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
|
Çağan E, Kızmaz MA, Akalın EH, Oral HB, Tezcan G, Budak F. New biological markers in diagnosis and follow-up of brucellosis cases. Diagn Microbiol Infect Dis 2025; 111:116587. [PMID: 39550977 DOI: 10.1016/j.diagmicrobio.2024.116587] [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: 07/29/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024]
Abstract
Brucellosis remains a significant public health issue in some parts of the world. It is clear that new laboratory methods are needed to diagnose brucellosis. Currently, no test method meets the criteria of high specificity, sensitivity, reliability, and low cost for the diagnosis of brucellosis, which could also predict chronicity. This study was conducted based on the data from a study conducted in 2015, which aimed to reveal genes with different transcript levels in chronic and acute patients and to evaluate their effects on the progression to chronicity by studying mRNA microarray and miRNA array in peripheral blood mononuclear cells in acute, chronic brucellosis and healthy control groups. According to the data obtained in this study, a second study was conducted to determine new markers that could aid in diagnosis and/or predict chronicity, with the most prominent gene products being [ABI3 (ABL interactor), PIAS4 (Protein Inhibitor of Activated STAT 4), PPP2R4 (Protein Phosphatase 2 Phosphatase Activator), DDIT4L (DNA Damage Inducible Transcript 4 Like), WDR33 (WD Repeat-Containing Protein 33), and IDO (Indoleamine 2,3-Dioxygenase)]. The study speculates that increased levels of ABI3, CLEC12B, PPP2R4 and decreased levels of DDIT4L, PIAS4, and IDO may be used as markers for the diagnosis of acute brucellosis, decreased levels of ABI3, CLEC12B, PPP2R4 and increased levels of DDIT4L, PIAS4, IDO may be assessed for treatment response. The study also suggested that maintaining consistent levels of ABI3, CLEC12B, PIAS4, PPP2R4, and IDO in subsequent titers may serve as a potential marker to predict chronic progression.
Collapse
Affiliation(s)
- E Çağan
- Department of Pediatric Infectious Diseases, Bursa Yuksek Ihtisas Training and Research Hospital, University of Health Sciences, Bursa, Turkey; Department of Immunology, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
| | - M A Kızmaz
- Department of Immunology, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
| | - E H Akalın
- Department of Infectious Diseases and Clinical Microbiology, Bursa Uludag University School of Medicine, Bursa, Turkey
| | - H B Oral
- Department of Immunology, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
| | - G Tezcan
- Department of Fundamental Sciences, Faculty of Dentistry, Bursa Uludag University, Bursa, Turkey
| | - F Budak
- Department of Immunology, Bursa Uludag University Faculty of Medicine, Bursa, Turkey.
| |
Collapse
|
3
|
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
|
4
|
Wen J, Li Z, Lv Y, Ding S, Zhu Y, Yang J, Tang J, Zhu M, Zhao Y, Zhao W. A subunit vaccine based on Brucella rBP26 induces Th1 immune responses and M1 macrophage activation. Acta Biochim Biophys Sin (Shanghai) 2024; 56:879-891. [PMID: 38419498 PMCID: PMC11292127 DOI: 10.3724/abbs.2024023] [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: 12/02/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Brucellosis is a global zoonotic infection caused by Brucella bacteria, which poses a significant burden on society. While transmission prevention is currently the most effective method, the absence of a licenced vaccine for humans necessitates the urgent development of a safe and effective vaccine. Recombinant protein-based subunit vaccines are considered promising options, and in this study, the Brucella BP26 protein is expressed using prokaryotic expression systems. The immune responses are evaluated using the well-established adjuvant CpG-ODN. The results demonstrate that rBP26 supplemented with a CpG adjuvant induces M1 macrophage polarization and stimulates cellular immune responses mediated by Th1 cells and CD8 + T cells. Additionally, it generates high levels of rBP26-specific antibodies in immunized mice. Furthermore, rBP26 immunization activates, proliferates, and produces cytokines in T lymphocytes while also maintaining immune memory for an extended period of time. These findings shed light on the potential biological function of rBP26, which is crucial for understanding brucellosis pathogenesis. Moreover, rBP26 holds promise as an effective subunit vaccine candidate for use in endemic areas.
Collapse
Affiliation(s)
- Jia Wen
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- General Medicine DepartmentGeneral Hospital of Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Zihua Li
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Yongxue Lv
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Shuqin Ding
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Yazhou Zhu
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Jihui Yang
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Jing Tang
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Mingxing Zhu
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Yinqi Zhao
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Wei Zhao
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| |
Collapse
|
5
|
Rezaeepoor M, Keramat F, Jourghasemi S, Rahmanpour M, Lipsa A, Hajilooi M, Solgi G. MicroRNA -21 expression as an auxiliary diagnostic biomarker of acute brucellosis. Mol Biol Rep 2024; 51:264. [PMID: 38302783 DOI: 10.1007/s11033-023-09193-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/21/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND This study aimed to measure the expression levels of peripheral blood miRNAs in brucellosis and their involvement in the different phases of the brucellosis. METHODS The expression levels of miRNAs including miR-210, miR-155, miR-150, miR-146a, miR-139-3p, miR-125a-5p, miR-29 and miR-21 were quantified in 57 brucellosis patients subgrouped into acute, under treatment & relapse phase and 30 healthy controls (HCs) using real-time polymerase chain reaction (RT-PCR). The receiver operating characteristic (ROC) analysis curve analysis was performed to find a biomarker for discrimination of different phases of brucellosis. RESULTS The expression of miR-155, miR-146a, miR-125a-5p, miR-29, and miR-21 was found to be elevated in the acute brucellosis patients compared to HCs. miR-29 changed in under-treatment patients, while miR-139-3p and miR-125a-5p showed alterations in relapse cases. The ROC curve analysis depicted the potential involvement of miR-21 in the pathogenesis of acute brucellosis. CONCLUSION The expression level of miR-21 is significantly augmented in acute brucellosis and has the potential to be a contributing diagnostic factor for acute infection.
Collapse
Affiliation(s)
- Mahsa Rezaeepoor
- Department of Immunology, Faculty of Medicine, Hamadan University of Medical Sciences, Shariati Ave, Opposite to Lona Park, P.O. Box: 6517838736, Hamadan, Iran
| | - Fariba Keramat
- Department of Immunology, Faculty of Medicine, Hamadan University of Medical Sciences, Shariati Ave, Opposite to Lona Park, P.O. Box: 6517838736, Hamadan, Iran
- Department of Infectious Diseases, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sanaz Jourghasemi
- Department of Immunology, Faculty of Medicine, Hamadan University of Medical Sciences, Shariati Ave, Opposite to Lona Park, P.O. Box: 6517838736, Hamadan, Iran
| | - Mina Rahmanpour
- Department of Immunology, Faculty of Medicine, Hamadan University of Medical Sciences, Shariati Ave, Opposite to Lona Park, P.O. Box: 6517838736, Hamadan, Iran
| | - Anuja Lipsa
- Cancer Genetic Laboratory, Advanced Centre for Treatment Research and Education in Cancer-Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Mehrdad Hajilooi
- Department of Immunology, Faculty of Medicine, Hamadan University of Medical Sciences, Shariati Ave, Opposite to Lona Park, P.O. Box: 6517838736, Hamadan, Iran.
| | - Ghasem Solgi
- Department of Immunology, Faculty of Medicine, Hamadan University of Medical Sciences, Shariati Ave, Opposite to Lona Park, P.O. Box: 6517838736, Hamadan, Iran.
| |
Collapse
|
6
|
Hou S, Kong F, Li X, Xu Y, Chen S, Zhang S, Zhang L, Li T, Fu Y, Li C, Wang W. Role of myeloid-derived suppressor cells in chronic brucellosis. Front Cell Infect Microbiol 2024; 14:1347883. [PMID: 38352057 PMCID: PMC10861671 DOI: 10.3389/fcimb.2024.1347883] [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: 12/01/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Human brucellosis, a Brucella infection caused most common zoonosis in the world, remains a serious public health burden in China. Brucella chronic infection always causes immunosuppressive status and results in severe organ or tissue damages. The aim of this work was to study the role of the myeloid-derived suppressor cells (MDSCs) in human chronic brucellosis. Methods Fifty cases of chronic brucellosis and 40 healthy individual controls were enrolled in this study. We analyzed the frequency and subsets of MDSCs in PBMC between the chronic brucellosis and healthy control groups by flow cytometry. Furthermore, we also measured the inflammatory-related cytokines in serum samples and the MDSCs inhibition ability to the proliferation of T cells in vitro. Results We found that the frequency of MDSCs in peripheral blood and the level of IL-6 and IL-10 Th2 cytokines and Arginase-1 were significantly increased in chronic brucellosis patients. In addition, we also found that the T cell function was suppressed in vitro by co-culturing with MDSCs from brucellosis patients. Conclusion Our study described an increase of immunosuppressive MDSCs in peripheral blood of chronic brucellosis patients. These results contribute to the understanding of Brucella persistent infection, which may provide an insight for effective treatment of chronic brucellosis patients in clinical practice.
