordinate to maintain survival, as well as document that both quorum sensing systems appear physiologically active.
Collapse
Affiliation(s)
- Mitchell T. Caudill
- Center for One Health Research, Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
| | - S. Tristan Stoyanof
- Center for One Health Research, Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
| | - Clayton C. Caswell
- Center for One Health Research, Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
| |
Collapse
|
4
|
Bányász B, Antal J, Dénes B. Ghostbuster-A Western Blot-Based Panel Method to Resolve False-Positive Brucellosis Serology Test Results. Microorganisms 2025; 13:574. [PMID: 40142466 PMCID: PMC11944415 DOI: 10.3390/microorganisms13030574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
False-positive serologic results (FPSRs) of brucellosis occur from time to time in various livestock with all the consequences (quarantine, compulsory slaughter, etc.) that follow true-positive laboratory results. A method based on the Polyacrylamide Gel Electrophoresis/Western Blot of a protein panel for resolving the FPSRs in the diagnosis of brucellosis was developed. Within the context of limited positive serum sample availability in Europe, the method successfully discriminates Brucella-positive sera from samples containing antibodies raised against infections caused by other Gram-negative bacteria causing FPSRs. An average CV% of 1.36 was determined for both repeatability and reproducibility for the whole separation mw range, and the test achieved 1.00 Diagnostic Sensitivity and 1.00 Diagnostic Specificity. The method with pre-prepared WB panels provides a rapid (less than 3 h), easily standardizable, and validatable alternative to existing confirmation methods. The whole WB process of the Brucella proteins and the subsequent densitometry can be accomplished with commercially available equipment, ready-to-use reagents, and open-source software, providing cost-effectiveness. The results of this study could attract broader attention, since molecular species in the 35.0-75.0 kDa range can serve as antigens in Brucella serology and the same fraction can be considered in the development of synthetic Brucella vaccines.
Collapse
Affiliation(s)
| | - József Antal
- Omixon Biocomputing Ltd., 1117 Budapest, Hungary
| | - Béla Dénes
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine Budapest, 1143 Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| |
Collapse
|
5
|
Yin Y, Tian M, Zhang G, Ding C, Yu S. A novel Brucella T4SS effector RS15060 acts on bacterial morphology, lipopolysaccharide core synthesis and host proinflammatory responses, which is beneficial for Brucella melitensis virulence. Microbiol Res 2025; 292:128015. [PMID: 39689431 DOI: 10.1016/j.micres.2024.128015] [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: 09/22/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
Brucella relies on the type IV secretion system (T4SS) to establish replication niches within host cells. However, the Brucella T4SS effectors and their functions have not been fully identified. In this study, we investigated the function of Brucella RS15060, a novel T4SS effector discovered in our previous study, on the bacterial biological characteristics and pathogenesis by construction of the gene deletion and complementation strains. We found that deletion of the rs15060 gene weakened abilities of Brucella to replicate within host cells and establish chronic infection in mice but enhanced abilities to adhere/invade HeLa cells and evade lysosomal degradation in the early stage of infection. In addition, the rs15060 deletion Brucella strain showed significant changes in bacterial shape, cell wall thickness, and sensitivity to bactericidal factors. Furthermore, the rs15060 deletion strain showed an increased synthesis of bacterial lipopolysaccharide core and induced a stronger host's inflammatory response. The Brucella rs15060 complementation strain restored the altered biological characteristics. Moreover, BLASTP prediction and 3D structure simulation revealed that the Brucella RS15060 contains NAD(P)-binding and active motifs in structure, which are important for proteins to exert NAD dependent epimerase/dehydratase activity. The complementation strain with mutation on NAD(P)-binding and/or active motifs of RS15060 did not restore the altered characteristics, suggesting that the Brucella RS15060 is a potential NAD dependent epimerase/dehydratase, and the predicted NAD(P)-binding and/or active motifs play an important role on bacterial cell wall and LPS core synthesis, which is crucial for maintaining bacterial morphology and exerting virulence.
Collapse
Affiliation(s)
- Yi Yin
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Mingxing Tian
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Guangdong Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China; Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Shengqing Yu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China; Jiangsu Agri-Animal Husbandry Vocational College, Veterinary Bio-Pharmaceutical, Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Taizhou, Jiangsu 225309, China.
| |
Collapse
|
6
|
Zhu D, Guo J, Deng X, Li M, Wang Y, Wang Z, Sun Z, Cao S, Zhao T, Xu Y, Liu L, Zhang H. Brucella abortus transcriptional regulator ArsR6 inhibits host pyroptosis via BAB_RS28760 by triggering the endoplasmic reticulum stress pathway. Int Immunopharmacol 2025; 147:114001. [PMID: 39787758 DOI: 10.1016/j.intimp.2024.114001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/15/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Pyroptosis, which is accompanied by inflammatory responses, is critical for pathogen clearance. However, the mechanism through which Brucella evades host pyroptosis remains unclear. The transcriptional regulator ArsR6 maintains bacterial intracellular homeostasis and possibly influences host cell death. However, whether ArsR6 acts on cellular pyroptosis is unknown. Therefore, we investigated pathogen-host interactions within macrophages infected with Brucella abortus (B. abortus), and found that ArsR6 is crucial for inhibiting host cell pyroptosis after B. abortus infection. The downstream target gene, BAB_RS28760 of ArsR6 was screened using chromatin immunoprecipitation sequencing. BAB_RS28760 belongs to the BA14K protein family and is strongly immunoreactive and induces humoral and cellular immune responses in the host during infection. Deleting ArsR6 in B. abortuspromotes pyroptosis and enhancs the intracellular survival of B. abortus. In addition, ArsR6 negatively regulated its target gene BAB_RS28760, whereas BAB_RS28760 deletion downregulated cellular pyroptosis by inhibiting endoplasmic reticulum stress and decreasing the intracellular survival of B. abortus. Our results reveal for the first time that Brucella ArsR6 reduces endoplasmic reticulum stress activation by negatively regulating its downstream target genes, thus inhibiting host cell pyroptosis. Our study provides new insights into the pathogenic mechanisms of Brucella, which can provide potential selectivity for the development of anti-Brucella therapies.
Collapse
Affiliation(s)
- Dexin Zhu
- College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Jia Guo
- College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Xingmei Deng
- College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Min Li
- Changji City Communist Youth League Committee, Changji, China.
| | - Yong Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Zhen Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Zhihua Sun
- College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Shuzhu Cao
- College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Tianyi Zhao
- College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Yimei Xu
- The Center for Disease Control and Prevention of Xinjiang Uygur Autonomous Region, Urumqi, China.
| | - Liangbo Liu
- College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Hui Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, China.
| |
Collapse
|
7
|
He J, Yin S, Deng X, Ma Z, Zhang H, Miao Y, Yi J, Chen C, Zhang J. The effector protein BspE affects Brucella survival by regulating the inflammatory response and apoptosis. Int Immunopharmacol 2025; 144:113576. [PMID: 39566384 DOI: 10.1016/j.intimp.2024.113576] [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: 08/11/2024] [Revised: 10/24/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024]
Abstract
Brucella T4SS secretes numerous effector proteins to disrupt host immune responses and apoptosis, enabling long-term survival. One such effector protein is BspE, whose role remains largely unknown. In this study, we demonstrated that BspE promotes the growth of Brucella, enhances its survival in macrophages, and affects the release of macrophage inflammatory factors. Furthermore, BspE facilitates Brucella colonization and pathological damage in mice. Our findings reveal that BspE can be translated in the host cell nucleus, where it interacts with the host RNA-binding protein PCBP1 to promote Brucella replication in macrophages. Knockdown of PCBP1 affects BspE-mediated proliferation of Brucella in macrophages. Furthermore, the BspE-PCBP1 interaction hinders P53 signaling and inhibits macrophage apoptosis. Although this interaction affects inflammatory cytokines, it does not significantly involve the NF-κB pathway. These findings contribute to a better understanding of how the Brucella effector protein BspE regulates host immune responses and apoptosis to influence its own survival.
Collapse
Affiliation(s)
- Jinke He
- 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 Xinjiang 832003, China; Department of Basic Medicine, Xinjiang Second Medical College, Kelamayi 834000, Xinjiang, China
| | - Shuanghong Yin
- Guizhou Provincial Key Laboratory for Biodiversity Conservation and Utilization in the Fanjing Mountain Region, Tongren University, Tongren 554300, Guizhou, China; School of Sports and Health Science, Tongren University, Tongren 554300, China
| | - Xiaoyu Deng
- 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 Xinjiang 832003, China; Department of Basic Medicine, Hunan University of Medicine, huaihua 418000, Hunan, China
| | - Zhongchen Ma
- 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 Xinjiang 832003, China
| | - Huan Zhang
- 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 Xinjiang 832003, China
| | - Yuhe Miao
- Fujian Sunvet Biological Technology Co., Ltd, Nanping 354100, Fujian, China
| | - Jihai Yi
- 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 Xinjiang 832003, China.
| | - Chuangfu Chen
- 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 Xinjiang 832003, China.
| | - Junbo Zhang
- Guizhou Provincial Key Laboratory for Biodiversity Conservation and Utilization in the Fanjing Mountain Region, Tongren University, Tongren 554300, Guizhou, China; College of A&F Engineering and Planning, Tongren University, Tongren 554300, Guizhou, China.
| |
Collapse
|
8
|
Pavlik P, Velecka E, Spidlova P. Breaking the cellular defense: the role of autophagy evasion in Francisella virulence. Front Cell Infect Microbiol 2024; 14:1523597. [PMID: 39776438 PMCID: PMC11703736 DOI: 10.3389/fcimb.2024.1523597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Many pathogens have evolved sophisticated strategies to evade autophagy, a crucial cellular defense mechanism that typically targets and degrades invading microorganisms. By subverting or inhibiting autophagy, these pathogens can create a more favorable environment for their replication and survival within the host. For instance, some bacteria secrete factors that block autophagosome formation, while others might escape from autophagosomes before degradation. These evasion tactics are critical for the pathogens' ability to establish and maintain infections. Understanding the mechanisms by which pathogens avoid autophagy is crucial for developing new therapeutic strategies, as enhancing autophagy could bolster the host's immune response and aid in the elimination of pathogenic bacteria. Francisella tularensis can manipulate host cell pathways to prevent its detection and destruction by autophagy, thereby enhancing its virulence. Given the potential for F. tularensis to be used as a bioterrorism agent due to its high infectivity and ability to cause severe disease, research into how this pathogen evades autophagy is of critical importance. By unraveling these mechanisms, new therapeutic approaches could be developed to enhance autophagic responses and strengthen host defense against this and other similarly evasive pathogens.
Collapse
Affiliation(s)
- Pavla Pavlik
- Department of Molecular Pathology and Biology, Military Faculty of Medicine, University of Defence, Hradec Kralove, Czechia
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czechia
| | - Eva Velecka
- Department of Molecular Pathology and Biology, Military Faculty of Medicine, University of Defence, Hradec Kralove, Czechia
| | - Petra Spidlova
- Department of Molecular Pathology and Biology, Military Faculty of Medicine, University of Defence, Hradec Kralove, Czechia
| |
Collapse
|
9
|
Li J, Yuan N, Zhai Y, Wang M, Hao M, Liu X, Zhou D, Liu W, Jin Y, Wang A. Protein disulfide isomerase A4 binds to Brucella BtpB and mediates intracellular NAD +/NADH metabolism in RAW264.7 cells. Int Immunopharmacol 2024; 142:113046. [PMID: 39226825 DOI: 10.1016/j.intimp.2024.113046] [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: 07/06/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
The Toll/interleukin-1 receptor (TIR) signaling domain is distributed widely in mammalian Toll-like receptors and adaptors, plant nucleotide-binding leucine-rich repeat receptors, and specific bacterial virulence proteins. Proteins that possess TIR domain exhibit NADase activity which is distinct from the canonical signaling function of these domains. However, the effects of bacterial TIR domain proteins on host metabolic switches and the underlying mechanism of NADase activity in these proteins remain unclear. Here, we utilized Brucella TIR domain-containing type IV secretion system effector protein, BtpB, to explore the mechanism of NADase activity in host cells. We showed that using ectopic expression BtpB not only generates depletion of NAD+ but also loss of NADH and ATP in RAW264.7 macrophage cells. Moreover, immunoprecipitation-mass spectrometry, co-immunoprecipitation, and confocal microscope assays revealed that BtpB interacted with host protein disulfide isomerase A4 (PDIA4). The Brucella mutant strain deleted the gene for BtpB, significantly decreased PDIA4 expression. Furthermore, our data revealed that PDIA4 played an important role in regulating intracellular NAD+/NADH levels in macrophages, and PDIA4 overexpression restored the decline of intracellular NAD+ and NADH levels induced by Brucella BtpB. The results provide new insights into the metabolic regulatory activity of TIR domain proteins in the critical human and animal pathogen Brucella.