Collapse
Affiliation(s)
- Shuiping Hou
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Department of Microbiology, Guangzhou Center for Disease Control and Prevention (CDC), Guangzhou, China
| | - Fandong Kong
- Department of Medical Administration, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xintong Li
- Department of Blood Components, Guangzhou Blood Center, Guangzhou, China
| | - Yanwen Xu
- Department of Obstetrics, He Xian Memorial Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shouyi Chen
- Department of Parasitic Disease and Endemic Disease Control and Prevention, Guangzhou Center for Disease Control and Prevention (CDC), Guangzhou, China
| | - Sheng Zhang
- Administration Office, Baoan Central Blood Station, Shenzhen, China
| | - Ling Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Tingting Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yongshui Fu
- Clinical Transfusion Institute, Guangzhou Blood Center, Guangzhou, China
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Wenjing Wang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Yang X, Goodwin ZI, Bhagyaraj E, Hoffman C, Pascual DW. Parenteral Vaccination with a Live Brucella melitensis Mutant Protects against Wild-Type B. melitensis 16M Challenge. Microorganisms 2024; 12:169. [PMID: 38257995 PMCID: PMC10820470 DOI: 10.3390/microorganisms12010169] [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: 12/05/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Susceptibility to brucellosis remains prevalent, even in herds vaccinated with conventional vaccines. Efforts are underway to develop an improved brucellosis vaccine, and possibly a universal vaccine, given that Brucella species are highly homologous. To this end, two B. melitensis mutants were developed, znBM-lacZ (znBMZ) and znBM-mCherry (znBM-mC), and were tested for their ability to confer systemic immunity against virulent B. melitensis challenge. To assess the extent of their attenuation, bone-marrow-derived macrophages and human TF-1 myeloid cells were infected with both mutants, and the inability to replicate within these cells was noted. Mice infected with varying doses of znBM-mC cleared the brucellae within 6-10 weeks. To test for efficacy against systemic disease, groups of mice were vaccinated once by the intraperitoneal route with either znBMZ or B. abortus S19 vaccine. Relative to the PBS-dosed mice, znBMZ vaccination greatly reduced splenic brucellae colonization by ~25,000-fold compared to 700-fold for S19-vaccinated mice. Not surprisingly, both znBMZ and S19 strains induced IFN-γ+ CD4+ T cells, yet only znBMZ induced IFN-γ+ CD8+ T cells. While both strains induced CD4+ effector memory T cells (Tems), only znBMZ induced CD8+ Tems. Thus, these results show that the described znBM mutants are safe, able to elicit CD4+ and CD8+ T cell immunity without a boost, and highly effective, rendering them promising vaccine candidates for livestock.
Collapse
Affiliation(s)
| | | | | | | | - David W. Pascual
- Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (X.Y.); (Z.I.G.); (E.B.); (C.H.)
| |
Collapse
|
8
|
Zhao T, Zhang Y, Liu L, Deng X, Guo J, Cao S, Zhu D, Xu J, Nikolaevna UV, Maratbek S, Wang Z, Sun Z, Gu X, Zhang H. Systemic Pharmacology Reveals the Potential Targets and Signaling Mechanisms in the Adjuvant Treatment of Brucellosis with Traditional Chinese Medicine. ACS OMEGA 2023; 8:28797-28812. [PMID: 37576692 PMCID: PMC10413447 DOI: 10.1021/acsomega.3c03716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023]
Abstract
Human brucellosis is one of the world's most common zoonoses, caused by Brucella infection and characterized by induced inflammation, which in severe cases can lead to abortion and sterility in humans and animals. There is growing evidence that traditional Chinese medicine (TCM) is beneficial as an adjunct to the treatment of brucellosis. However, its specific targets of action and molecular mechanisms remain unclear. In this study, a systematic pharmacological approach was applied to demonstrate pharmacological targets, biological functions, and signaling pathways of TCM as an adjunct to the treatment of brucellosis (TCMTB). The results of network pharmacology were further verified by in vitro experiments. Network analysis revealed that 133 active ingredients and 247 targets were screened in TCMTB. Further data analysis identified 21 core targets and 5 core compounds in TCMTB, including beta-sitosterol, quercetin, kaempferol, luteolin, and paeoniflorin. Gene ontology and the Kyoto Encyclopedia of Gene and Genome analysis showed that TCMTB might actively treat brucellosis by regulating inflammatory response, enhancing immune function, and targeting signaling pathways such as tuberculosis and TNF. Molecular docking results showed that multiple compounds could bind to multiple targets. Further, in vitro experiments confirmed that quercetin, among the active compounds screened, induced the strongest immunomodulatory and pro-inflammatory cytokine production during Brucella abortus infection. Further, quercetin induced nitric oxide production, which attenuated the ability of B. abortus to internalize THP-1 cells as well as intracellular survival. This study reveals the mechanism by which TCMTB aids in the treatment of brucellosis through a synergistic multicomponent, multipathway, and multitarget action. The contribution of quercetin treatment to B. abortus infection was demonstrated for the first time, which may be related to the quercetin-induced production of nitric oxide and immunomodulatory and inflammatory cytokines. These predictions of the core compounds and targets may be used in the future for the clinical treatment of brucellosis.
Collapse
Affiliation(s)
- Tianyi Zhao
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| | - Yu Zhang
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| | - Liangbo Liu
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| | - Xingmei Deng
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| | - Jia Guo
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| | - Shuzhu Cao
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| | - Dexin Zhu
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| | - Jian Xu
- Herbivorous
Animal Bacterial Disease Innovation Team, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural
Sciences, Lanzhou, Gansu 730046, China
| | - Usevich Vera Nikolaevna
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
- College
of Veterinary, Ural State Agricultural University, Yekaterinburg 620000, Russia
| | - Suleimenov Maratbek
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
- College
of Veterinary, Kazakh National Agricultural
University, Nur Sultan 050001, Kazakhstan
| | - Zhen Wang
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| | - Zhihua Sun
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| | - Xinli Gu
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| | - Hui Zhang
- State
International Joint Research Center for Animal Health Breeding, College
of Animal Science and Technology, Shihezi
University, Shihezi, Xinjiang 832003, China
| |
Collapse
|
9
|
Pascual DW, Goodwin ZI, Bhagyaraj E, Hoffman C, Yang X. Activation of mucosal immunity as a novel therapeutic strategy for combating brucellosis. Front Microbiol 2022; 13:1018165. [PMID: 36620020 PMCID: PMC9814167 DOI: 10.3389/fmicb.2022.1018165] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Brucellosis is a disease of livestock that is commonly asymptomatic until an abortion occurs. Disease in humans results from contact of infected livestock or consumption of contaminated milk or meat. Brucella zoonosis is primarily caused by one of three species that infect livestock, Bacillus abortus in cattle, B. melitensis in goats and sheep, and B. suis in pigs. To aid in disease prophylaxis, livestock vaccines are available, but are only 70% effective; hence, improved vaccines are needed to mitigate disease, particularly in countries where disease remains pervasive. The absence of knowing which proteins confer complete protection limits development of subunit vaccines. Instead, efforts are focused on developing new and improved live, attenuated Brucella vaccines, since these mimic attributes of wild-type Brucella, and stimulate host immune, particularly T helper 1-type responses, required for protection. In considering their development, the new mutants must address Brucella's defense mechanisms normally active to circumvent host immune detection. Vaccination approaches should also consider mode and route of delivery since disease transmission among livestock and humans is believed to occur via the naso-oropharyngeal tissues. By arming the host's mucosal immune defenses with resident memory T cells (TRMs) and by expanding the sources of IFN-γ, brucellae dissemination from the site of infection to systemic tissues can be prevented. In this review, points of discussion focus on understanding the various immune mechanisms involved in disease progression and which immune players are important in fighting disease.
Collapse
|
10
|
Mitroulis I, Chrysanthopoulou A, Divolis G, Ioannidis C, Ntinopoulou M, Tasis A, Konstantinidis T, Antoniadou C, Soteriou N, Lallas G, Mitka S, Lesche M, Dahl A, Gembardt S, Panopoulou M, Sideras P, Wielockx B, Coskun Ü, Ritis K, Skendros P. A gene expression map of host immune response in human brucellosis. Front Immunol 2022; 13:951232. [PMID: 35979363 PMCID: PMC9376622 DOI: 10.3389/fimmu.2022.951232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/13/2022] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is a common zoonotic disease caused by intracellular pathogens of the genus Brucella. Brucella infects macrophages and evades clearance mechanisms, thus resulting in chronic parasitism. Herein, we studied the molecular changes that take place in human brucellosis both in vitro and ex vivo. RNA sequencing was performed in primary human macrophages (Mφ) and polymorphonuclear neutrophils (PMNs) infected with a clinical strain of Brucella spp. We observed a downregulation in the expression of genes involved in host response, such as TNF signaling, IL-1β production, and phagosome formation in Mφ, and phosphatidylinositol signaling and TNF signaling in PMNs, being in line with the ability of the pathogen to survive within phagocytes. Further transcriptomic analysis of isolated peripheral blood mononuclear cells (PBMCs) and PMNs from patients with acute brucellosis before treatment initiation and after successful treatment revealed a positive correlation of the molecular signature of active disease with pathways associated with response to interferons (IFN). We identified 24 common genes that were significantly altered in both PMNs and PBMCs, including genes involved in IFN signaling that were downregulated after treatment in both cell populations, and IL1R1 that was upregulated. The concentration of several inflammatory mediators was measured in the serum of these patients, and levels of IFN-γ, IL-1β and IL-6 were found significantly increased before the treatment of acute brucellosis. An independent cohort of patients with chronic brucellosis also revealed increased levels of IFN-γ during relapse compared to remissions. Taken together, this study provides for the first time an in-depth analysis of the transcriptomic alterations that take place in human phagocytes upon infection, and in peripheral blood immune populations during active disease.
Collapse
Affiliation(s)
- Ioannis Mitroulis
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Akrivi Chrysanthopoulou
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - Georgios Divolis
- Biomedical Research Foundation Academy of Athens, Center for Clinical, Experimental Surgery and Translational Research, Athens, Greece
| | - Charalampos Ioannidis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Maria Ntinopoulou
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Athanasios Tasis
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Theocharis Konstantinidis
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
- Laboratory of Microbiology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Christina Antoniadou
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | | | - George Lallas
- R&D Department, P. Zafiropoulos S.A., Athens, Greece
| | - Stella Mitka
- School of Biomedical Sciences, International Hellenic University, Thessaloniki, Greece
| | - Mathias Lesche
- DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Andreas Dahl
- DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Stephanie Gembardt
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Maria Panopoulou
- Laboratory of Microbiology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Paschalis Sideras
- Biomedical Research Foundation Academy of Athens, Center for Clinical, Experimental Surgery and Translational Research, Athens, Greece
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ünal Coskun
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Konstantinos Ritis
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Panagiotis Skendros
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
- *Correspondence: Panagiotis Skendros,
| |
Collapse
|
11
|
Gheitasi R, Keramat F, Khosravi S, Hajilooi M, Pletz MW, Makarewicz O. Evaluation of Th2 and Th17 Immunity-Related Factors as Indicators of Brucellosis. Front Cell Infect Microbiol 2022; 11:786994. [PMID: 35071039 PMCID: PMC8777051 DOI: 10.3389/fcimb.2021.786994] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/13/2021] [Indexed: 01/18/2023] Open
Abstract
Objective Brucellosis is a common bacterial zoonotic infection, and greater than half a million new cases are diagnosed annually. This study investigates the expression of Th2 and Th17 immunity-related factors (Th2-LCR lncRNA, IL-25, TRAF3IP2, and IL-17RB) in different stages of Brucella infections. Material and Methods In total, 99 brucellosis patients were divided into three groups (acute = first infection before treatment, relapse = before treatment, and treated = after treatment for 6–8 weeks with doxycycline and rifampin). Thirty-three healthy volunteers represented the control group. Gene expression levels were assessed by quantitative amplification in reference to the 18S rRNA gene and statistically evaluated. Results No significant differences in the expression of these genes were observed between the control group and patients after completion of antibiotic treatment. Compared to these two groups, only Th2-LCR lncRNA and TRAF3IP2 were significantly more highly expressed in the acute group. Th2-LCR lncRNA was also significantly elevated in the relapse group. TRAF3IP2 expression was additionally significantly increased in the acute group compared to the relapse group. Conclusion IL-25 and IL-17RB failed to differentiate between the infected and noninfected groups. TRAF3IP2 and Th2-LCR lncRNA might be good indicators of brucellosis during the acute phase, but the expression levels varied strongly among patients. To verify the suitability of these factors as an indicator for brucellosis, acute infection or relapse should be investigated in further studies on larger cohorts with well-defined inclusion criteria.
Collapse
Affiliation(s)
- Reza Gheitasi
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany.,Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fariba Keramat
- Brucellosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sara Khosravi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehrdad Hajilooi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mathias W Pletz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Oliwia Makarewicz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| |
Collapse
|
12
|
Tang Y, Ma C, Sun H, Yang S, Yu F, Li X, Wang L. Serum Levels of Seven General Cytokines in Acute Brucellosis Before and After Treatment. Infect Drug Resist 2021; 14:5501-5510. [PMID: 34955644 PMCID: PMC8694408 DOI: 10.2147/idr.s341331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/02/2021] [Indexed: 01/02/2023] Open
Abstract
Purpose Human brucellosis is the most common bacterial zoonosis globally that poses a severe health threat. Despite the availability of antibiotic therapy for human brucellosis, its tendencies of chronicity and persistence may lead to severe debilitating and disabling conditions in patients. Comprehensive understanding of the immune response in brucellosis will be helpful in improving the treatment strategies. In this study, we measured serum levels of T helper cell type 1 (Th1), Th2, and Th17 cytokines in patients with acute brucellosis before and after treatment. Patients and Methods Overall, 30 patients with acute brucellosis from the Beijing Di Tan Hospital and 26 healthy controls were enrolled in this study. All the patients received a 6-week therapy regimen comprising ceftriaxone, doxycycline, and rifampicin. Serum samples were collected from patients with acute Brucella infection and healthy controls before and after treatment. Serum seven cytokine levels of Th1 (IL-2, IFN-γ, and TNF-α), Th2 (IL-4, IL-6, IL-10), and Th17 (IL-17A) were measured using cytometric bead array. Results In patients with acute infection, the IL-2, IFN-γ, and IL-10 levels were significantly increased compared with those in healthy controls (P < 0.001). After treatment, IL-2, IFN-γ, and IL-10 levels significantly decreased (P < 0.05) and the TNF-α level significantly increased compared with the corresponding baseline levels and those in healthy controls (P < 0.05). Furthermore, the IFN-γ, IL-4, and IL-10 levels were higher in patients after treatment than in healthy controls (P < 0.05). IL-2 and IL-6 levels exhibited a positive correlation with the C-reactive protein (CRP) level in acute brucellosis (P < 0.05). Conclusion Levels of most serum Th1 and Th2 cytokines were simultaneously increased in acute infection, followed by reduction in the corresponding cytokine levels and residual cytokine response during treatment. This residual immune response could represent a therapeutic opportunity that may improve the long-term clinical outcomes in patients with acute brucellosis after treatment.