Collapse
Affiliation(s)
- Junmei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Department of Veterinary Medicine, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Ningqiu Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yunyi Zhai
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Minghui Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Mingyue Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Xiaofang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Dong Zhou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Wei Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, China; Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
10
|
Zhi F, Liu K, Geng H, Su M, Xu J, Fu L, Ma K, Gao P, Yuan L, Chu Y. Copper sensing transcription factor ArsR2 regulates VjbR to sustain virulence in Brucella abortus. Emerg Microbes Infect 2024; 13:2406274. [PMID: 39295505 PMCID: PMC11425708 DOI: 10.1080/22221751.2024.2406274] [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: 05/24/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 09/21/2024]
Abstract
Brucellosis, caused by the intracellular pathogen Brucella, is a major zoonotic infection that promotes reproductive disease in domestic animals and chronic debilitating conditions in humans. The ArsR family of transcriptional regulators plays key roles in diverse cellular processes, including metal ion homeostasis, responding to adverse conditions, and virulence. However, little is known about the function of ArsR family members in Brucella. Here, we identified ArsR2 as a nonclassical member of the family that lacks autoregulatory function, but which nevertheless plays a vital role in maintaining copper homeostasis in B. abortus. ArsR2 is a global regulator of 241 genes, including those involved in the VirB type IV secretion system (T4SS). Significantly, ArsR2 regulates T4SS production in B. abortus by targeting VjbR which encodes a LuxR-type family transcriptional regulator. Moreover, copper modulates transcriptional activity of ArsR2, but not of VjbR. Furthermore, deletion of arsR2 attenuated virulence in a mouse model. Collectively, these findings enhance understanding of the mechanism by which ArsR proteins regulate virulence gene expression in pathogenic Brucella species.
Collapse
Affiliation(s)
- Feijie Zhi
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Kemeng Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Hao Geng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Mengru Su
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Jian Xu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Lei Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Ke Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Pengcheng Gao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Lvfeng Yuan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - YueFeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| |
Collapse
|
11
|
Allam NAT, Abdelsalam ME, Elsharkawy HI, Kandil MM, Mohamed AMM, Ali F, Gebely MA, Nour SY, Sedky D, El-Gawad MEHA, Zaki HM, Al-Gallas N, Aboelmaaty AM, Sobhy MM, Ata NS, Abdel-Hamid MS, Badawy GA. Comprehensive epidemiological evaluation of ruminant brucellosis and associated risk factors in some Egyptian Governorates. Vet World 2024; 17:2780-2796. [PMID: 39897353 PMCID: PMC11784036 DOI: 10.14202/vetworld.2024.2780-2796] [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: 06/20/2024] [Accepted: 10/16/2024] [Indexed: 02/04/2025] Open
Abstract
Background and Aim Brucellosis contributes to significant economic losses due to abortion, weak newborns, infertility, and up to 20% reductions in milk yield in carrier animals. This study aimed to estimate the prevalence of ruminant brucellosis in six Egyptian governorates. This study aimed to estimate the prevalence of ruminant brucellosis and evaluate the risk factors regarding the epidemiological status, highlighting the importance of early carrier detection for the success of control programs. Materials and Methods A total of 3000 ruminants were investigated. Blood and serum samples were collected for routine hemato-biochemical analysis (complete blood picture and metabolic panel). In addition, genotoxicity analysis was performed, whereas tissue samples were collected for histopathological analysis. The buffered acidified plate antigen test (BAPAT), Rose Bengal plate test (RBPT), and complement fixation test (CFT) were used for serological diagnosis of brucellosis. The obtained bacterial colonies were typed using Brucella abortus-, melitensis-, ovis-, and suis-polymerase chain reaction (AMOS-PCR), depending on the variability of the IS711 fragment among Brucella spp. Serum trace elements, oxidative stress, and acute phase proteins were compared according to body condition score (BCS) and clinical condition images within the study population. Results Mastitis and abortion were the key recorded symptoms (9.966%, 299/3000 and 6%, 180/3000, respectively); however, symptomless individuals were predominant (82.9%, 2487/3000). Blood lymphocytosis was prominent even in asymptomatic animals. Nutritional and food conversion conditions were defined as low, moderate, or high BCS. Brucella overall seropositivity by BAPAT, RBPT, and CFT was 6.1% (182/3000), 5.6% (168/3000), and 5.1% (154/3000) in ruminant species within the included governorates, respectively. Upon diagnosis, 154 seropositive cases developed 93 bacterial isolates and a 731-bp PCR fragment whose sequences confirmed Brucella melitensis biovar 3. Serum metabolic and biochemical profiles, acute phase proteins, trace elements, and oxidative stress concentrations were indicative of loss of functionality in the liver and kidneys, malnutrition and malabsorption syndrome, and DNA damage, particularly in the low-BCS groups (p < 0.0001). Granulomatous lesions were most prominent in the lymph nodes, spleen, uterus, and udder of the dams, while placental multifocal necrosis with thrombosis was recorded in aborted fetuses. There were 8 types of chromosomal aberrations detected in peripheral white blood cells. The highest frequency was for dicentric aberrations 0.025% (25/1000), whereas the lowest 0.009% (9/1000) was for acentric, ring, fusion, and polyploidy. The difference between species was significant for BCS; 14.2% in low-BCS cattle and camels and 8.4% in high-BCS buffaloes. Conclusion B. melitensis biovar 3 is prevalent in Egypt. Mixed-rearing systems are the main risk factors for interspecies transmission among ruminants. The difficulty in accurately diagnosing all infected animals, particularly carriers, is a major limitation of eradication and control programs. Different biomarkers could be indicators and/or sensors for performance and/or infectivity conditions in animal herds; however, they require further optimization. Early detection using molecular technologies, highly descriptive, quantitative, sensitive, and specific methods, as alternatives to serological diagnosis (CFT, BAPAT, and RBT), is urgently needed to enhance the efficiency of brucellosis-specific prophylaxis. Such a comprehensive procedure is the World Organization for Animal Health dependent decision.
Collapse
Affiliation(s)
- Nesreen Allam Tantawy Allam
- Department of Parasitology and Animal Diseases, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Cairo, Egypt
| | - Mahinour Ezzeldin Abdelsalam
- Department of General Biology, Center of Basic Sciences, Misr University for Science and Technology, Al Motamayez District, 6 of October, Giza, Egypt
| | - Hend I. Elsharkawy
- Brucella Reference Laboratory, Animal Health Research Institute, Agricultural Research Center, P.O. Box 264-Giza, Cairo, 12618, Egypt
| | - Mai Mohamed Kandil
- Department of Microbiology and Immunology, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Cairo, Egypt
| | - Amany Mohamed Mohamed Mohamed
- Department of Parasitology and Animal Diseases, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Cairo, Egypt
| | - Fatma Ali
- Department of Physiology, Faculty of Veterinary Medicine, Aswan University, Egypt
| | - Mohamed A. Gebely
- Department of Parasitology and Animal Diseases, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Cairo, Egypt
| | - Safaa Y. Nour
- Animal Medicine Department, Faculty of Veterinary Medicine, Aswan University, Egypt
| | - Doaa Sedky
- Department of Parasitology and Animal Diseases, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Cairo, Egypt
| | | | - Hoda M. Zaki
- Brucella Reference Laboratory, Animal Health Research Institute, Agricultural Research Center, P.O. Box 264-Giza, Cairo, 12618, Egypt
| | - Nazek Al-Gallas
- Department of Biology, Faculty of Science, University of Hafr Al-Batin, P.O. Box: 1803, Hafr Al-Batin, 31991, Kingdom of Saudi Arabia
- Water and Food Control Lab., National Center of Salmonella, Shigella, Vibrio, E. coli-Enteropathogens, Institute Pasteur de Tunis, Tunis
| | - Amal M. Aboelmaaty
- Department of Reproduction, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Cairo, Egypt
| | - Mona Mohamed Sobhy
- Department of Reproductive Diseases, Animal Reproduction Research Institute, Agricultural Research Center, Al-Haram, Giza, Egypt
| | - Nagwa Sayed Ata
- Department of Microbiology and Immunology, Veterinary Research Institute, National Research Centre, 33 El Buhouth Street, Dokki, P.O. Box: 12622, Cairo, Egypt
| | - Marwa Salah Abdel-Hamid
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City
| | - Ghada A. Badawy
- Department of Botany, Faculty of Science, El-Fayoum University, Fayoum, 63514, Egypt
- Department of Biology, Faculty of Science, University of Tabuk, Umluj 46429, Kingdom of Saudi Arabia
| |
Collapse
|
12
|
Mallik SR, Joshi K, Radhakrishnan GK. The arginine/ornithine binding protein ArgT plays an essential role in Brucella neotomae/ Brucella melitensis to prevent intracellular killing and contribute to chronic persistence in the host. Virulence 2024; 15:2421983. [PMID: 39463062 PMCID: PMC11540086 DOI: 10.1080/21505594.2024.2421983] [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: 02/14/2024] [Revised: 09/13/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
Brucella species are facultative intracellular bacterial pathogens that cause the contagious zoonotic disease, brucellosis. Brucella spp. infect a wide range of animals, including livestock, wild animals, and marine mammals. Compared with other invasive bacterial pathogens, partial information is available on the virulence factors of Brucella that enable them to survive in the host. Here, we performed transposon-based random mutagenesis of B. neotomae and identified the arginine/ornithine binding protein, ArgT, as one of the crucial virulence determinants of Brucella. Deleting ArgT from B. neotomae or B. melitensis resulted in its attenuation in macrophages, which was restored upon complementation with an ArgT expression plasmid. We observed that macrophages infected with ΔArgT-B. neotomae produced elevated levels of NO due to the inability of these mutants to deplete the host intracellular arginine through their importer. Furthermore, defective survival of ΔArgT B. neotomae and B. melitensis was observed in the infected mice, which correlated with enhanced NO production in the mice. Our studies revealed that ArgT plays a vital role in preventing intracellular killing and contributes to the chronic persistence of B. neotomae/B. melitensis in the host. This study highlights the essential role of arginine in clearing intracellular infections and the subversion of this host defense mechanism by intracellular pathogens for their chronic persistence.
Collapse
Affiliation(s)
- Sushree Rekha Mallik
- Laboratory of Immunology and Microbial Pathogenesis, BRIC-National Institute of Animal Biotechnology (BRIC-NIAB), Hyderabad, Telangana, India
- BRIC-Regional Centre for Biotechnology (BRIC-RCB), Faridabad, Haryana, India
| | - Kiranmai Joshi
- Laboratory of Immunology and Microbial Pathogenesis, BRIC-National Institute of Animal Biotechnology (BRIC-NIAB), Hyderabad, Telangana, India
- BRIC-Regional Centre for Biotechnology (BRIC-RCB), Faridabad, Haryana, India
| | - Girish K. Radhakrishnan
- Laboratory of Immunology and Microbial Pathogenesis, BRIC-National Institute of Animal Biotechnology (BRIC-NIAB), Hyderabad, Telangana, India
| |
Collapse
|
13
|
Wang J, Yan Z, Zhang W, Liu X, Wang J, Peng Q. Upregulation of TREM2 expression in M2 macrophages promotes Brucella abortus chronic infection. Front Immunol 2024; 15:1466520. [PMID: 39497817 PMCID: PMC11532147 DOI: 10.3389/fimmu.2024.1466520] [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: 07/18/2024] [Accepted: 09/27/2024] [Indexed: 11/07/2024] Open
Abstract
Brucella abortus (B.abortus) is a zoonotic bacterial pathogen that causes chronic host infections. The eradication of brucellosis using antibiotic therapy is often incomplete or slow. In a mouse model, the predominance of alternatively activated macrophages (also known as M2) plays an essential role in sustaining chronic infection. The underlying functional mechanism by which M2 sustains chronic infection remains unclear. Here, we show that B. abortus can enter M2 via triggering receptor expressed on myeloid cells 2 (TREM2) and promotes the upregulation of TREM2 expression of M2 in a type IV secretion system (T4SS)-dependent manner. Increased TREM2 enhances B. abortus growth within M2 by suppressing intracellular ROS production, preventing M2 pyroptosis via suppression of mitochondrial ROS (mROS), and promoting M2 proliferation by increasing β-catenin expression. In line with these results, downregulation of TREM2 expression suppressed B. abortus intracellular growth and M2 proliferation and induced M2 pyroptosis. In our mouse model, upregulation of TREM2 expression sustained the accumulation of M2 and B. abortus chronic infection, whereas downregulation of TREM2 expression restricted M2 proliferation and chronic infection. Collectively, our results suggest that targeting TREM2 may be a potential adjunct to antibiotic therapy for the prevention of chronic Brucella infection.