Collapse
Affiliation(s)
- Yunxia Tang
- Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China.,Clinical Center for HIV/AIDS, Capital Medical University, Beijing, People's Republic of China
| | - Chenjie Ma
- Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China.,Clinical Center for HIV/AIDS, Capital Medical University, Beijing, People's Republic of China
| | - Huali Sun
- Department of Infectious Diseases, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Siyuan Yang
- Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China.,Clinical Center for HIV/AIDS, Capital Medical University, Beijing, People's Republic of China
| | - Fengting Yu
- Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China.,Clinical Center for HIV/AIDS, Capital Medical University, Beijing, People's Republic of China
| | - Xingwang Li
- Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China.,Clinical Center for HIV/AIDS, Capital Medical University, Beijing, People's Republic of China
| | - Linghang Wang
- Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China.,Clinical Center for HIV/AIDS, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
13
|
Kazemi S, Mirzaei R, Sholeh M, Karampoor S, Keramat F, Saidijam M, Alikhani MY. microRNAs in human brucellosis: A promising therapeutic approach and biomarker for diagnosis and treatment. Immun Inflamm Dis 2021; 9:1209-1218. [PMID: 34449979 PMCID: PMC8589381 DOI: 10.1002/iid3.519] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION Human brucellosis is a zoonotic bacterial disease with up to 500,000 new cases each year. The major evasion mechanisms from the host immune system by Brucella are restraint of complement pathway and Toll-like receptors signaling pathways, interference with efficient antigen presentation to CD4-positive T lymphocytes, selective subversion of autophagy pathways, inhibition of dendritic cell stimulation, inhibition of autophagolysosomal fusion, and macrophage apoptosis. Many molecular and cellular pathways contribute to brucellosis that microRNAs have a vital function in the immunopathogenesis of this disease. In this regard, these molecules apply for their roles by modulating various events like inflammatory reactions and immune defense. Recently, in the case of immunity to human brucellosis, it has been shown that microRNAs play an important role in immunity against these bacteria. METHODS AND RESULTS In this study, we tried to review the immune defense and immunopathogenesis of Brucella infection and highlight the current knowledge of the microRNAs in infected cells by Brucella pathogens. The recent findings suggest that the regulation of microRNAs expression is impaired during brucellosis infection, which may contribute to disease progression or inhibition by modulating immune responses against this pathogen. CONCLUSIONS The interplay between miRNAs and Brucella pathogens and the underlying process required comprehensive examination to unravel the novel therapeutic or diagnostic approaches.
Collapse
Affiliation(s)
- Sima Kazemi
- Department of Microbiology, School of MedicineHamadan University of Medical SciencesHamadanIran
| | - Rasoul Mirzaei
- Department of Microbiology, School of MedicineHamadan University of Medical SciencesHamadanIran
| | - Mohammad Sholeh
- Department of Microbiology, School of MedicineIran University of Medical SciencesTehranIran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research CenterIran University of Medical SciencesTehranIran
- Department of Virology, School of MedicineIran University of Medical SciencesTehranIran
| | - Fariba Keramat
- Brucellosis Research CenterHamadan University of Medical SciencesHamadanIran
| | - Massoud Saidijam
- Research Center for Molecular Medicine, School of MedicineHamadan University of Medical SciencesHamadanIran
| | - Mohammad Yousef Alikhani
- Department of Microbiology, School of MedicineHamadan University of Medical SciencesHamadanIran
- Brucellosis Research CenterHamadan University of Medical SciencesHamadanIran
| |
Collapse
|
14
|
Ukita M, Hozé N, Nemoto T, Cauchemez S, Asakura S, Makingi G, Kazwala R, Makita K. Quantitative evaluation of the infection dynamics of bovine brucellosis in Tanzania. Prev Vet Med 2021; 194:105425. [PMID: 34256237 DOI: 10.1016/j.prevetmed.2021.105425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 11/18/2022]
Abstract
Brucellosis is endemic in Tanzania. A cross-sectional study was conducted at 17 cattle farms in agro-pastoral areas in Tanzania to identify risk factors associated with the within-farm prevalence of bovine brucellosis and to quantitatively assess the infection dynamics through disease modelling. Cattle blood sampling and interviews with farmers using a structured questionnaire were conducted. A total of 673 serum samples were screened using the Rose-Bengal plate test (RBPT), and sero-positivity of RBPT-positive samples was confirmed using a competitive enzyme-linked immunosorbent assay. Zero-inflated binomial regression was performed for univariable and multivariable risk factor analyses of within-farm prevalence. Several susceptible-infectious (SI) models were compared based on deviance information criteria, and age-dependent force of infection (FOI) was measured using age-specific prevalence data for the 10 infection-positive farms. Using the diagnoses of cows on the 17 farms, the basic reproduction number, R0, was also calculated. The farm-level prevalence and animal-level adjusted prevalence were 58.8 % (10/17, 95 % confidence interval: 33.5-80.6 %) and 7.0 % (28/673, 95 % credible interval: 5.7-8.4 %), respectively. The risk factor for high within-farm prevalence was introduction of cattle from other herds. A mathematical model with constant FOI showed the annual probability of infection as 1.4 % (95 % credible interval: 1.0 %-2.0 %). The R0 was 1.07. The constant FOI could have been due to the predominant mode of infection being transmission of Brucella from contaminated aborted materials during grazing. Direct purchase of infected cattle could facilitate efficient transmission between susceptible animals through abortion.
Collapse
Affiliation(s)
- Makoto Ukita
- Veterinary Epidemiology Unit, Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai Midorimachi, Ebetsu, 069-8501, Japan
| | - Nathanael Hozé
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, UMR2000, CNRS, Paris, France
| | - Takahiro Nemoto
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, UMR2000, CNRS, Paris, France
| | - Simon Cauchemez
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, UMR2000, CNRS, Paris, France
| | - Shingo Asakura
- Veterinary Epidemiology Unit, Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai Midorimachi, Ebetsu, 069-8501, Japan
| | - George Makingi
- Sokoine University of Agriculture, Chuo Kikuu, Morogoro, P.O. Box 3000, Tanzania
| | - Rudovick Kazwala
- Sokoine University of Agriculture, Chuo Kikuu, Morogoro, P.O. Box 3000, Tanzania
| | - Kohei Makita
- Veterinary Epidemiology Unit, Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai Midorimachi, Ebetsu, 069-8501, Japan.
| |
Collapse
|
15
|
Sun HL, Ma CJ, Du XF, Yang SY, Lv X, Zhao H, Wang LH, Tang YX, Li XW, Jiang RM. Soluble IL-2Rα correlates with imbalances of Th1/Th2 and Tc1/Tc2 cells in patients with acute brucellosis. Infect Dis Poverty 2020; 9:92. [PMID: 32660627 PMCID: PMC7359011 DOI: 10.1186/s40249-020-00699-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/11/2020] [Indexed: 01/18/2023] Open
Abstract
Background Previous studies showed that soluble IL-2Rα is an important marker of cellular immune activation and might be a marker of treatment efficacy for children with brucellosis. However, data regarding adult patients with brucellosis were unknown. The aim of study was to explore the potential role of serum sIL-2Rα evaluating treatment responses in adult patients with brucellosis, and T cell immune status was also examined. Methods During January 2016–April 2017, 30 patients with acute brucellosis from the Third People’s Hospital of Linfen in Shanxi Province and Beijing Di Tan Hospital, and 28 healthy controls were included in this study. Peripheral blood samples were collected before and after six weeks of antibiotic treatment. Serum sIL-2Rα levels were measured by enzyme-linked immunosorbent assay, and the percentage of Th1, Th2, Tc1, Tc2, and Tregs was detected by flow cytometry after intracellular staining for cytokines (interferon-γ and interleukin-4) and Foxp3 in T lymphocytes from peripheral blood. The obtained data were analyzed with Wilcoxon ranked sum tests for paired values, Mann-Whitney U-tests for comparisons between patients and healthy controls, and Spearman rank tests for correlation analyses. Results Serum sIL-2Rα levels were significantly higher in patients than in controls (P = 0.001). A significant decline was observed in patients after the cessation of treatment (P < 0.001) and return to normal (P > 0.05). Th1, Tc1, Th2, and Tc2 cell frequencies were higher in patients than in healthy subjects (P < 0.05), while the Th1/Th2 and Tc1/Tc2 ratios were significantly lower (P = 0.0305 and 0.0005, respectively) and returned to normal levels after treatment. In patients with acute brucellosis, serum sIL-2Rα levels were negatively correlated with the Th1/Th2 ratio (r = − 0.478, P = 0.028), Tc1/Tc2 ratio (r = − 0.677, P = 0.001), and Tc1 percentage (r = − 0.516, P = 0.017). Serum sIL-2Rα and Tc2 percentages were positively correlated (r = 0.442, P = 0.045). Conclusions Based on the correlations with Th1/Th2 and Tc1/Tc2 ratios, serum sIL-2Rα levels may reflect the immune response status. sIL-2Rα may be a marker for therapeutic efficacy in acute brucellosis.
Collapse
Affiliation(s)
- Hua-Li Sun
- Department of Infectious Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Cheng-Jie Ma
- The Laboratory of Infectious Diseases Centre, Beijing Di Tan Hospital, Capital Medical University, Beijing, China
| | - Xiu-Fang Du
- Department of Infectious Diseases, the Third People Hospital, Linfen City, Shanxi Province, China
| | - Si-Yuan Yang
- The Laboratory of Infectious Diseases Centre, Beijing Di Tan Hospital, Capital Medical University, Beijing, China
| | - Xiao Lv
- Department of Laboratory Medicine, the Third People Hospital, Linfen City, Shanxi Province, China
| | - Hong Zhao
- Department of Laboratory Medicine, the Third People Hospital, Linfen City, Shanxi Province, China
| | - Ling-Hang Wang
- The Laboratory of Infectious Diseases Centre, Beijing Di Tan Hospital, Capital Medical University, Beijing, China
| | - Yun-Xia Tang
- The Laboratory of Infectious Diseases Centre, Beijing Di Tan Hospital, Capital Medical University, Beijing, China
| | - Xing-Wang Li
- Center for Infectious Diseases, Beijing Di Tan Hospital, Capital Medical University, Beijing, China
| | - Rong-Meng Jiang
- Center for Infectious Diseases, Beijing Di Tan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
16
|
Investigation of Linc-MAF-4 expression as an effective marker in brucellosis. Mol Immunol 2020; 123:60-63. [PMID: 32417631 DOI: 10.1016/j.molimm.2020.04.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 11/24/2022]
Abstract
Brucellosis is a zoonotic disease that is one of the most common infectious diseases. Cellular immunity is the main immune response against brucella. Long non coding RNAs are a new subset of genes that could regulate cell function and may gene regulation. We aim to investigate whether the level of Linc-MAF-4 and cMAF have considerable differences in brucella infection. In this experiment 99 patients with brucellosis were divided into three groups of acute, undertreatment and relapse and 30 volunteers with negative serologic tests as control group. The expression levels were detected using quantitative polymerase chain reaction (qPCR). Statistical analysis was performed using SPSS software version 25.0. Results showed that the expression of Linc-MAF-4 was significantly increased in the acute group in comparison to control and relapse groups. Also, cMAF expression was significantly increased in the relapse group versus the control group. Our study showed these genes play important roles in the immune response include regulating naïve T cell differentiation to T helper cells in Brucella infection. We propose that Linc-MAF-4 could be a potential biomarker for the screening, diagnosis and treatment of brucellosis.
Collapse
|
17
|
Eini P, Majzoobi MM, Ghasemi Basir HR, Moosavi Z, Moradi A. Comparison of the serum level of interleukin-4 in patients with brucellosis and healthy controls. J Clin Lab Anal 2020; 34:e23267. [PMID: 32100374 PMCID: PMC7370742 DOI: 10.1002/jcla.23267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Evaluation of cytokines such as interleukin-4 (IL-4) can be an important tool in examining immune responses to brucellosis. Also, determining the response rate to treatment is necessary for controlling and eradicating of disease. The review of previous studies reveals contradictory results that require further research in this regard. The aim of this study was to compare the serum level of IL-4 in patients with brucellosis and healthy controls. MATERIAL AND METHODS In this descriptive-analytical study for comparison of two groups, a total of 165 participants, including 83 patients with brucellosis and 82 non-infected people, were evaluated after matching of sex and age in Hamadan (northwest of Iran) in 2017 and the serum level of IL-4 was compared by ELISA method. The collected data were analyzed by SPSS software version 21 at 95% significant level. RESULTS Mean of age in the case and control groups were 50.25 ± 16.01 and 43.26 ± 15.6 years, respectively. The serum levels of IL-4 in the case and control groups were 1.42 ± 0.51 pg/mL and 1.31 ± 1.02 pg/mL, respectively. Based on the non-parametric Mann-Whitney test, the IL-4 level was significantly higher in the case group, compared with the control (P < .001), but no statistically significant relationship was found between serum levels of IL-4 with age, sex, and serologic titers of Wright and 2ME. CONCLUSION In patients with brucellosis, the level of IL-4 increases independently of the duration and severity of the disease, which indicates the role of this cytokine of immune system in this infectious disease.
Collapse
Affiliation(s)
- Peyman Eini
- Brucellosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Hamid Reza Ghasemi Basir
- Department of Pathology, School of medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zahra Moosavi
- Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abbas Moradi
- Department of Community Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
18
|
Priyanka, Shringi BN, Choudhary OP, Kashyap SK. Cytokines in brucellosis: biological rhythm at the interface of innate and adaptive immunity. BIOL RHYTHM RES 2019. [DOI: 10.1080/09291016.2019.1613794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Priyanka
- Department of Veterinary Microbiology and Biotechnology, College of Veterinary and Animal Sciences, Rajasthan University of Veterinary and Animal Sciences, Bikaner, Rajasthan, India
| | - Brij Nandan Shringi
- Department of Veterinary Microbiology and Biotechnology, College of Veterinary and Animal Sciences, Rajasthan University of Veterinary and Animal Sciences, Bikaner, Rajasthan, India
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy and Histology, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, Mizoram, India
| | - Sudhir Kumar Kashyap
- Department of Veterinary Microbiology and Biotechnology, College of Veterinary and Animal Sciences, Rajasthan University of Veterinary and Animal Sciences, Bikaner, Rajasthan, India
| |
Collapse
|
19
|
Amjadi O, Rafiei A, Mardani M, Zafari P, Zarifian A. A review of the immunopathogenesis of Brucellosis. Infect Dis (Lond) 2019; 51:321-333. [PMID: 30773082 DOI: 10.1080/23744235.2019.1568545] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Brucellosis, caused by the intracellular pathogens Brucella, is one of the major zoonotic infections. Considering the economic burden, its prevalence has been a health concern especially in endemic regions. Brucella is able to survive and replicate within host cells by expressing different virulence factors and using various strategies to avoid the host's immune response. This leads to progression of the disease from an acute phase to chronic brucellosis. Exploration of genetic variations has confirmed the expected influence of gene polymorphisms on susceptibility and resistance to brucellosis of humans. Since there is no approved human vaccine and treatment is uncertain with risk of relapse, it is important to increase knowledge about pathogenesis, diagnosis and treatment of brucellosis in order to manage and control this infection, especially in endemic regions.
Collapse
Affiliation(s)
- Omolbanin Amjadi
- a Student Research Committee, Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran
| | - Alireza Rafiei
- b Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran
| | - Masoud Mardani
- c Infectious Diseases and Tropical Medicine Research Center , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Parisa Zafari
- a Student Research Committee, Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran.,b Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran
| | - Ahmadreza Zarifian
- d Infectious Disease Research Group, Student Research Committee, Medical School , Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
20
|
Lacey CA, Chambers CA, Mitchell WJ, Skyberg JA. IFN-γ-dependent nitric oxide suppresses Brucella-induced arthritis by inhibition of inflammasome activation. J Leukoc Biol 2019; 106:27-34. [PMID: 30748031 DOI: 10.1002/jlb.4mia1018-409r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/14/2019] [Accepted: 01/31/2019] [Indexed: 12/28/2022] Open
Abstract
Brucellosis, caused by the intracellular bacterial pathogen Brucella, is a globally important zoonotic disease for which arthritis is the most common focal complication in humans. Wild-type mice infected systemically with Brucella typically do not exhibit arthritis, but mice lacking IFN-γ develop arthritis regardless of the route of Brucella infection. Here, we investigated mechanisms by which IFN-γ suppresses Brucella-induced arthritis. Several cell types, including innate lymphoid cells, contributed to IFN-γ production and suppression of joint swelling. IFN-γ deficiency resulted in elevated joint IL-1β levels, and severe joint inflammation that was entirely inflammasome dependent, and in particular, reliant on the NLRP3 inflammasome. IFN-γ was vital for induction of the nitric oxide producing enzyme, iNOS, in infected joints, and nitric oxide directly inhibited IL-1β production and inflammasome activation in Brucella-infected macrophages in vitro. During in vivo infection, iNOS deficiency resulted in an increase in IL-1β and inflammation in Brucella-infected joints. Collectively, this data indicate that IFN-γ prevents arthritis both by limiting Brucella infection, and by inhibiting excessive inflammasome activation through the induction of nitric oxide.