Collapse
Affiliation(s)
- Jingyu Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhirong Yan
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Weiyu Zhang
- Institute of Microbiology Department, Jilin Provincial Center for Disease Control and Prevention, Changchun, China
| | - Xiaofeng Liu
- Tumor Hospital of Jilin Province, Changchun, China
| | - Jun Wang
- Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Qisheng Peng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
14
|
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
|
15
|
Loperena-Barber M, Elizalde-Bielsa A, Salvador-Bescós M, Ruiz-Rodríguez P, Pellegrini JM, Renau-Mínguez C, Lancaster R, Zúñiga-Ripa A, Iriarte M, Bengoechea JA, Coscollá M, Gorvel JP, Moriyón I, Conde-Álvarez R. "Phylogenomic insights into brucellaceae: The Pseudochrobactrum algeriensis case". INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 123:105625. [PMID: 38906517 DOI: 10.1016/j.meegid.2024.105625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
The genus Pseudochrobactrum encompasses free-living bacteria phylogenetically close to Ochrobactrum opportunistic pathogens and to Brucella, facultative intracellular parasites causing brucellosis, a worldwide-extended and grave zoonosis. Recently, Pseudochrobactrum strains were isolated from Brucella natural hosts on Brucella selective media, potentially causing diagnostic confusions. Strikingly, P. algeriensis was isolated from cattle lymph nodes, organs that are inimical to bacteria. Here, we analyse P. algeriensis potential virulence factors in comparison with Ochrobactrum and Brucella. Consistent with genomic analyses, Western-Blot analyses confirmed that P. algeriensis lacks the ability to synthesize the N-formylperosamine O-polysaccharide characteristic of the lipopolysaccharide (LPS) of smooth Brucella core species. However, unlike other Pseudochrobactrum but similar to some early diverging brucellae, P. algeriensis carries genes potentially synthetizing a rhamnose-based O-polysaccharide LPS. Lipid A analysis by MALDI-TOF demonstrated that P. algeriensis LPS bears a lipid A with a reduced pathogen-associated molecular pattern, a trait shared with Ochrobactrum and Brucella that is essential to generate a highly stable outer membrane and to delay immune activation. Also, although not able to multiply intracellularly in macrophages, the analysis of P. algeriensis cell lipid envelope revealed the presence of large amounts of cationic aminolipids, which may account for the extremely high resistance of P. algeriensis to bactericidal peptides and could favor colonization of mucosae and transient survival in Brucella hosts. However, two traits critical in Brucella pathogenicity are either significantly different (T4SS [VirB]) or absent (erythritol catabolic pathway) in P. algeriensis. This work shows that, while diverging in other characteristics, lipidic envelope features relevant in Brucella pathogenicity are conserved in Brucellaceae. The constant presence of these features strongly suggests that reinforcement of the envelope integrity as an adaptive advantage in soil was maintained in Brucella because of the similarity of some environmental challenges, such as the action of cationic peptide antibiotics and host defense peptides. This information adds knowledge about the evolution of Brucellaceae, and also underlines the taxonomical differences of the three genera compared.
Collapse
Affiliation(s)
- Maite Loperena-Barber
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Aitor Elizalde-Bielsa
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Miriam Salvador-Bescós
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Paula Ruiz-Rodríguez
- Institute for Integrative Systems Biology, Universidad de Valencia-CSIC, Valencia, Spain
| | | | - Chantal Renau-Mínguez
- Institute for Integrative Systems Biology, Universidad de Valencia-CSIC, Valencia, Spain
| | - Rebecca Lancaster
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Amaia Zúñiga-Ripa
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Maite Iriarte
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Jose A Bengoechea
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Mireia Coscollá
- Institute for Integrative Systems Biology, Universidad de Valencia-CSIC, Valencia, Spain
| | - Jean-Pierre Gorvel
- Centre d'Immunologie de Marseille-Luminy, CNRS, INSERM, Aix-Marseille University, Marseille, France
| | - Ignacio Moriyón
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Raquel Conde-Álvarez
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
16
|
Pannetta M, Smal M, Ferravante C, Eletto D, Di Rosa D, Alexandrova E, Rizzo F, Voli A, Tosco A, Weisz A, Porta A. Transcriptome analysis of macrophages during Brucella abortus infection clarifies the survival mechanisms of the bacteria. Diagn Microbiol Infect Dis 2024; 110:116401. [PMID: 38878343 DOI: 10.1016/j.diagmicrobio.2024.116401] [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: 04/08/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/30/2024]
Abstract
Brucellosis is a critical zoonotic disease impacting humans and animals globally, causing symptoms like fever and arthritis in humans and reproductive issues in animals. The disease stems from the Brucella genus, adept at evading the immune system and proliferating within host cells. This study explores how Brucella abortus manipulates host cellular mechanisms to sustain infection, focusing on the interaction with murine macrophages over 24 h. Initial host defenses involve innate immune responses, while Brucella's survival strategies include evading lysosomal degradation and modulating host cell functions through various pathways. The research identified significant transcriptional changes in macrophages post-infection, highlighting pathways such as cytokine storm, pyroptosis signaling, Toll-like receptor pathways, and LXRs/RXRs signaling. The findings shed light on Brucella's complex mechanisms to undermine host defenses and underscore the need for further investigation into therapeutic targets to combat brucellosis.
Collapse
Affiliation(s)
- Martina Pannetta
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy; Ph.D. Program in Drug Discovery and Development, University of Salerno, Fisciano, SA, Italy
| | - Marharyta Smal
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana" University of Salerno, Baronissi, SA, Italy
| | - Carlo Ferravante
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana" University of Salerno, Baronissi, SA, Italy; Medical Genomics Program and Division of Oncology, AOU "S. Giovanni di Dio e Ruggi d'Aragona", University of Salerno, Salerno, Italy
| | - Daniela Eletto
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | - Domenico Di Rosa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana" University of Salerno, Baronissi, SA, Italy
| | - Elena Alexandrova
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana" University of Salerno, Baronissi, SA, Italy
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana" University of Salerno, Baronissi, SA, Italy; Medical Genomics Program and Division of Oncology, AOU "S. Giovanni di Dio e Ruggi d'Aragona", University of Salerno, Salerno, Italy; Genome Research Center for Health-CRGS, Campus of Medicine of the University of Salerno, Baronissi, SA, Italy
| | - Antonia Voli
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy; Ph.D. Program in Drug Discovery and Development, University of Salerno, Fisciano, SA, Italy
| | - Alessandra Tosco
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana" University of Salerno, Baronissi, SA, Italy; Medical Genomics Program and Division of Oncology, AOU "S. Giovanni di Dio e Ruggi d'Aragona", University of Salerno, Salerno, Italy; Genome Research Center for Health-CRGS, Campus of Medicine of the University of Salerno, Baronissi, SA, Italy
| | - Amalia Porta
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy.
| |
Collapse
|
17
|
Sun P, Yang X, Liu J, Bao Y, Qi J, Han X, Liu G, Wang S, Tian M. Development and Application of Colloidal Gold Test Strips for the Rapid Detection of Canine Brucellosis. BIOSENSORS 2024; 14:388. [PMID: 39194617 DOI: 10.3390/bios14080388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024]
Abstract
Brucellosis is a global problem, with the causative agent being the genus Brucella. B. canis can cause undulant fever in dogs, which is a zoonotic disease that can spread not only among dogs but also to humans. This poses a public health threat to society. In this study, a rapid and straightforward immune colloidal gold test strip was developed for the diagnosis of canine brucellosis through the detection of anti-LPS antibodies in serum samples. Rabbit anti-canine IgG conjugated with colloidal gold was employed as the colloidal gold-labeled antibody. The extracted high-purity R-LPS was employed as the capture antigen in the test line (T-line), while goat anti-rabbit IgG was utilized as the capture antibody in the control line (C-line). The colloidal gold strip exhibited high specificity in the detection of brucellosis, with no cross-reaction observed with the common clinical canine diseases caused by Canine coronavirus (CCV), Canine distemper virus (CDV), and Canine parvovirus (CPV). In comparison to the commercial iELISA kit, the sensitivity and specificity of the colloidal gold test strip were found to be 95.23% and 98.76%, respectively. The diagnostic coincidence rate was 98.47%. The findings of this study indicate that colloidal gold test strips may be employed as a straightforward, expeditious, sensitive, and specific diagnostic instrument for the identification of canine brucellosis, particularly in resource-limited regions.
Collapse
Affiliation(s)
- Pengxiang Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - Xinmei Yang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jinyue Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yanqing Bao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jingjing Qi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Guanhui Liu
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - Shaohui Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Mingxing Tian
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| |
Collapse
|
18
|
Giménez A, Del Giudice MG, López PV, Guaimas F, Sámano-Sánchez H, Gibson TJ, Chemes LB, Arregui CO, Ugalde JE, Czibener C. Brucella NpeA is a secreted Type IV effector containing an N-WASP-binding short linear motif that promotes niche formation. mBio 2024; 15:e0072624. [PMID: 38847540 PMCID: PMC11253601 DOI: 10.1128/mbio.00726-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/01/2024] [Indexed: 07/18/2024] Open
Abstract
The modulation of actin polymerization is a common theme among microbial pathogens. Even though microorganisms show a wide repertoire of strategies to subvert the activity of actin, most of them converge in the ones that activate nucleating factors, such as the Arp2/3 complex. Brucella spp. are intracellular pathogens capable of establishing chronic infections in their hosts. The ability to subvert the host cell response is dependent on the capacity of the bacterium to attach, invade, avoid degradation in the phagocytic compartment, replicate in an endoplasmic reticulum-derived compartment and egress. Even though a significant number of mechanisms deployed by Brucella in these different phases have been identified and characterized, none of them have been described to target actin as a cellular component. In this manuscript, we describe the identification of a novel virulence factor (NpeA) that promotes niche formation. NpeA harbors a short linear motif (SLiM) present within an amphipathic alpha helix that has been described to bind the GTPase-binding domain (GBD) of N-WASP and stabilizes the autoinhibited state. Our results show that NpeA is secreted in a Type IV secretion system-dependent manner and that deletion of the gene diminishes the intracellular replication capacity of the bacterium. In vitro and ex vivo experiments demonstrate that NpeA binds N-WASP and that the short linear motif is required for the biological activity of the protein.IMPORTANCEThe modulation of actin-binding effectors that regulate the activity of this fundamental cellular protein is a common theme among bacterial pathogens. The neural Wiskott-Aldrich syndrome protein (N-WASP) is a protein that several pathogens target to hijack actin dynamics. The highly adapted intracellular bacterium Brucella has evolved a wide repertoire of virulence factors that modulate many activities of the host cell to establish successful intracellular replication niches, but, to date, no effector proteins have been implicated in the modulation of actin dynamics. We present here the identification of a virulence factor that harbors a short linear motif (SLiM) present within an amphipathic alpha helix that has been described to bind the GTPase-binding domain (GBD) of N-WASP stabilizing its autoinhibited state. We demonstrate that this protein is a Type IV secretion effector that targets N-WASP-promoting intracellular survival and niche formation.
Collapse
Affiliation(s)
- Agostina Giménez
- 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
| | - Mariela G. Del Giudice
- 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
| | - Paula V. López
- 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
| | - Francisco Guaimas
- 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
| | - Hugo Sámano-Sánchez
- Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, China
- Biomedical Sciences, Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Toby J. Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Lucía B. Chemes
- 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
| | - Carlos O. Arregui
- 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 E. Ugalde
- 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
| | - Cecilia Czibener
- 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
| |
Collapse
|
19
|
Zhang G, Hu H, Yin Y, Tian M, Bu Z, Ding C, Yu S. Brucella Manipulates Host Cell Ferroptosis to Facilitate Its Intracellular Replication and Egress in RAW264.7 Macrophages. Antioxidants (Basel) 2024; 13:577. [PMID: 38790682 PMCID: PMC11118192 DOI: 10.3390/antiox13050577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Brucella virulence relies on its successful intracellular life cycle. Modulating host cell death is a strategy for Brucella to survive and replicate intracellularly. Ferroptosis is a novel regulated cell death characterized by iron-triggered excessive lipid peroxidation, which has been proven to be associated with pathogenic bacteria infection. Thus, we attempted to explore if smooth-type Brucella infection triggers host cell ferroptosis and what role it plays in Brucella infection. We assessed the effects of Brucella infection on the lactate dehydrogenase release and lipid peroxidation levels of RAW264.7 macrophages; subsequently, we determined the effect of Brucella infection on the expressions of ferroptosis defense pathways. Furthermore, we determined the role of host cell ferroptosis in the intracellular replication and egress of Brucella. The results demonstrated that Brucella M5 could induce ferroptosis of macrophages by inhibiting the GPX4-GSH axis at the late stage of infection but mitigated ferroptosis by up-regulating the GCH1-BH4 axis at the early infection stage. Moreover, elevating host cell ferroptosis decreased Brucella intracellular survival and suppressing host cell ferroptosis increased Brucella intracellular replication and egress. Collectively, Brucella may manipulate host cell ferroptosis to facilitate its intracellular replication and egress, extending our knowledge about the underlying mechanism of how Brucella completes its intracellular life cycle.
Collapse
Affiliation(s)
- Guangdong Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China; (G.Z.); (H.H.); (Y.Y.); (M.T.)
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Harbin 150069, China;
| | - Hai Hu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China; (G.Z.); (H.H.); (Y.Y.); (M.T.)
| | - Yi Yin
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China; (G.Z.); (H.H.); (Y.Y.); (M.T.)
| | - Mingxing Tian
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China; (G.Z.); (H.H.); (Y.Y.); (M.T.)
| | - Zhigao Bu
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Harbin 150069, China;
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China; (G.Z.); (H.H.); (Y.Y.); (M.T.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Shengqing Yu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China; (G.Z.); (H.H.); (Y.Y.); (M.T.)
| |
Collapse
|
20
|
Chatterjee R, Setty SRG, Chakravortty D. SNAREs: a double-edged sword for intravacuolar bacterial pathogens within host cells. Trends Microbiol 2024; 32:477-493. [PMID: 38040624 DOI: 10.1016/j.tim.2023.11.002] [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: 08/05/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 12/03/2023]
Abstract
In the tug-of-war between host and pathogen, both evolve to combat each other's defence arsenals. Intracellular phagosomal bacteria have developed strategies to modify the vacuolar niche to suit their requirements best. Conversely, the host tries to target the pathogen-containing vacuoles towards the degradative pathways. The host cells use a robust system through intracellular trafficking to maintain homeostasis inside the cellular milieu. In parallel, intracellular bacterial pathogens have coevolved with the host to harbour strategies to manipulate cellular pathways, organelles, and cargoes, facilitating the conversion of the phagosome into a modified pathogen-containing vacuole (PCV). Key molecular regulators of intracellular traffic, such as changes in the organelle (phospholipid) composition, recruitment of small GTPases and associated effectors, soluble N-ethylmaleimide-sensitive factor-activating protein receptors (SNAREs), etc., are hijacked to evade lysosomal degradation. Legionella, Salmonella, Coxiella, Chlamydia, Mycobacterium, and Brucella are examples of pathogens which diverge from the endocytic pathway by using effector-mediated mechanisms to overcome the challenges and establish their intracellular niches. These pathogens extensively utilise and modulate the end processes of secretory pathways, particularly SNAREs, in repurposing the PCV into specialised compartments resembling the host organelles within the secretory network; at the same time, they avoid being degraded by the host's cellular mechanisms. Here, we discuss the recent research advances on the host-pathogen interaction/crosstalk that involves host SNAREs, conserved cellular processes, and the ongoing host-pathogen defence mechanisms in the molecular arms race against each other. The current knowledge of SNAREs, and intravacuolar bacterial pathogen interactions, enables us to understand host cellular innate immune pathways, maintenance of homeostasis, and potential therapeutic strategies to combat ever-growing antimicrobial resistance.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka, India
| | - Subba Rao Gangi Setty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka, India.
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka, India; Adjunct Faculty, Indian Institute of Science Research and Education, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
21
|
Khairullah AR, Kurniawan SC, Puspitasari Y, Aryaloka S, Silaen OSM, Yanestria SM, Widodo A, Moses IB, Effendi MH, Afnani DA, Ramandinianto SC, Hasib A, Riwu KHP. Brucellosis: Unveiling the complexities of a pervasive zoonotic disease and its global impacts. Open Vet J 2024; 14:1081-1097. [PMID: 38938422 PMCID: PMC11199761 DOI: 10.5455/ovj.2024.v14.i5.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/08/2024] [Indexed: 06/29/2024] Open
Abstract
One zoonotic infectious animal disease is brucellosis. The bacteria that cause brucellosis belong to the genus Brucella. Numerous animal and human species are affected by brucellosis, with an estimated 500,000 human cases recorded annually worldwide. The occurrence of new areas of infection and the resurgence of infection in already infected areas indicate how dynamically brucellosis is distributed throughout different geographic regions. Bacteria originate from the blood and are found in the reticuloendothelial system, the liver, the spleen, and numerous other locations, including the joints, kidneys, heart, and genital tract. Diagnosis of this disease can be done by bacterial isolation, molecular tests, modified acid-fast stain, rose bengal test (RBT), milk ring test, complement fixation test, enzyme-linked immunosorbent assay, and serum agglutination test. The primary sign of a Brucella abortus infection is infertility, which can result in abortion and the birth of a frail fetus that may go on to infect other animals. In humans, the main symptoms are acute febrile illness, with or without localization signs, and chronic infection. Female cattle have a greater risk of contracting Brucella disease. Human populations at high risk of contracting brucellosis include those who care for cattle, veterinarians, slaughterhouse employees, and butchers. Antibiotic treatment of brucellosis is often unsuccessful due to the intracellular survival of Brucella and its adaptability in macrophages. A "one health" strategy is necessary to control illnesses like brucellosis.
Collapse
Affiliation(s)
- Aswin Rafif Khairullah
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Shendy Canadya Kurniawan
- Master Program of Animal Sciences, Department of Animal Sciences, Specialisation in Molecule, Cell and Organ Functioning, Wageningen University and Research, Wageningen, The Netherlands
| | - Yulianna Puspitasari
- Division of Veterinary Microbiology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Suhita Aryaloka
- Master Program of Veterinary Agribusiness, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Otto Sahat Martua Silaen
- Doctoral Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | - Agus Widodo
- Department of Health, Faculty of Vocational Studies, Universitas Airlangga, Surabaya, Indonesia
| | - Ikechukwu Benjamin Moses
- Department of Applied Microbiology, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | - Mustofa Helmi Effendi
- Division of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Daniah Ashri Afnani
- Department of Microbiology and Parasitology, Faculty of Veterinary Medicine, Universitas Pendidikan Mandalika, Mataram, Indonesia
| | | | - Abdullah Hasib
- School of Agriculture and Food Sustainability, The University of Queensland, Gatton, Queensland
| | - Katty Hendriana Priscilia Riwu
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Pendidikan Mandalika, Mataram, Indonesia
| |
Collapse
|
22
|
Elizalde-Bielsa A, Muñoz PM, Zúñiga-Ripa A, Conde-Álvarez R. A Review on the Methodology and Use of the Pregnant Mouse Model in the Study of Brucella Reproductive Pathogenesis and Its Abortifacient Effect. Microorganisms 2024; 12:866. [PMID: 38792696 PMCID: PMC11123383 DOI: 10.3390/microorganisms12050866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
Brucellosis is one of the most common and widespread bacterial zoonoses and is caused by Gram-negative bacteria belonging to the genus Brucella. These organisms are able to infect and replicate within the placenta, resulting in abortion, one of the main clinical signs of brucellosis. Although the mouse model is widely used to study Brucella virulence and, more recently, to evaluate the protection of new vaccines, there is no clear consensus on the experimental conditions (e.g., mouse strains, doses, routes of inoculation, infection/pregnancy time) and the natural host reproducibility of the pregnant mouse model for reproductive brucellosis. This lack of consensus calls for a review that integrates the major findings regarding the effect of Brucella wild-type and vaccine strains infections on mouse pregnancy. We found sufficient evidence on the utility of the pregnant mouse model to study Brucella-induced placentitis and abortion and propose suitable experimental conditions (dose, time of infection) and pregnancy outcome readouts for B. abortus and B. melitensis studies. Finally, we discuss the utility and limitations of the pregnant mouse as a predictive model for the abortifacient effect of live Brucella vaccines.
Collapse
Affiliation(s)
- Aitor Elizalde-Bielsa
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain;
| | - Pilar M. Muñoz
- Department of Animal Science, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), 50059 Zaragoza, Spain;
- Instituto Agroalimentario de Aragón—IA2, CITA-Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Amaia Zúñiga-Ripa
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain;
| | - Raquel Conde-Álvarez
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain;
| |
Collapse
|
23
|
Xu H, Lu J, Huang F, Zhang Q, Liu S, Chen Z, Li S. A genome-wide CRISPR screen identified host genes essential for intracellular Brucella survival. Microbiol Spectr 2024; 12:e0338323. [PMID: 38376367 PMCID: PMC10986529 DOI: 10.1128/spectrum.03383-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 01/26/2024] [Indexed: 02/21/2024] Open
Abstract
Brucella is a zoonotic intracellular bacterium that poses threats to human health and economic security. Intracellular infection is a hallmark of the agent Brucella and a primary cause of distress, through which the bacterium regulates the host intracellular environment to promote its own colonization and replication, evading host immunity and pharmaceutical killing. Current studies of Brucella intracellular processes are typically premised on bacterial phenotype such as intracellular bacterial survival, followed by biochemical or molecular biological approaches to reveal detailed mechanisms. While such processes can deepen the understanding of Brucella-host interaction, the insights into host alterations in infection would be easily restricted to known pathways. In the current study, we applied CRISPR Cas9 screen to identify host genes that are most affected by Brucella infection on cell viability at the genomic level. As a result of CRISPR screening, we firstly identified that knockout of the negatively selected genes GOLGA6L6, DEFB103B, OR4F29, and ERCC6 attenuate the viability of both the host cells and intracellular Brucella, suggesting these genes to be potential therapeutic targets for Brucella control. In particular, knockout of DEFB103B diminished Brucella intracellular survival by altering host cell autophagy. Conversely, knockout of positive screening genes promoted intracellular proliferation of Brucella. In summary, we screened host genes at the genomic level throughout Brucella infection, identified host genes that are previously not recognized to be involved in Brucella infection, and provided targets for intracellular infection control.IMPORTANCEBrucella is a Gram-negative bacterium that infects common mammals causing arthritis, myalgia, neuritis, orchitis, or miscarriage and is difficult to cure with antibiotics due to its intracellular parasitism. Therefore, unraveling the mechanism of Brucella-host interactions will help controlling Brucella infections. CRISPR-Cas9 is a gene editing technology that directs knockout of individual target genes by guided RNA, from which genome-wide gene-knockout cell libraries can be constructed. Upon infection with Brucella, the cell library would show differences in viability as a result of the knockout and specific genes could be revealed by genomic DNA sequencing. As a result, genes affecting cell viability during Brucella infection were identified. Further testing of gene function may reveal the mechanisms of Brucella-host interactions, thereby contributing to clinical therapy.
Collapse
Affiliation(s)
- Heling Xu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, China
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Jingjing Lu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Qi Zhang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, China
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Shuang Liu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, China
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Zeliang Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
24
|
Lázaro-Antón L, Veiga-da-Cunha M, Elizalde-Bielsa A, Chevalier N, Conde-Álvarez R, Iriarte M, Letesson JJ, Moriyón I, Van Schaftingen E, Zúñiga-Ripa A. A novel gluconeogenic route enables efficient use of erythritol in zoonotic Brucella. Front Vet Sci 2024; 11:1328293. [PMID: 38601913 PMCID: PMC11005471 DOI: 10.3389/fvets.2024.1328293] [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: 10/26/2023] [Accepted: 01/29/2024] [Indexed: 04/12/2024] Open
Abstract
Brucellosis is a worldwide extended zoonosis caused by pathogens of the genus Brucella. While most B. abortus, B. melitensis, and B. suis biovars grow slowly in complex media, they multiply intensely in livestock genitals and placenta indicating high metabolic capacities. Mutant analyses in vitro and in infection models emphasize that erythritol (abundant in placenta and genitals) is a preferred substrate of brucellae, and suggest hexoses, pentoses, and gluconeogenic substrates use in host cells. While Brucella sugar and erythritol catabolic pathways are known, growth on 3-4 carbon substrates persists in Fbp- and GlpX-deleted mutants, the canonical gluconeogenic fructose 1,6-bisphosphate (F1,6bP) bisphosphatases. Exploiting the prototrophic and fast-growing properties of B. suis biovar 5, we show that gluconeogenesis requires fructose-bisphosphate aldolase (Fba); the existence of a novel broad substrate bisphosphatase (Bbp) active on sedoheptulose 1,7-bisphosphate (S1,7bP), F1,6bP, and other phosphorylated substrates; that Brucella Fbp unexpectedly acts on S1,7bP and F1,6bP; and that, while active in B. abortus and B. melitensis, GlpX is disabled in B. suis biovar 5. Thus, two Fba-dependent reactions (dihydroxyacetone-phosphate + glyceraldehyde 3-phosphate ⇌ F1,6bP; and dihydroxyacetone-phosphate + erythrose 4-phosphate ⇌ S1,7bP) can, respectively, yield fructose 6-phosphate and sedoheptulose 7-phosphate for classical gluconeogenesis and the Pentose Phosphate Shunt (PPS), the latter reaction opening a new gluconeogenic route. Since erythritol generates the PPS-intermediate erythrose 4-phosphate, and the Fba/Fbp-Bbp route predicts sedoheptulose 7-phosphate generation from erythrose 4-phosphate, we re-examined the erythritol connections with PPS. Growth on erythritol required transaldolase or the Fba/Fbp-Bbp pathway, strongly suggesting that Fba/Fbp-Bbp works as a PPS entry for both erythritol and gluconeogenic substrates in Brucella. We propose that, by increasing erythritol channeling into PPS through these peculiar routes, brucellae proliferate in livestock genitals and placenta in the high numbers that cause abortion and infertility, and make brucellosis highly contagious. These findings could be the basis for developing attenuated brucellosis vaccines safer in pregnant animals.