Collapse
Affiliation(s)
- Carolyn A Lacey
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA.,Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, USA
| | - Catherine A Chambers
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA.,Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, USA
| | - William J Mitchell
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Jerod A Skyberg
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA.,Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
21
|
Higgins JL, Bowen RA, Gonzalez-Juarrero M. Cell mediated immune response in goats after experimental challenge with the virulent Brucella melitensis strain 16M and the reduced virulence strain Rev. 1. Vet Immunol Immunopathol 2018; 202:74-84. [DOI: 10.1016/j.vetimm.2018.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 06/02/2018] [Accepted: 06/05/2018] [Indexed: 11/26/2022]
|
22
|
Meta-Analysis of the Changes of Peripheral Blood T Cell Subsets in Patients with Brucellosis. J Immunol Res 2018; 2018:8439813. [PMID: 29888294 PMCID: PMC5985067 DOI: 10.1155/2018/8439813] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/24/2018] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is one of the most prevalent zoonotic diseases in the world, but its pathogenesis is not very clear. At present, it is thought that it may be related to the immunity of T cells. The conclusions of related studies are inconsistent, and its clinical significance is not explicit. We searched published articles in electronic databases up to December 2017 identified as relating to the clinical features of human brucellosis in China. Only eight studies had sufficient quality for data extraction. Meta-analysis showed a significantly decreased proportion of CD4+ T cells in human brucellosis patients compared to healthy subject individuals. The frequency of CD8+ T cells was significantly higher in human brucellosis patients than that in the healthy control group. The pooled analysis presented a significant decrease of the CD4+/CD8+ ratio in human brucellosis patients compared to healthy subjects. There is immunologic dysfunction of T lymphocyte in patients with human brucellosis, the CD4+ and CD8+ T cells might be the important factors affecting the progress of brucellosis.
Collapse
|
23
|
Cytokine Profiles in Acute Brucellosis, Compared with Those in Other Febrile Illnesses. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2017. [DOI: 10.5812/archcid.66077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
24
|
Lacey CA, Keleher LL, Mitchell WJ, Brown CR, Skyberg JA. CXCR2 Mediates Brucella-Induced Arthritis in Interferon γ-Deficient Mice. J Infect Dis 2016; 214:151-60. [PMID: 26951819 DOI: 10.1093/infdis/jiw087] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/29/2016] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Brucella species are facultative intracellular gram-negative bacteria that cause brucellosis, a common global zoonosis. Infection of the joints is the most common focal complication of brucellosis in humans. The purpose of this study was to identify mediators of focal inflammation during brucellosis. METHODS Wild-type (WT) mice are naturally resistant to Brucella infection; therefore, we infected anti-interferon γ (IFN-γ)-treated, or IFN-γ(-/-) mice with Brucella to induce osteoarticular and musculoskeletal inflammation, as we previously described. Mice were infected intraperitoneally with Brucella melitensis, and the clinical course of disease, histopathologic changes, and cytokine levels were compared among groups. RESULTS Rag1(-/-) mice (B- and T-cell deficient) and µMT(-/-) mice (B-cell deficient) developed paw inflammation at a similar rate and severity as WT mice following infection with B. melitensis and treatment with anti-IFN-γ. Joints from B. melitensis-infected IFN-γ(-/-) mice had markedly increased levels of CCR2 and CXCR2 ligands. While anti-IFN-γ-treated CCR2(-/-) and WT mice behaved similarly, anti-IFN-γ-treated CXCR2(-/-) or IFN-γ(-/-)/CXCR2(-/-) mice had strikingly reduced focal swelling relative to anti-IFN-γ-treated WT or IFN-γ(-/-) mice, respectively. Additionally, neutrophil recruitment was dependent on CXCR2. CONCLUSIONS Adaptive immune cells and CCR2 are dispensable, while CXCR2 is necessary for Brucella-induced focal neutrophil recruitment and inflammation.
Collapse
Affiliation(s)
- Carolyn A Lacey
- Department of Veterinary Pathobiology, College of Veterinary Medicine Laboratory for Infectious Disease Research, University of Missouri, Columbia
| | - Lauren L Keleher
- Department of Veterinary Pathobiology, College of Veterinary Medicine Laboratory for Infectious Disease Research, University of Missouri, Columbia
| | | | - Charles R Brown
- Department of Veterinary Pathobiology, College of Veterinary Medicine
| | - Jerod A Skyberg
- Department of Veterinary Pathobiology, College of Veterinary Medicine Laboratory for Infectious Disease Research, University of Missouri, Columbia
| |
Collapse
|
25
|
Kazemi S, Saidijam M, Hashemi SH, Karami M, Vaisi-Raygani A, Alikhani MY. Analysis of IL-10 and IL-6 Gene Polymorphisms and Their Serum Levels in Patients with Brucellosis: A Case Control Study. Immunol Invest 2016; 45:107-15. [PMID: 26849072 DOI: 10.3109/08820139.2015.1096285] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND It seems that polymorphism in the regulatory areas of cytokine genes affects the cytokine production capacity and may play a role in the development of infectious diseases. Interleukin-10 (IL-10) and Interleukin-6 (IL-6), which are cytokines of Th2, cause the macrophage become inactive and patient conditions get worse. METHODS In this case-control study, 60 patients with brucellosis and 60 healthy participants were recruited. IL-10 genotyping at positions -1082 (G/A), -819 (C/T), and -592 (C/A) and IL-6 genotyping at position -174 (G/C) were analyzed by amplification refractory mutation system-polymerase chain reaction (ARMS-PCR) and restriction fragment length polymorphism-polymerase chain reaction (RFLP-PCR) methods. The levels of IL-10 and IL-6 were determined by a sandwich enzyme-linked immunosorbent assay in sera of study population. RESULTS The AA and CC genotypes of the IL-10 gene at positions -1082 G/A and -819 C/T were significantly more frequent in patients in comparison to controls, respectively. The AG genotype of the IL-10 gene at positions -1082 G/A was significantly more frequent in control groups than the patients. Serum levels of IL-10 and IL-6 were significantly more frequent in the patients than in the control groups. CONCLUSIONS Our study showed that the AA and CC genotypes at positions -1082 and -819 are very important, respectively. These results suggest that IL-10 (-1082 G/A) GG genotype may be considered as a risk factor for brucellosis, while the AG genotype might be a protective factor against the disease.
Collapse
Affiliation(s)
- Sima Kazemi
- a Department of Microbiology, Faculty of Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Massoud Saidijam
- b Research Center for Molecular Medicine, School of Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Seyed Hamid Hashemi
- c Brucellosis Research Center , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Manoochehr Karami
- d Department of Biostatistics and Epidemiology , School of Public Health, Hamadan University of Medical Sciences , Hamadan , Iran
| | - Asad Vaisi-Raygani
- e Department of Clinical Biochemistry , Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Mohammad Yousef Alikhani
- a Department of Microbiology, Faculty of Medicine , Hamadan University of Medical Sciences , Hamadan , Iran.,b Research Center for Molecular Medicine, School of Medicine , Hamadan University of Medical Sciences , Hamadan , Iran
| |
Collapse
|
26
|
Abstract
Brucellosis, caused by bacteria of the genus Brucella, is an important zoonotic infection that causes reproductive disease in domestic animals and chronic debilitating disease in humans. An intriguing aspect of Brucella infection is the ability of these bacteria to evade the host immune response, leading to pathogen persistence. Conversely, in the reproductive tract of infected animals, this stealthy pathogen is able to cause an acute severe inflammatory response. In this review, we discuss the different mechanisms used by Brucella to cause disease, with emphasis on its virulence factors and the dichotomy between chronic persistence and reproductive disease.
Collapse
Affiliation(s)
| | - Renee M Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616; ,
| |
Collapse
|
27
|
de Souza Filho JA, de Paulo Martins V, Campos PC, Alves-Silva J, Santos NV, de Oliveira FS, Menezes GB, Azevedo V, Cravero SL, Oliveira SC. Mutant Brucella abortus membrane fusogenic protein induces protection against challenge infection in mice. Infect Immun 2015; 83:1458-64. [PMID: 25644010 PMCID: PMC4363440 DOI: 10.1128/iai.02790-14] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/25/2015] [Indexed: 01/10/2023] Open
Abstract
Brucella species can cause brucellosis, a zoonotic disease that causes serious livestock economic losses and represents a public health threat. The mechanism of virulence of Brucella spp. is not yet fully understood. Therefore, it is crucial to identify new molecules that serve as virulence factors to better understand this host-pathogen interplay. Here, we evaluated the role of the Brucella membrane fusogenic protein (Mfp) and outer membrane protein 19 (Omp19) in bacterial pathogenesis. In this study, we showed that B. abortus Δmfp::kan and Δomp19::kan deletion mutant strains have reduced persistence in vivo in C57BL/6 and interferon regulatory factor 1 (IRF-1) knockout (KO) mice. Additionally, 24 h after macrophage infection with a Δmfp::kan or Δomp19::kan strain expressing green fluorescent protein (GFP) approximately 80% or 65% of Brucella-containing vacuoles (BCVs) retained the late endosomal/lysosomal marker LAMP-1, respectively, whereas around 60% of BCVs containing wild-type S2308 were found in LAMP-1-negative compartments. B. abortus Δomp19::kan was attenuated in vivo but had a residual virulence in C57BL/6 and IRF-1 KO mice, whereas the Δmfp::kan strain had a lower virulence in these same mouse models. Furthermore, Δmfp::kan and Δomp19::kan strains were used as live vaccines. Challenge experiments revealed that in C57BL/6 and IRF-1 KO mice, the Δmfp::kan strain induced greater protection than the vaccine RB51 and protection similar that of vaccine S19. However, a Δomp19::kan strain induced protection similar to that of RB51. Thus, these results demonstrate that Brucella Mfp and Omp19 are critical for full bacterial virulence and that the Δmfp::kan mutant may serve as a potential vaccine candidate in future studies.