Collapse
Affiliation(s)
- Leticia Lázaro-Antón
- Departamento de Microbiología y Parasitología – IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Maria Veiga-da-Cunha
- Groupe de Recherches Metaboliques, De Duve Institute, UCLouvain, Brussels, Belgium
| | - Aitor Elizalde-Bielsa
- Departamento de Microbiología y Parasitología – IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Nathalie Chevalier
- Groupe de Recherches Metaboliques, De Duve Institute, UCLouvain, Brussels, Belgium
| | - Raquel Conde-Álvarez
- Departamento de Microbiología y Parasitología – IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Maite Iriarte
- Departamento de Microbiología y Parasitología – IDISNA, Universidad de Navarra, Pamplona, Spain
| | | | - Ignacio Moriyón
- Departamento de Microbiología y Parasitología – IDISNA, Universidad de Navarra, Pamplona, Spain
| | | | - Amaia Zúñiga-Ripa
- Departamento de Microbiología y Parasitología – IDISNA, Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
25
|
Carvalho TP, Toledo FAO, Bautista DFA, Silva MF, Oliveira JBS, Lima PA, Costa FB, Ribeiro NQ, Lee JY, Birbrair A, Paixão TA, Tsolis RM, Santos RL. Pericytes modulate endothelial inflammatory response during bacterial infection. mBio 2024; 15:e0325223. [PMID: 38289074 PMCID: PMC10936204 DOI: 10.1128/mbio.03252-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 03/14/2024] Open
Abstract
Pericytes are located around blood vessels, in close contact with endothelial cells. We discovered that pericytes dampen pro-inflammatory endothelial cell responses. Endothelial cells co-cultured with pericytes had markedly reduced expression of adhesion molecules (PECAM-1 and ICAM-1) and proinflammatory cytokines (CCL-2 and IL-6) in response to bacterial stimuli (Brucella ovis, Listeria monocytogenes, or Escherichia coli lipopolysaccharide). Pericyte-depleted mice intraperitoneally inoculated with either B. ovis, a stealthy pathogen that does not trigger detectable inflammation, or Listeria monocytogenes, developed peritonitis. Further, during Citrobacter rodentium infection, pericyte-depleted mice developed severe intestinal inflammation, which was not evident in control mice. The anti-inflammatory effect of pericytes required connexin 43, as either chemical inhibition or silencing of connexin 43 abrogated pericyte-mediated suppression of endothelial inflammatory responses. Our results define a mechanism by which pericytes modulate inflammation during infection, which shifts our understanding of pericyte biology: from a structural cell to a pro-active player in modulating inflammation. IMPORTANCE A previously unknown mechanism by which pericytes modulate inflammation was discovered. The absence of pericytes or blocking interaction between pericytes and endothelium through connexin 43 results in stronger inflammation, which shifts our understanding of pericyte biology, from a structural cell to a player in controlling inflammation.
Collapse
Affiliation(s)
- Thaynara P. Carvalho
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Frank A. O. Toledo
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Diego F. A. Bautista
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Monique F. Silva
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jefferson B. S. Oliveira
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pâmela A. Lima
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fabíola B. Costa
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Noelly Q. Ribeiro
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jee-Yon Lee
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Alexander Birbrair
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tatiane A. Paixão
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Reneé M. Tsolis
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Renato L. Santos
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| |
Collapse
|
26
|
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
|
27
|
Ward JA, Ng'ang'a SI, Randhawa IAS, McHugo GP, O'Grady JF, Flórez JM, Browne JA, Pérez O’Brien AM, Landaeta-Hernández AJ, Garcia JF, Sonstegard TS, Frantz LAF, Salter-Townshend M, MacHugh DE. Genomic insights into the population history and adaptive traits of Latin American Criollo cattle. ROYAL SOCIETY OPEN SCIENCE 2024; 11:231388. [PMID: 38571912 PMCID: PMC10990470 DOI: 10.1098/rsos.231388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/04/2024] [Accepted: 01/31/2024] [Indexed: 04/05/2024]
Abstract
Criollo cattle, the descendants of animals brought by Iberian colonists to the Americas, have been the subject of natural and human-mediated selection in novel tropical agroecological zones for centuries. Consequently, these breeds have evolved distinct characteristics such as resistance to diseases and exceptional heat tolerance. In addition to European taurine (Bos taurus) ancestry, it has been proposed that gene flow from African taurine and Asian indicine (Bos indicus) cattle has shaped the ancestry of Criollo cattle. In this study, we analysed Criollo breeds from Colombia and Venezuela using whole-genome sequencing (WGS) and single-nucleotide polymorphism (SNP) array data to examine population structure and admixture at high resolution. Analysis of genetic structure and ancestry components provided evidence for African taurine and Asian indicine admixture in Criollo cattle. In addition, using WGS data, we detected selection signatures associated with a myriad of adaptive traits, revealing genes linked to thermotolerance, reproduction, fertility, immunity and distinct coat and skin coloration traits. This study underscores the remarkable adaptability of Criollo cattle and highlights the genetic richness and potential of these breeds in the face of climate change, habitat flux and disease challenges. Further research is warranted to leverage these findings for more effective and sustainable cattle breeding programmes.
Collapse
Affiliation(s)
- James A. Ward
- Animal Genomics Laboratory, School of Agriculture and Food Science, University College Dublin, DublinD04 V1W8, Ireland
| | - Said I. Ng'ang'a
- Palaeogenomics Group, Department of Veterinary Sciences, Ludwig Maximilian University, MunichD-80539, Germany
- School of Biological and Chemical Sciences, Queen Mary University of London, LondonE1 4NS, UK
| | | | - Gillian P. McHugo
- Animal Genomics Laboratory, School of Agriculture and Food Science, University College Dublin, DublinD04 V1W8, Ireland
| | - John F. O'Grady
- Animal Genomics Laboratory, School of Agriculture and Food Science, University College Dublin, DublinD04 V1W8, Ireland
| | - Julio M. Flórez
- Acceligen, Eagan, MN55121, USA
- Department of Preventive Veterinary Medicine and Animal Reproduction, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | - John A. Browne
- Animal Genomics Laboratory, School of Agriculture and Food Science, University College Dublin, DublinD04 V1W8, Ireland
| | | | - Antonio J. Landaeta-Hernández
- Unidad de Investigaciones Zootécnicas, Facultad de Ciencias Veterinarias, Universidad del Zulia, Maracaibo, Venezuela
| | - Jóse F. Garcia
- Department of Preventive Veterinary Medicine and Animal Reproduction, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
| | | | - Laurent A. F. Frantz
- Palaeogenomics Group, Department of Veterinary Sciences, Ludwig Maximilian University, MunichD-80539, Germany
- School of Biological and Chemical Sciences, Queen Mary University of London, LondonE1 4NS, UK
| | | | - David E. MacHugh
- Animal Genomics Laboratory, School of Agriculture and Food Science, University College Dublin, DublinD04 V1W8, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, DublinD04 V1W8, Ireland
| |
Collapse
|
28
|
Flores-Concha M, Gómez LA, Soto-Shara R, Molina RE, Coloma-Rivero RF, Montero DA, Ferrari Í, Oñate Á. Brucella abortus triggers the differential expression of immunomodulatory lncRNAs in infected murine macrophages. Front Immunol 2024; 15:1352306. [PMID: 38464511 PMCID: PMC10921354 DOI: 10.3389/fimmu.2024.1352306] [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: 12/07/2023] [Accepted: 01/30/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction The lncRNAs (long non-coding RNAs) are the most diverse group of non-coding RNAs and are involved in most biological processes including the immune response. While some of them have been recognized for their influence on the regulation of inflammatory activity, little is known in the context of infection by Brucella abortus, a pathogen that presents significant challenges due to its ability to manipulate and evade the host immune system. This study focuses on characterize the expression profile of LincRNA-cox2, Lethe, lincRNA-EPS, Malat1 and Gas5 during infection of macrophages by B. abortus. Methods Using public raw RNA-seq datasets we constructed for a lncRNA expression profile in macrophages Brucella-infected. In addition, from public RNA-seq raw datasets of RAW264.7 cells infected with B. abortus we constructed a transcriptomic profile of lncRNAs in order to know the expression of the five immunomodulating lncRNAs studied here at 8 and 24 h post-infection. Finally, we performed in vitro infection assays in RAW264.7 cells and peritoneal macrophages to detect by qPCR changes in the expression of these lncRNAs at first 12 hours post infection, a key stage in the infection cycle where Brucella modulates the immune response to survive. Results Our results demonstrate that infection of macrophages with Brucella abortus, induces significant changes in the expression of LincRNA-Cox2, Lethe, LincRNA-EPS, Gas5, and Malat1. Discussion The change in the expression profile of these immunomodulatory lncRNAs in response to infection, suggest a potential involvement in the immune evasion strategy employed by Brucella to facilitate its intracellular survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ángel Oñate
- Laboratory of Molecular Immunology, Department of Microbiology, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
29
|
Joshi K, Mazumdar V, Nandi BR, Radhakrishnan GK. Brucella targets the host ubiquitin-specific protease, Usp8, through the effector protein, TcpB, for facilitating infection of macrophages. Infect Immun 2024; 92:e0028923. [PMID: 38174929 PMCID: PMC10863413 DOI: 10.1128/iai.00289-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/12/2023] [Indexed: 01/05/2024] Open
Abstract
Brucella species are Gram-negative intracellular bacterial pathogens that cause the worldwide zoonotic disease brucellosis. Brucella can infect many mammals, including humans and domestic and wild animals. Brucella manipulates various host cellular processes to invade and multiply in professional and non-professional phagocytic cells. However, the host targets and their modulation by Brucella to facilitate the infection process remain obscure. Here, we report that the host ubiquitin-specific protease, USP8, negatively regulates the invasion of Brucella into macrophages through the plasma membrane receptor, CXCR4. Upon silencing or chemical inhibition of USP8, the membrane localization of the CXCR4 receptor was enriched, which augmented the invasion of Brucella into macrophages. Activation of USP8 through chemical inhibition of 14-3-3 protein affected the invasion of Brucella into macrophages. Brucella suppressed the expression of Usp8 at its early stage of infection in the infected macrophages. Furthermore, we found that only live Brucella could negatively regulate the expression of Usp8, suggesting the role of secreted effector protein of Brucella in modulating the gene expression. Subsequent studies revealed that the Brucella effector protein, TIR-domain containing protein from Brucella, TcpB, plays a significant role in downregulating the expression of Usp8 by targeting the cyclic-AMP response element-binding protein pathway. Treatment of mice with USP8 inhibitor resulted in enhanced survival of B. melitensis, whereas mice treated with CXCR4 or 14-3-3 antagonists showed a diminished bacterial load. Our experimental data demonstrate a novel role of Usp8 in the host defense against microbial intrusion. The present study provides insights into the microbial subversion of host defenses, and this information may ultimately help to develop novel therapeutic interventions for infectious diseases.
Collapse
Affiliation(s)
- Kiranmai Joshi
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Varadendra Mazumdar
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, 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, India
| | - Girish K. Radhakrishnan
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| |
Collapse
|
30
|
King KA, Benton AH, Caudill MT, Stoyanof ST, Kang L, Michalak P, Lahmers KK, Dunman PM, DeHart TG, Ahmad SS, Jutras BL, Poncin K, De Bolle X, Caswell CC. Post-transcriptional control of the essential enzyme MurF by a small regulatory RNA in Brucella abortus. Mol Microbiol 2024; 121:129-141. [PMID: 38082493 DOI: 10.1111/mmi.15207] [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: 09/27/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 01/15/2024]
Abstract
Brucella abortus is a facultative, intracellular, zoonotic pathogen that resides inside macrophages during infection. This is a specialized niche where B. abortus encounters various stresses as it navigates through the macrophage. In order to survive this harsh environment, B. abortus utilizes post-transcriptional regulation of gene expression through the use of small regulatory RNAs (sRNAs). Here, we characterize a Brucella sRNAs called MavR (for MurF- and virulence-regulating sRNA), and we demonstrate that MavR is required for the full virulence of B. abortus in macrophages and in a mouse model of chronic infection. Transcriptomic and proteomic studies revealed that a major regulatory target of MavR is MurF. MurF is an essential protein that catalyzes the final cytoplasmic step in peptidoglycan (PG) synthesis; however, we did not detect any differences in the amount or chemical composition of PG in the ΔmavR mutant. A 6-nucleotide regulatory seed region within MavR was identified, and mutation of this seed region resulted in dysregulation of MurF production, as well as significant attenuation of infection in a mouse model. Overall, the present study underscores the importance of sRNA regulation in the physiology and virulence of Brucella.
Collapse
Affiliation(s)
- Kellie A King
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Angela H Benton
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Mitchell T Caudill
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - S Tristan Stoyanof
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Lin Kang
- Department of Biomedical Sciences, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, USA
- College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
- Center for One Health Research, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia, USA
| | - Pawel Michalak
- Department of Biomedical Sciences, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, USA
- Center for One Health Research, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia, USA
- Institute for Evolution, University of Haifa, Haifa, Israel
| | - Kevin K Lahmers
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Paul M Dunman
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Tanner G DeHart
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, USA
| | - Saadman S Ahmad
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, USA
| | - Brandon L Jutras
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, USA
| | - Katy Poncin
- URBM, Narilis, University of Namur, Namur, Belgium
| | | | - Clayton C Caswell
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
31
|
Djokic V, Freddi L, de Massis F, Lahti E, van den Esker MH, Whatmore A, Haughey A, Ferreira AC, Garofolo G, Melzer F, Sacchini F, Koets A, Wyllie S, Fontbonne A, Girault G, Vicente AF, McGiven J, Ponsart C. The emergence of Brucella canis as a public health threat in Europe: what we know and what we need to learn. Emerg Microbes Infect 2023; 12:2249126. [PMID: 37649455 PMCID: PMC10540651 DOI: 10.1080/22221751.2023.2249126] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023]
Abstract
The zoonotic bacteria, Brucella canis, is becoming the leading cause of canine brucellosis in Europe. In dogs, it causes reproductive problems as well as non-specific lameness or discospondilitis. In humans, B. canis can be origin of chronic debilitating conditions characteristic to its genus such as undulant fever, splenomegaly, and lymphadenopathy. Although B. canis shows some pathogenic characteristics similar to B. abortus and B. melitensis, it lacks surface O-polysaccharide, like nonzoonotic B. ovis. This review shows that host-B. canis interactions are still poorly understood, with many knowledge and capability gaps, causing relatively poor sensitivity and specificity of existing diagnostic tools. Currently, there is no vaccine for this rough Brucella species. Besides, antimicrobial therapy does not guarantee bacterial elimination, and infection relapses are frequently reported, increasing the risks of antibiotic resistance development. B. canis has been detected in dogs in almost all European countries which increased human exposure, but currently there is no systematic surveillance. Moreover, B. canis caused brucellosis is not included in Animal Health Law, and therefore there is no legal framework to tackle this emerging infectious disease. To map out the diagnostic strategies, identify risks for human infections and propose management scheme for infected pet and kennel dogs, we present current understanding of canine B. canis caused brucellosis, outline major knowledge gaps and propose future steps. To address and highlight challenges veterinary and public health services encounter in Europe, we developed two B. canis infection scenarios: of a single household pet and of a kennel dog in larger group.
Collapse
Affiliation(s)
- Vitomir Djokic
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, Animal Health Laboratory, Paris-Est University/ANSES, Maisons-Alfort, France
| | - Luca Freddi
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, Animal Health Laboratory, Paris-Est University/ANSES, Maisons-Alfort, France
| | - Fabrizio de Massis
- National and WOAH Reference Laboratory for Brucellosis, Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise “G. Caporale”, Teramo, Italy
| | - Elina Lahti
- Department of Epidemiology and Disease Control, National Veterinary Institute (SVA), Uppsala, Sweden
| | - Marielle H. van den Esker
- Department of Bacteriology, Host-Pathogen Interaction and Diagnostics, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Adrian Whatmore
- WOAH and FAO Brucellosis Reference Laboratory, Animal and Plant Health Agency (APHA), Addlestone, UK
| | - Anna Haughey
- WOAH and FAO Brucellosis Reference Laboratory, Animal and Plant Health Agency (APHA), Addlestone, UK
| | - Ana Cristina Ferreira
- National Institute for Agrarian and Veterinary Research, I.P. (INIAV, IP), Oeiras, Portugal
| | - Giuliano Garofolo
- National and WOAH Reference Laboratory for Brucellosis, Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise “G. Caporale”, Teramo, Italy
| | - Falk Melzer
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Jena, Germany
| | - Flavio Sacchini
- National and WOAH Reference Laboratory for Brucellosis, Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise “G. Caporale”, Teramo, Italy
| | - Ad Koets
- Department of Bacteriology, Host-Pathogen Interaction and Diagnostics, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Stephen Wyllie
- WOAH and FAO Brucellosis Reference Laboratory, Animal and Plant Health Agency (APHA), Addlestone, UK
| | - Alain Fontbonne
- Unité de Médecine de L'Elevage et du Sport (UMES), École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Guillaume Girault
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, Animal Health Laboratory, Paris-Est University/ANSES, Maisons-Alfort, France
| | - Acacia Ferreira Vicente
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, Animal Health Laboratory, Paris-Est University/ANSES, Maisons-Alfort, France
| | - John McGiven
- WOAH and FAO Brucellosis Reference Laboratory, Animal and Plant Health Agency (APHA), Addlestone, UK
| | - Claire Ponsart
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, Animal Health Laboratory, Paris-Est University/ANSES, Maisons-Alfort, France
| |
Collapse
|
32
|
Arce-Gorvel V, Hysenaj L, de Laval B, Sieweke MH, Sarrazin S, Gorvel JP. [The dance between Brucella and hematopoietic stem cells]. Med Sci (Paris) 2023; 39:822-824. [PMID: 38018922 DOI: 10.1051/medsci/2023152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023] Open
Affiliation(s)
- Vilma Arce-Gorvel
- Aix Marseille univ, CNRS, Inserm, Centre d'immunologie de Marseille-Luminy, Marseille, France
| | - Lisiena Hysenaj
- Aix Marseille univ, CNRS, Inserm, Centre d'immunologie de Marseille-Luminy, Marseille, France - Department of anatomy, University of California, San Francisco, États-Unis
| | - Bérengère de Laval
- Aix Marseille univ, CNRS, Inserm, Centre d'immunologie de Marseille-Luminy, Marseille, France
| | - Michael H Sieweke
- Aix Marseille univ, CNRS, Inserm, Centre d'immunologie de Marseille-Luminy, Marseille, France - Center for regenerative therapies Dresden, Technische Universität Dresden, Dresde, Allemagne
| | - Sandrine Sarrazin
- Aix Marseille univ, CNRS, Inserm, Centre d'immunologie de Marseille-Luminy, Marseille, France
| | - Jean-Pierre Gorvel
- Aix Marseille univ, CNRS, Inserm, Centre d'immunologie de Marseille-Luminy, Marseille, France
| |
Collapse
|
33
|
Dawood AS, Elrashedy A, Nayel M, Salama A, Guo A, Zhao G, Algharib SA, Zaghawa A, Zubair M, Elsify A, Mousa W, Luo W. Brucellae as resilient intracellular pathogens: epidemiology, host-pathogen interaction, recent genomics and proteomics approaches, and future perspectives. Front Vet Sci 2023; 10:1255239. [PMID: 37876633 PMCID: PMC10591102 DOI: 10.3389/fvets.2023.1255239] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 09/15/2023] [Indexed: 10/26/2023] Open
Abstract
Brucellosis is considered one of the most hazardous zoonotic diseases all over the world. It causes formidable economic losses in developed and developing countries. Despite the significant attempts to get rid of Brucella pathogens in many parts of the world, the disease continues to spread widely. Recently, many attempts proved to be effective for the prevention and control of highly contagious bovine brucellosis, which could be followed by others to achieve a prosperous future without rampant Brucella pathogens. In this study, the updated view for worldwide Brucella distribution, possible predisposing factors for emerging Brucella pathogens, immune response and different types of Brucella vaccines, genomics and proteomics approaches incorporated recently in the field of brucellosis, and future perspectives for prevention and control of bovine brucellosis have been discussed comprehensively. So, the current study will be used as a guide for researchers in planning their future work, which will pave the way for a new world without these highly contagious pathogens that have been infecting and threatening the health of humans and terrestrial animals.
Collapse
Affiliation(s)
- Ali Sobhy Dawood
- Engineering Laboratory for Tarim Animal Diseases Diagnosis and Control, College of Animal Science and Technology, Tarim University, Alar, Xinjiang, China
- The State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Alyaa Elrashedy
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Mohamed Nayel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Akram Salama
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Aizhen Guo
- The State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Gang Zhao
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, School of Life Sciences, Ningxia University, Yinchuan, China
| | - Samah Attia Algharib
- Engineering Laboratory for Tarim Animal Diseases Diagnosis and Control, College of Animal Science and Technology, Tarim University, Alar, Xinjiang, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues (HZAU), Wuhan, China
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Ahmed Zaghawa
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Muhammed Zubair
- Key Laboratory of Veterinary Biological Engineering and Technology, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Ahmed Elsify
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Walid Mousa
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Wanhe Luo
- Engineering Laboratory for Tarim Animal Diseases Diagnosis and Control, College of Animal Science and Technology, Tarim University, Alar, Xinjiang, China
| |
Collapse
|
34
|
Liu X, Wang P, Shi Y, Cui Y, Li S, Wu Dong G, Li J, Hao M, Zhai Y, Zhou D, Liu W, Wang A, Jin Y. (P)ppGpp synthetase Rsh participates in rifampicin tolerance of persister cells in Brucella abortus in vitro. Microb Pathog 2023; 183:106310. [PMID: 37604214 DOI: 10.1016/j.micpath.2023.106310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/30/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023]
Abstract
Brucella abortus is facultative intracellular pathogen that causes chronic persistent infections and results in abortion and infertility in food animals. Recurrent infections can be one of the results of persister cells formation that transiently displays phenotypic tolerance to high dose of antibiotics treatment. We examined persister cells formation of B. abortus strain A19 in stationary phase and investigated a potential role for the (p)ppGpp synthetase Rsh in this process. We found that B. abortus stationary phase cells can produce higher levels of multi-drugs tolerant persister cells in vitro under high dose of antibiotics (20 × MIC) exposure than do exponential phase cells. Persister cell formation was also induced with environmental stressors pH 4.5, 0.01 M PBS (pH7.0), 2% NaCl and 25 °C, upon exposure to ampicillin, enrofloxacin and rifampicin. Persister cells were not formed following exposure to 1 mM H2O2. The numbers of persister cells were significantly increased following uptake of B. abortus stationary phase cells by RAW264.7 macrophages in contrast with cultures in TSB liquid medium. Environmental stressors to B. abortus significantly increased expression of rsh mRNA level. The rsh null mutant (Δrsh) formed significantly fewer persister cells than the complemented (CΔrsh) and wildtype (WT) strains under high dose of rifampicin in vitro. These data for the first time demonstrate that B. abortus can produce multi-drug tolerant persister cells in stationary phase. The (p)ppGpp synthetase Rsh is necessary for persister cell formation in B. abortus in the presence of rifampicin. On this basis, a new understanding of the recurrent infections of Brucella was advanced, thus provided a new basis for revelation of pathogenic mechanism of the chronic persistent infection in Brucella.
Collapse
Affiliation(s)
- Xiaofang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Pingping Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Yong Shi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Yimeng Cui
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Shengnan Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Gaowa Wu Dong
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Junmei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Mingyue Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Yunyi Zhai
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Dong Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Wei Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| | - Aihua Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China.
| | - Yaping Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University; Yangling, Shaanxi 712100, China
| |
Collapse
|
35
|
Gómez LA, Molina RE, Soto RI, Flores MR, Coloma-Rivero RF, Montero DA, Oñate ÁA. Unraveling the Role of the Zinc-Dependent Metalloproteinase/HTH-Xre Toxin/Antitoxin (TA) System of Brucella abortus in the Oxidative Stress Response: Insights into the Stress Response and Virulence. Toxins (Basel) 2023; 15:536. [PMID: 37755962 PMCID: PMC10538038 DOI: 10.3390/toxins15090536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Toxin/antitoxin (TA) systems have been scarcely studied in Brucella abortus, the causative agent of brucellosis, which is one of the most prevalent zoonotic diseases worldwide. In this study, the roles of a putative type II TA system composed by a Zinc-dependent metalloproteinase (ZnMP) and a transcriptional regulator HTH-Xre were evaluated. The deletion of the open reading frame (ORF) BAB1_0270, coding for ZnMP, used to produce a mutant strain, allowed us to evaluate the survival and gene expression of B. abortus 2308 under oxidative conditions. Our results showed that the B. abortus mutant strain exhibited a significantly reduced capacity to survive under hydrogen peroxide-induced oxidative stress. Furthermore, this mutant strain showed a decreased expression of genes coding for catalase (katE), alkyl hydroperoxide reductase (ahpC) and transcriptional regulators (oxyR and oxyR-like), as well as genes involved in the general stress response, phyR and rpoE1, when compared to the wild-type strain. These findings suggest that this type II ZnMP/HTH-Xre TA system is required by B. abortus to resist oxidative stress. Additionally, previous evidence has demonstrated that this ZnMP also participates in the acidic stress resistance and virulence of B. abortus 2308. Therefore, we propose a hypothetical regulatory function for this ZnMP/HTH-Xre TA system, providing insight into the stress response and its potential roles in the pathogenesis of B. abortus.
Collapse
Affiliation(s)
- Leonardo A. Gómez
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción 4030000, Chile; (R.E.M.); (R.I.S.); (M.R.F.); (R.F.C.-R.); (D.A.M.)
| | | | | | | | | | | | - Ángel A. Oñate
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción 4030000, Chile; (R.E.M.); (R.I.S.); (M.R.F.); (R.F.C.-R.); (D.A.M.)
| |
Collapse
|
36
|
Freddi L, Vicente AF, Petit E, Ribeiro M, Game Y, Locatelli Y, Jacques I, Riou M, Jay M, Garin-Bastuji B, Rossi S, Djokic V, Ponsart C. Evaluation of a Lateral Flow Immunochromatography Assay (LFIA) for Diagnosis and Surveillance of Brucellosis in French Alpine Ibex ( Capra ibex). Microorganisms 2023; 11:1976. [PMID: 37630536 PMCID: PMC10459811 DOI: 10.3390/microorganisms11081976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
France has been officially free of bovine brucellosis since 2005. Nevertheless, in 2012, as the source of two human cases, a bovine outbreak due to B. melitensis biovar 3 was confirmed in the French Alpine Bargy massif, due to a spillover from wild, protected Alpine ibex (Capra ibex). In order to reduce high Brucella prevalence in the local ibex population, successive management strategies have been implemented. Lateral flow immunochromatography assay (LFIA) was thus identified as a promising on-site screening test, allowing for a rapid diagnosis far from the laboratory. This study compared a commercial LFIA for brucellosis diagnosis with the WOAH-recommended tests for small ruminants (i.e., Rose Bengal test (RBT), Complement fixation test, (CFT) and Indirect ELISA, (iELISA)). LFIA showed the same analytical sensitivity as iELISA on successive dilutions of the International Standard anti-Brucella melitensis Serum (ISaBmS) and the EU Goat Brucella Standard Serum (EUGBSS). Selectivity was estimated at 100% when vaccinated ibex sera were analyzed. When used on samples from naturally infected ibex, LFIA showed high concordance, as well as relative sensitivity and specificity (>97.25%) in comparison with RBT and CFT. This work shows high reliability and ensures a better standardization of LFIA testing for wild ruminants.