Collapse
Affiliation(s)
- Job Alves de Souza Filho
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Priscila Carneiro Campos
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Juliana Alves-Silva
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Nathalia V Santos
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fernanda Souza de Oliveira
- Departamento de Bioquímica, Universidade Federal de Juiz de Fora Gerais, Governador Valadares, MG, Brazil
| | - Gustavo B Menezes
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vasco Azevedo
- Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Sergio Costa Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
28
|
Abstract
Macrophages are a diverse population of phagocytic cells that reside in tissues throughout the body. At sites of infection, macrophages encounter and engulf invading microbes. Accordingly, macrophages possess specialized effector functions to kill or coordinate the elimination of their prey. Nevertheless, many intracellular bacterial pathogens preferentially replicate inside macrophages. Here we consider explanations for what we call "the macrophage paradox:" why do so many pathogenic bacteria replicate in the very cells equipped to destroy them? We ask whether replication in macrophages is an unavoidable fate that essentially defines a key requirement to be a pathogen. Conversely, we consider whether fundamental aspects of macrophage biology provide unique cellular or metabolic environments that pathogens can exploit. We conclude that resolution of the macrophage paradox requires acknowledgment of the richness and complexity of macrophages as a replicative niche.
Collapse
Affiliation(s)
- Jordan V Price
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Russell E Vance
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
29
|
Bahador A, Hadjati J, Hassannejad N, Ghazanfari H, Maracy M, Jafari S, Nourizadeh M, Nejadeh A. Frequencies of CD4+ T Regulatory Cells and their CD25(high) and FoxP3(high) Subsets Augment in Peripheral Blood of Patients with Acute and Chronic Brucellosis. Osong Public Health Res Perspect 2014; 5:161-8. [PMID: 25180149 PMCID: PMC4147229 DOI: 10.1016/j.phrp.2014.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 04/27/2014] [Accepted: 04/28/2014] [Indexed: 01/18/2023] Open
Abstract
Objectives Brucellosis remains one of the most common zoonotic diseases worldwide. In humans, brucellosis can be a serious, debilitating, and sometimes chronic disease. Different mechanisms can be postulated as to the basis for the induction of the chronic status of infectious diseases that T regulatory cells are one of the most important related mechanisms. The current study was designed to determine whether percentage of CD4+Treg cells and their CD25high and FoxP3high subpopulations in peripheral blood are changed in human brucellosis samples in comparison to a control group. Methods In total, 68 brucellosis patients (acute form: n = 43, chronic form: n = 25) and 36 healthy volunteers entered our study. After isolating of peripheral blood mononuclear cells, heparinized venous blood samples were obtained from both patients and healthy donors, CD4, CD25, and FoxP3 molecules were evaluated by two- and three-color flow cytometric methods. Results The results revealed a new finding in relation to Treg cells and human brucellosis. The numbers of CD4+Treg cells and their CD25high and FoxP3high subsets increase significantly in the peripheral blood of acute and chronic forms of brucellosis samples compared with healthy groups, with this increase being greater in the chronic group. Conclusion There seems to be a correlation between increase of CD4+Treg cells and their subsets and the disease progress from healthy state to acute and chronic brucellosis.
Collapse
Affiliation(s)
- Abbas Bahador
- Department of Microbiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamshid Hadjati
- Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Hassannejad
- Department of Cellular and Molecular Biology, Faculty of Sciences, Science and Research Branch of Islamic Azad University, Tehran, Iran
| | - Hadi Ghazanfari
- Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Maracy
- Department of Biostatistics and Epidemiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sirous Jafari
- Department of Infectious Diseases, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Nourizadeh
- Immunology, Asthma and Allergy Research Institute, Children Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
30
|
Spera JM, Comerci DJ, Ugalde JE. Brucella alters the immune response in a prpA-dependent manner. Microb Pathog 2014; 67-68:8-13. [PMID: 24508400 DOI: 10.1016/j.micpath.2014.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 01/15/2014] [Accepted: 01/27/2014] [Indexed: 01/18/2023]
Abstract
Brucellosis, a disease caused by the gram-negative bacterium Brucella spp., is a widespread zoonosis that inflicts important animal and human health problems, especially in developing countries. One of the hallmarks of Brucella infection is its capacity to establish a chronic infection, characteristic that depends on a wide repertoire of virulence factors among which are immunomodulatory proteins such as PrpA (encoding the proline racemase protein A or hydroxyproline-2-epimerase), involved in the establishment of the chronic phase of the infectious process that we have previously identified and characterized. We report here that, in vivo, Brucella abortus prpA is responsible for an increment in the B-cell number and in the specific antibody response and that these antibodies promote cell infection. We additionally found that Brucella alters the cytokine levels of IFN-γ, IL-10, TGFβ1 and TNFα during the acute phase of the infectious process in a prpA dependent manner.
Collapse
Affiliation(s)
- Juan M Spera
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto Tecnológico de Chascomús, IIB-INTECH, CONICET, Universidad Nacional de San Martín, San Martín, Buenos Aires, Argentina
| | - Diego J Comerci
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto Tecnológico de Chascomús, IIB-INTECH, CONICET, Universidad Nacional de San Martín, San Martín, Buenos Aires, Argentina
| | - Juan E Ugalde
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto Tecnológico de Chascomús, IIB-INTECH, CONICET, Universidad Nacional de San Martín, San Martín, Buenos Aires, Argentina.
| |
Collapse
|
31
|
Jain-Gupta N, Contreras-Rodriguez A, Smith G, Garg V, Witonsky S, Isloor S, Vemulapalli R, Boyle S, Sriranganathan N. Immunotherapeutics to prevent the replication of Brucella in a treatment failure mouse model. Vaccine 2014; 32:918-23. [DOI: 10.1016/j.vaccine.2013.12.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 12/12/2013] [Accepted: 12/18/2013] [Indexed: 01/18/2023]
|
32
|
Brucella melitensis T cell epitope recognition in humans with brucellosis in Peru. Infect Immun 2013; 82:124-31. [PMID: 24126518 DOI: 10.1128/iai.00796-13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Brucella melitensis, one of the causative agents of human brucellosis, causes acute, chronic, and relapsing infection. While T cell immunity in brucellosis has been extensively studied in mice, no recognized human T cell epitopes that might provide new approaches to classifying and prognosticating B. melitensis infection have ever been delineated. Twenty-seven pools of 500 major histocompatibility complex class II (MHC-II) restricted peptides were created by computational prediction of promiscuous MHC-II CD4(+) T cell derived from the top 50 proteins recognized by IgG in human sera on a genome level B. melitensis protein microarray. Gamma interferon (IFN-γ) and interleukin-5 (IL-5) enzyme-linked immunospot (ELISPOT) analyses were used to quantify and compare Th1 and Th2 responses of leukapheresis-obtained peripheral blood mononuclear cells from Peruvian subjects cured after acute infection (n = 9) and from patients who relapsed (n = 5). Four peptide epitopes derived from 3 B. melitensis proteins (BMEI 1330, a DegP/HtrA protease; BMEII 0029, type IV secretion system component VirB5; and BMEII 0691, a predicted periplasmic binding protein of a peptide transport system) were found repeatedly to produce significant IFN-γ ELISPOT responses in both acute-infection and relapsing patients; none of the peptides distinguished the patient groups. IL-5 responses against the panel of peptides were insignificant. These experiments are the first to systematically identify B. melitensis MHC-II-restricted CD4(+) T cell epitopes recognized by the human immune response, with the potential for new approaches to brucellosis diagnostics and understanding the immunopathogenesis related to this intracellular pathogen.