Collapse
Affiliation(s)
- Luca Freddi
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, ANSES/Paris-Est University, 94700 Maisons-Alfort, France (M.R.); (M.J.)
| | - Acacia Ferreira Vicente
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, ANSES/Paris-Est University, 94700 Maisons-Alfort, France (M.R.); (M.J.)
| | - Elodie Petit
- French Office for Biodiversity (OFB), Research and Scientific Support, 74320 Sévrier, France
- The Biometrics and Evolutionary Biology Laboratory UMR 5558, CNRS, VetAgro Sup, Université de Lyon, 69622 Villeurbanne, France
| | - Maëline Ribeiro
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, ANSES/Paris-Est University, 94700 Maisons-Alfort, France (M.R.); (M.J.)
| | - Yvette Game
- Departmental Veterinary Laboratory of Savoie (LDAV 73), 73000 Chambéry, France
| | - Yann Locatelli
- Réserve Zoologique de la Haute Touche, Muséum National d’Histoire Naturelle (MNHN), 36290 Obterre, France
| | - Isabelle Jacques
- Institut Universitaire Technologique (IUT), Département Génie Biologique, Université de Tours, 37082 Tours, France
- UMR-1282 Infectiologie et Santé Publique (ISP), INRAE Centre Val de Loire, Université de Tours, 37380 Nouzilly, France
| | - Mickaël Riou
- UE-1277 Plateforme d’Infectiologie Expérimentale, INRAE Centre Val de Loire, 37380 Nouzilly, France
| | - Maryne Jay
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, ANSES/Paris-Est University, 94700 Maisons-Alfort, France (M.R.); (M.J.)
| | - Bruno Garin-Bastuji
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, ANSES/Paris-Est University, 94700 Maisons-Alfort, France (M.R.); (M.J.)
| | - Sophie Rossi
- French Office for Biodiversity (OFB), Research and Scientific Support, 74320 Sévrier, France
| | - Vitomir Djokic
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, ANSES/Paris-Est University, 94700 Maisons-Alfort, France (M.R.); (M.J.)
| | - Claire Ponsart
- EU/WOAH & National Reference Laboratory for Animal Brucellosis, ANSES/Paris-Est University, 94700 Maisons-Alfort, France (M.R.); (M.J.)
| |
Collapse
|
37
|
Distel JS, Di Venanzio G, Mackel JJ, Rosen DA, Feldman MF. Replicative Acinetobacter baumannii strains interfere with phagosomal maturation by modulating the vacuolar pH. PLoS Pathog 2023; 19:e1011173. [PMID: 37294840 DOI: 10.1371/journal.ppat.1011173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/23/2023] [Indexed: 06/11/2023] Open
Abstract
Bacterial pneumonia is a common infection of the lower respiratory tract that can afflict patients of all ages. Multidrug-resistant strains of Acinetobacter baumannii are increasingly responsible for causing nosocomial pneumonias, thus posing an urgent threat. Alveolar macrophages play a critical role in overcoming respiratory infections caused by this pathogen. Recently, we and others have shown that new clinical isolates of A. baumannii, but not the common lab strain ATCC 19606 (19606), can persist and replicate in macrophages within spacious vacuoles that we called Acinetobacter Containing Vacuoles (ACV). In this work, we demonstrate that the modern A. baumannii clinical isolate 398, but not the lab strain 19606, can infect alveolar macrophages and produce ACVs in vivo in a murine pneumonia model. Both strains initially interact with the alveolar macrophage endocytic pathway, as indicated by EEA1 and LAMP1 markers; however, the fate of these strains diverges at a later stage. While 19606 is eliminated in an autophagy pathway, 398 replicates in ACVs and are not degraded. We show that 398 reverts the natural acidification of the phagosome by secreting large amounts of ammonia, a by-product of amino acid catabolism. We propose that this ability to survive within macrophages may be critical for the persistence of clinical A. baumannii isolates in the lung during a respiratory infection.
Collapse
Affiliation(s)
- Jesus S Distel
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Gisela Di Venanzio
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Joseph J Mackel
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - David A Rosen
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Mario F Feldman
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
38
|
Kambarev S, Borghesan E, Miller CN, Myeni S, Celli J. The Brucella abortus Type IV Effector BspA Inhibits MARCH6-Dependent ERAD To Promote Intracellular Growth. Infect Immun 2023; 91:e0013023. [PMID: 37129527 PMCID: PMC10187129 DOI: 10.1128/iai.00130-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/03/2023] Open
Abstract
Brucella abortus, the intracellular causative agent of brucellosis, relies on type IV secretion system (T4SS) effector-mediated modulation of host cell functions to establish a replicative niche, the Brucella-containing vacuole (BCV). Brucella exploits the host's endocytic, secretory, and autophagic pathways to modulate the nature and function of its vacuole from an endocytic BCV (eBCV) to an endoplasmic reticulum (ER)-derived replicative BCV (rBCV) to an autophagic egress BCV (aBCV). A role for the host ER-associated degradation pathway (ERAD) in the B. abortus intracellular cycle was recently uncovered, as it is enhanced by the T4SS effector BspL to control the timing of aBCV-mediated egress. Here, we show that the T4SS effector BspA also interferes with ERAD, yet to promote B. abortus intracellular proliferation. BspA was required for B. abortus replication in bone marrow-derived macrophages and interacts with membrane-associated RING-CH-type finger 6 (MARCH6), a host E3 ubiquitin ligase involved in ERAD. Pharmacological inhibition of ERAD and small interfering RNA (siRNA) depletion of MARCH6 did not affect the replication of wild-type B. abortus but rescued the replication defect of a bspA deletion mutant, while depletion of the ERAD component UbxD8 affected replication of B. abortus and rescued the replication defect of the bspA mutant. BspA affected the degradation of ERAD substrates and destabilized the MARCH6 E3 ligase complex. Taken together, these findings indicate that BspA inhibits the host ERAD pathway via targeting of MARCH6 to promote B. abortus intracellular growth. Our data reveal that targeting ERAD components by type IV effectors emerges as a multifaceted theme in Brucella pathogenesis.
Collapse
Affiliation(s)
- Stanimir Kambarev
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Elizabeth Borghesan
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Cheryl N. Miller
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Sebenzile Myeni
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Jean Celli
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
- Department of Microbiology and Molecular Genetics, Larner College of Medicine at the University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
39
|
de Carvalho TP, da Silva LA, Castanheira TLL, de Souza TD, da Paixão TA, Lazaro-Anton L, Tsolis RM, Santos RL. Cell and Tissue Tropism of Brucella spp. Infect Immun 2023; 91:e0006223. [PMID: 37129522 PMCID: PMC10187126 DOI: 10.1128/iai.00062-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
Brucella spp. are facultatively intracellular bacteria that can infect, survive, and multiply in various host cell types in vivo and/or in vitro. The genus Brucella has markedly expanded in recent years with the identification of novel species and hosts, which has revealed additional information about the cell and tissue tropism of these pathogens. Classically, Brucella spp. are considered to have tropism for organs that contain large populations of phagocytes such as lymph nodes, spleen, and liver, as well as for organs of the genital system, including the uterus, epididymis, testis, and placenta. However, experimental infections of several different cultured cell types indicate that Brucella may actually have a broader cell tropism than previously thought. Indeed, recent studies indicate that certain Brucella species in particular hosts may display a pantropic distribution in vivo. This review discusses the available knowledge on cell and tissue tropism of Brucella spp. in natural infections of various host species, as well as in experimental animal models and cultured cells.
Collapse
Affiliation(s)
- Thaynara Parente de Carvalho
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| | - Laice Alves da Silva
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thaís Larissa Lourenço Castanheira
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto Federal de Educação Ciência e Tecnologia do Norte de Minas Gerais, Salinas, Brazil
| | - Tayse Domingues de Souza
- Escuela de Medicina Veterinaria, Facultad de Agronomía e Ingeniería Forestal, Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tatiane Alves da Paixão
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leticia Lazaro-Anton
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| | - Renee M. Tsolis
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| | - Renato Lima Santos
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| |
Collapse
|
40
|
Bányász B, Antal J, Dénes B. False Positives in Brucellosis Serology: Wrong Bait and Wrong Pond? Trop Med Infect Dis 2023; 8:tropicalmed8050274. [PMID: 37235322 DOI: 10.3390/tropicalmed8050274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
This review summarizes the status of resolving the problem of false positive serologic results (FPSR) in Brucella serology, compiles our knowledge on the molecular background of the problem, and highlights some prospects for its resolution. The molecular basis of the FPSRs is reviewed through analyzing the components of the cell wall of Gram-negative bacteria, especially the surface lipopolysaccharide (LPS) with details related to brucellae. After evaluating the efforts that have been made to solve target specificity problems of serologic tests, the following conclusions can be drawn: (i) resolving the FPSR problem requires a deeper understanding than we currently possess, both of Brucella immunology and of the current serology tests; (ii) the practical solutions will be as expensive as the related research; and (iii) the root cause of FPSRs is the application of the same type of antigen (S-type LPS) in the currently approved tests. Thus, new approaches are necessary to resolve the problems stemming from FPSR. Such approaches suggested by this paper are: (i) the application of antigens from R-type bacteria; or (ii) the further development of specific brucellin-based skin tests; or (iii) the application of microbial cell-free DNA as analyte, whose approach is detailed in this paper.
Collapse
Affiliation(s)
- Borbála Bányász
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine Budapest, 1143 Budapest, Hungary
- Laboratory of Immunology, Veterinary Diagnostic Directorate, National Food Chain Safety Office, 1143 Budapest, Hungary
| | - József Antal
- Omixon Biocomputing Ltd., 1117 Budapest, Hungary
| | - Béla Dénes
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine Budapest, 1143 Budapest, Hungary
| |
Collapse
|
41
|
Altamirano-Silva P, Meza-Torres J, Zúñiga-Pereira AM, Zamora-Jaen S, Pietrosemoli N, Cantos G, Peltier J, Pizarro-Cerdá J, Moreno E, Chacón-Díaz C, Guzmán-Verri C, Chaves-Olarte E. Phenotypes controlled by the Brucella abortus two component system BvrR/BvrS are differentially impacted by BvrR phosphorylation. Front Microbiol 2023; 14:1148233. [PMID: 37234533 PMCID: PMC10206243 DOI: 10.3389/fmicb.2023.1148233] [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: 01/19/2023] [Accepted: 04/14/2023] [Indexed: 05/28/2023] Open
Abstract
Brucella abortus is a zoonotic pathogen whose virulence depends on its ability to survive intracellularly at the endoplasmic reticulum derived compartment. The two-component system BvrR/BvrS (BvrRS) is essential for intracellular survival due to the transcriptional control of the type IV secretion system VirB and its transcriptional regulator VjbR. It is a master regulator of several traits including membrane homeostasis by controlling gene expression of membrane components, such as Omp25. BvrR phosphorylation is related to DNA binding at target regions, thereby repressing or activating gene transcription. To understand the role of BvrR phosphorylation we generated dominant positive and negative versions of this response regulator, mimicking phosphorylated and non-phosphorylated BvrR states and, in addition to the wild-type version, these variants were introduced in a BvrR negative background. We then characterized BvrRS-controlled phenotypes and assessed the expression of proteins regulated by the system. We found two regulatory patterns exerted by BvrR. The first pattern was represented by resistance to polymyxin and expression of Omp25 (membrane conformation) which were restored to normal levels by the dominant positive and the wild-type version, but not the dominant negative BvrR. The second pattern was represented by intracellular survival and expression of VjbR and VirB (virulence) which were, again, complemented by the wild-type and the dominant positive variants of BvrR but were also significantly restored by complementation with the dominant negative BvrR. These results indicate a differential transcriptional response of the genes controlled to the phosphorylation status of BvrR and suggest that unphosphorylated BvrR binds and impacts the expression of a subset of genes. We confirmed this hypothesis by showing that the dominant negative BvrR did not interact with the omp25 promoter whereas it could interact with vjbR promoter. Furthermore, a global transcriptional analysis revealed that a subset of genes responds to the presence of the dominant negative BvrR. Thus, BvrR possesses diverse strategies to exert transcriptional control on the genes it regulates and, consequently, impacting on the phenotypes controlled by this response regulator.
Collapse
Affiliation(s)
- Pamela Altamirano-Silva
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Jazmín Meza-Torres
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Ana Mariel Zúñiga-Pereira
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Sigrid Zamora-Jaen
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Natalia Pietrosemoli
- Bioinformatics and Biostatistics Hub, CNRS USR3756, Institut Pasteur, Université Paris Cité, Paris, France
| | - Gabriela Cantos
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Johann Peltier
- Laboratoire Pathogenèse des Bactéries Anaérobies, CNRS UMR6047, Institut Pasteur, Université Paris Cité, Paris, France
- CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, Gif-sur-Yvette, France
| | - Javier Pizarro-Cerdá
- Yersinia Research Unit, CNRS UMR6047, Institut Pasteur, Université Paris Cité, Paris, France
| | - Edgardo Moreno
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Carlos Chacón-Díaz
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Caterina Guzmán-Verri
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Esteban Chaves-Olarte
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
42
|
English BC, Savage HP, Mahan SP, Diaz-Ochoa VE, Young BM, Abuaita BH, Sule G, Knight JS, O’Riordan MX, Bäumler AJ, Tsolis RM. The IRE1α-XBP1 Signaling Axis Promotes Glycolytic Reprogramming in Response to Inflammatory Stimuli. mBio 2023; 14:e0306822. [PMID: 36475773 PMCID: PMC9973330 DOI: 10.1128/mbio.03068-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 12/14/2022] Open
Abstract
Immune cells must be able to adjust their metabolic programs to effectively carry out their effector functions. Here, we show that the endoplasmic reticulum (ER) stress sensor Inositol-requiring enzyme 1 alpha (IRE1α) and its downstream transcription factor X box binding protein 1 (XBP1) enhance the upregulation of glycolysis in classically activated macrophages (CAMs). The IRE1α-XBP1 signaling axis supports this glycolytic switch in macrophages when activated by lipopolysaccharide (LPS) stimulation or infection with the intracellular bacterial pathogen Brucella abortus. Importantly, these different inflammatory stimuli have distinct mechanisms of IRE1α activation; while Toll-like receptor 4 (TLR4) supports glycolysis under both conditions, TLR4 is required for activation of IRE1α in response to LPS treatment but not B. abortus infection. Though IRE1α and XBP1 are necessary for maximal induction of glycolysis in CAMs, activation of this pathway is not sufficient to increase the glycolytic rate of macrophages, indicating that the cellular context in which this pathway is activated ultimately dictates the cell's metabolic response and that IRE1α activation may be a way to fine-tune metabolic reprogramming. IMPORTANCE The immune system must be able to tailor its response to different types of pathogens in order to eliminate them and protect the host. When confronted with bacterial pathogens, macrophages, frontline defenders in the immune system, switch to a glycolysis-driven metabolism to carry out their antibacterial functions. Here, we show that IRE1α, a sensor of ER stress, and its downstream transcription factor XBP1 support glycolysis in macrophages during infection with Brucella abortus or challenge with Salmonella LPS. Interestingly, these stimuli activate IRE1α by independent mechanisms. While the IRE1α-XBP1 signaling axis promotes the glycolytic switch, activation of this pathway is not sufficient to increase glycolysis in macrophages. This study furthers our understanding of the pathways that drive macrophage immunometabolism and highlights a new role for IRE1α and XBP1 in innate immunity.
Collapse
Affiliation(s)
- Bevin C. English
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Hannah P. Savage
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Scott P. Mahan
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Vladimir E. Diaz-Ochoa
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Briana M. Young
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Basel H. Abuaita
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Gautam Sule
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mary X. O’Riordan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Renée M. Tsolis
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| |
Collapse
|
43
|
Abstract
Host cell egress is a critical step in the life cycle of intracellular pathogens, especially in microbes capable of establishing chronic infections. The Gram-negative bacterium Brucella belongs to such a group of pathogens. Even though much has been done to understand how Brucella avoids killing and multiplies in its intracellular niche, the mechanism that this bacterium deploys to egress from the cell to complete its cycle has been poorly studied. In the manuscript, we quantify the kinetics of bacterial egress and show that Brucella exploits multivesicular bodies to exit host cells. For the first time, we visualized the process of egress in real time by live video microscopy and showed that a population of intracellular bacteria exit from host cells in vacuoles containing multivesicular body-like features. We observed the colocalization of Brucella with two multivesicular markers, namely, CD63 and LBPA, both during the final stages of the intracellular life cycle and in egressed bacteria. Moreover, drugs that either promote or inhibit multivesicular bodies either increased or decreased the number of extracellular bacteria, respectively. Our results strongly suggest that Brucella hijacks multivesicular bodies to exit the host cells to initiate new infection events. IMPORTANCE How intracellular bacterial pathogens egress from host cells has been poorly studied. This is particularly important because this stage of the infectious cycle can have a strong impact on how the host resolves the infection. Brucella is an intracellular pathogen that infects mammals, including humans, and causes a chronic debilitating illness. The bacterium has evolved a plethora of mechanisms to invade host cells, avoid degradation in the endocytic pathway, and actively multiply within a specialized intracellular compartment. However, how this pathogen exits from infected cells to produce reinfection and complete its life cycle is poorly understood. In the manuscript, we shed some light on the mechanisms that are exploited by Brucella to egress from host cells. We observed for the first time the egress of Brucella from infected cells by time-lapse video microscopy, and we found that the bacterium exits in vesicles containing multivesicular bodies (MVBs) features. Moreover, the drug manipulation of MVBs resulted in the alteration of bacterial egress efficiency. Our results indicate that Brucella hijacks MVBs to exit host cells and that this strongly contributes to the reinfection cycle.
Collapse
|
44
|
Structure, Substrate Specificity and Role of Lon Protease in Bacterial Pathogenesis and Survival. Int J Mol Sci 2023; 24:ijms24043422. [PMID: 36834832 PMCID: PMC9961632 DOI: 10.3390/ijms24043422] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Proteases are the group of enzymes that carry out proteolysis in all forms of life and play an essential role in cell survival. By acting on specific functional proteins, proteases affect the transcriptional and post-translational pathways in a cell. Lon, FtsH, HslVU and the Clp family are among the ATP-dependent proteases responsible for intracellular proteolysis in bacteria. In bacteria, Lon protease acts as a global regulator, governs an array of important functions such as DNA replication and repair, virulence factors, stress response and biofilm formation, among others. Moreover, Lon is involved in the regulation of bacterial metabolism and toxin-antitoxin systems. Hence, understanding the contribution and mechanisms of Lon as a global regulator in bacterial pathogenesis is crucial. In this review, we discuss the structure and substrate specificity of the bacterial Lon protease, as well as its ability to regulate bacterial pathogenesis.
Collapse
|
45
|
Analysis of the Brucella suis Twin Arginine Translocation System and Its Substrates Shows That It Is Essential for Viability. Infect Immun 2023; 91:e0045922. [PMID: 36448838 PMCID: PMC9872638 DOI: 10.1128/iai.00459-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Bacteria use the twin arginine translocator (Tat) system to export folded proteins from the cytosol to the bacterial envelope or to the extracellular environment. As with most Gram-negative bacteria, the Tat system of the zoonotic pathogen Brucella spp. is encoded by a three-gene operon, tatABC. Our attempts, using several different strategies, to create a Brucella suis strain 1330 tat mutant were all unsuccessful. This suggested that, for B. suis, Tat is essential, in contrast to a recent report for Brucella melitensis. This was supported by our findings that two molecules that inhibit the Pseudomonas aeruginosa Tat system also inhibit B. suis, B. melitensis, and Brucella abortus growth in vitro. In a bioinformatic screen of the B. suis 1330 proteome, we identified 28 proteins with putative Tat signal sequences. We used a heterologous reporter assay based on export of the Tat-dependent amidase AmiA by using the Tat signal sequences from the Brucella proteins to confirm that 20 of the 28 candidates can engage the Tat pathway.
Collapse
|
46
|
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
|
47
|
Li J, Qi L, Diao Z, Zhang M, Li B, Zhai Y, Hao M, Zhou D, Liu W, Jin Y, Wang A. Brucella BtpB Manipulates Apoptosis and Autophagic Flux in RAW264.7 Cells. Int J Mol Sci 2022; 23:ijms232214439. [PMID: 36430916 PMCID: PMC9693124 DOI: 10.3390/ijms232214439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/02/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Brucella transfers effectors into host cells, manipulating cellular processes to its advantage; however, the mechanism by which effectors regulate cellular processes during infection is poorly understood. A growing number of studies have shown that apoptosis and autophagy are critical mechanisms for target cells to cope with pathogens and maintain cellular homeostasis. BtpB is a Brucella type IV secretion system effector with a complex mechanism for manipulating host infection. Here, we show that the ectopic expression of BtpB promoted DNA fragmentation. In contrast, an isogenic mutant strain, ΔbtpB, inhibited apoptosis compared to the wild-type strain B. suis S2 in RAW264.7 cells. In addition, BtpB inhibited autophagy, as determined by LC3-II protein levels, the number of LC3 puncta, and p62 degradation. We also found that BtpB reduced autophagolysosome formation and blocked the complete autophagic flux. Moreover, our results revealed that the autophagy inhibitor, chloroquine, reduces Brucella's intracellular survival. Overall, our data unveil new mechanisms of virulence implicating the effector BtpB in regulating host intracellular infection.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Ziyang Diao
- 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
| | - 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
| | - Bin 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
| | - 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
- Correspondence: or
| |
Collapse
|
48
|
Serafino A, Marin Franco JL, Maio M, Trotta A, Genoula M, Castillo LA, Birnberg Weiss F, Pittaluga JR, Balboa L, Barrionuevo P, Milillo MA. Brucella abortus RNA does not polarize macrophages to a particular profile but interferes with M1 polarization. PLoS Negl Trop Dis 2022; 16:e0010950. [PMID: 36441810 PMCID: PMC9731426 DOI: 10.1371/journal.pntd.0010950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 12/08/2022] [Accepted: 11/14/2022] [Indexed: 11/29/2022] Open
Abstract
Monocytes and macrophages play a central role in chronic brucellosis. Brucella abortus (Ba) is an intracellular pathogen that survives inside these cells. On the other hand, macrophages could be differentiated into classical (M1), alternative (M2) or other less-identified profiles. We have previously shown that Ba RNA (a bacterial viability-associated PAMP or vita-PAMP) is a key molecule by which Ba can evade the host immune response. However, we did not know if macrophages could be polarized by this vita-PAMP. To assess this, we used two different approaches: we evaluated if Ba RNA per se was able to differentiate macrophages to M1 or M2 or, given that Ba survives inside macrophages once a Th1 response is established (i.e., in the presence of IFN-γ), we also analysed if Ba RNA could interfere with M1 polarization. We found that Ba RNA alone does not polarize to M1 or M2 but activates human macrophages instead. However, our results show that Ba RNA does interfere with M1 polarization while they are being differentiated. This vita-PAMP diminished the M1-induced CD64, and MHC-II surface expression on macrophages at 48 h. This phenomenon was not associated with an alternative activation of these cells (M2), as shown by unchanged CD206, DC-SIGN and CD163 surface expression. When evaluating glucose metabolism, we found that Ba RNA did not modify M1 glucose consumption or lactate production. However, production of Nitrogen Reactive Species (NRS) did diminish in Ba RNA-treated M1 macrophages. Overall, our results show that Ba RNA could alter the proper immune response set to counterattack the bacteria that could persist in the host establishing a chronic infection.
Collapse
Affiliation(s)
- Agustina Serafino
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - José L. Marin Franco
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Mariano Maio
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Aldana Trotta
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Melanie Genoula
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Luis A. Castillo
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Federico Birnberg Weiss
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - José R. Pittaluga
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Luciana Balboa
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Paula Barrionuevo
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - M. Ayelén Milillo
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| |
Collapse
|
49
|
Guo J, Deng X, Zhang Y, Song S, Zhao T, Zhu D, Cao S, Baryshnikov PI, Cao G, Blair HT, Chen C, Gu X, Liu L, Zhang H. The Flagellar Transcriptional Regulator FtcR Controls Brucella melitensis 16M Biofilm Formation via a betI-Mediated Pathway in Response to Hyperosmotic Stress. Int J Mol Sci 2022; 23:ijms23179905. [PMID: 36077302 PMCID: PMC9456535 DOI: 10.3390/ijms23179905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
The expression of flagellar proteins in Brucella species likely evolved through genetic transference from other microorganisms, and contributed to virulence, adaptability, and biofilm formation. Despite significant progress in defining the molecular mechanisms behind flagellar gene expression, the genetic program controlling biofilm formation remains unclear. The flagellar transcriptional factor (FtcR) is a master regulator of the flagellar system’s expression, and is critical for B. melitensis 16M’s flagellar biogenesis and virulence. Here, we demonstrate that FtcR mediates biofilm formation under hyperosmotic stress. Chromatin immunoprecipitation with next-generation sequencing for FtcR and RNA sequencing of ftcR-mutant and wild-type strains revealed a core set of FtcR target genes. We identified a novel FtcR-binding site in the promoter region of the osmotic-stress-response regulator gene betI, which is important for the survival of B. melitensis 16M under hyperosmotic stress. Strikingly, this site autoregulates its expression to benefit biofilm bacteria’s survival under hyperosmotic stress. Moreover, biofilm reduction in ftcR mutants is independent of the flagellar target gene fliF. Collectively, our study provides new insights into the extent and functionality of flagellar-related transcriptional networks in biofilm formation, and presents phenotypic and evolutionary adaptations that alter the regulation of B. melitensis 16M to confer increased tolerance to hyperosmotic stress.
Collapse
Affiliation(s)
- Jia Guo
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
| | - Xingmei Deng
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
| | - Yu Zhang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
| | - Shengnan Song
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
| | - Tianyi Zhao
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
| | - Dexin Zhu
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
| | - Shuzhu Cao
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
| | - Peter Ivanovic Baryshnikov
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
- College of Veterinary, Altai State Agricultural University, 656000 Barnaul, Russia
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430000, China
| | - Hugh T. Blair
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
- International Sheep Research Center, School of Agriculture and Environment, Massey University, Palmerston North 4442, New Zealand
| | - Chuangfu Chen
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
| | - Xinli Gu
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
| | - Liangbo Liu
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
- Correspondence: (L.L.); (H.Z.); Tel.: +86-0993-2057971 (L.L. & H.Z.)
| | - Hui Zhang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
- Correspondence: (L.L.); (H.Z.); Tel.: +86-0993-2057971 (L.L. & H.Z.)
| |
Collapse
|
50
|
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
|