Collapse
|
33
|
Feldman KE, Loriaux PM, Saito M, Tuero I, Villaverde H, Siva T, Gotuzzo E, Gilman RH, Hoffmann A, Vinetz JM. Ex vivo innate immune cytokine signature of enhanced risk of relapsing brucellosis. PLoS Negl Trop Dis 2013; 7:e2424. [PMID: 24040434 PMCID: PMC3764229 DOI: 10.1371/journal.pntd.0002424] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 07/30/2013] [Indexed: 01/18/2023] Open
Abstract
Background Brucellosis, a zoonotic infection caused by one of the Gram-negative intracellular bacteria of the Brucella genus, is an ongoing public health problem in Perú. While most patients who receive standard antibiotic treatment recover, 5–40% suffer a brucellosis relapse. In this study, we examined the ex vivo immune cytokine profiles of recovered patients with a history of acute and relapsing brucellosis. Methodology/Principal Findings Blood was taken from healthy control donors, patients with a history of acute brucellosis, or patients with a history of relapsing brucellosis. Peripheral blood mononuclear cells were isolated and remained in culture without stimulation or were stimulated with a panel of toll-like receptor agonists or heat-killed Brucella melitensis (HKBM) isolates. Innate immune cytokine gene expression and protein secretion were measured by quantitative real-time polymerase chain reaction and a multiplex bead-based immunoassay, respectively. Acute and relapse patients demonstrated consistently elevated cytokine gene expression and secretion levels compared to controls. Notably, these include: basal and stimulus-induced expression of GM-CSF, TNF-α, and IFN-γ in response to LPS and HKBM; basal secretion of IL-6, IL-8, and TNF-α; and HKBM or Rev1-induced secretion of IL-1β, IL-2, GM-CSF, IFN-Υ, and TNF-α. Although acute and relapse patients were largely indistinguishable by their cytokine gene expression profiles, we identified a robust cytokine secretion signature that accurately discriminates acute from relapse patients. This signature consists of basal IL-6 secretion, IL-1β, IL-2, and TNF-α secretion in response to LPS and HKBM, and IFN-γ secretion in response to HKBM. Conclusions/Significance This work demonstrates that informative cytokine variations in brucellosis patients can be detected using an ex vivo assay system and used to identify patients with differing infection histories. Targeted diagnosis of this signature may allow for better follow-up care of brucellosis patients through improved identification of patients at risk for relapse. Brucellosis is a disease caused by transmission of bacteria of the Brucella genus from infected animals to humans. The main route of infection occurs through consumption of contaminated dairy products or contact with infected animals. While most patients treated with antibiotics will be cured of the infection, between 5–40% of patients experience a relapse of brucellosis. The mechanisms underlying these recurring infections remain poorly understood. In this study, we examined blood cells from control donors, patients who previously had acute infections, and patients who previously had relapsing infections. We identified an inflammatory cytokine signature from measurements of unstimulated and stimulated cells that showed statistically significant differences between relapsing and non-relapsing brucellosis patients. Future applications of this assay system may allow for better follow-up care of brucellosis through the diagnosis of this cytokine signature and predictive or improved identification of patients at risk for relapse.
Collapse
Affiliation(s)
- Kristyn E. Feldman
- Signaling Systems Laboratory, University of California San Diego, La Jolla, California, United States of America
| | - Paul M. Loriaux
- Signaling Systems Laboratory, University of California San Diego, La Jolla, California, United States of America
| | - Mayuko Saito
- Division of Infectious Diseases, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Iskra Tuero
- Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Homarh Villaverde
- Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Tenaya Siva
- Signaling Systems Laboratory, University of California San Diego, La Jolla, California, United States of America
| | - Eduardo Gotuzzo
- Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Perú
- Departmento de Enfermedades Infecciosas y Tropicales, Hospital Nacional Cayetano Heredia, Lima, Perú
| | - Robert H. Gilman
- Laboratorio de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Alexander Hoffmann
- Signaling Systems Laboratory, University of California San Diego, La Jolla, California, United States of America
- * E-mail: (AH); (JMV)
| | - Joseph M. Vinetz
- Division of Infectious Diseases, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
- Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Perú
- Laboratorio de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
- * E-mail: (AH); (JMV)
| |
Collapse
|
34
|
Xavier MN, Winter MG, Spees AM, den Hartigh AB, Nguyen K, Roux CM, Silva TMA, Atluri VL, Kerrinnes T, Keestra AM, Monack DM, Luciw PA, Eigenheer RA, Bäumler AJ, Santos RL, Tsolis RM. PPARγ-mediated increase in glucose availability sustains chronic Brucella abortus infection in alternatively activated macrophages. Cell Host Microbe 2013; 14:159-70. [PMID: 23954155 PMCID: PMC3777723 DOI: 10.1016/j.chom.2013.07.009] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 05/17/2013] [Accepted: 06/20/2013] [Indexed: 01/16/2023]
Abstract
Eradication of persistent intracellular bacterial pathogens with antibiotic therapy is often slow or incomplete. However, strategies to augment antibiotics are hampered by our poor understanding of the nutritional environment that sustains chronic infection. Here we show that the intracellular pathogen Brucella abortus survives and replicates preferentially in alternatively activated macrophages (AAMs), which are more abundant during chronic infection. A metabolic shift induced by peroxisome proliferator-activated receptor γ (PPARγ), which increases intracellular glucose availability, is identified as a causal mechanism promoting enhanced bacterial survival in AAMs. Glucose uptake was crucial for increased replication of B. abortus in AAMs, and for chronic infection, as inactivation of the bacterial glucose transporter gluP reduced both intracellular survival in AAMs and persistence in mice. Thus, a shift in intracellular nutrient availability induced by PPARγ promotes chronic persistence of B. abortus within AAMs, and targeting this pathway may aid in eradicating chronic infection.
Collapse
Affiliation(s)
- Mariana N. Xavier
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Maria G. Winter
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Alanna M. Spees
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Andreas B. den Hartigh
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Kim Nguyen
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Christelle M. Roux
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Teane M. A. Silva
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Vidya L. Atluri
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Tobias Kerrinnes
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - A. Marijke Keestra
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Denise M. Monack
- Department of Microbiology & Immunology, School of Medicine, Stanford University, Palo Alto, CA, 94305, USA
| | - Paul A. Luciw
- Center for Comparative Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Richard A. Eigenheer
- Proteomics Core Facility, University of California at Davis Genome Center, Davis, CA, 95616, USA
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Renato L. Santos
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Renée M. Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, 95616, USA
| |
Collapse
|
35
|
Gomez G, Adams LG, Rice-Ficht A, Ficht TA. Host-Brucella interactions and the Brucella genome as tools for subunit antigen discovery and immunization against brucellosis. Front Cell Infect Microbiol 2013; 3:17. [PMID: 23720712 PMCID: PMC3655278 DOI: 10.3389/fcimb.2013.00017] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 04/26/2013] [Indexed: 01/18/2023] Open
Abstract
Vaccination is the most important approach to counteract infectious diseases. Thus, the development of new and improved vaccines for existing, emerging, and re-emerging diseases is an area of great interest to the scientific community and general public. Traditional approaches to subunit antigen discovery and vaccine development lack consideration for the critical aspects of public safety and activation of relevant protective host immunity. The availability of genomic sequences for pathogenic Brucella spp. and their hosts have led to development of systems-wide analytical tools that have provided a better understanding of host and pathogen physiology while also beginning to unravel the intricacies at the host-pathogen interface. Advances in pathogen biology, host immunology, and host-agent interactions have the potential to serve as a platform for the design and implementation of better-targeted antigen discovery approaches. With emphasis on Brucella spp., we probe the biological aspects of host and pathogen that merit consideration in the targeted design of subunit antigen discovery and vaccine development.
Collapse
Affiliation(s)
- Gabriel Gomez
- Department of Veterinary Pathobiology, Texas A&M University College Station, TX 77843, USA.
| | | | | | | |
Collapse
|
36
|
Jain S, Afley P, Kumar S. Immunological responses to recombinant cysteine synthase A of Brucella abortus in BALB/c mice. World J Microbiol Biotechnol 2013; 29:907-13. [PMID: 23269507 DOI: 10.1007/s11274-012-1247-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 12/20/2012] [Indexed: 10/27/2022]
Abstract
Brucellosis is a worldwide zoonotic disease. No Brucella vaccine is available for use in humans, and existing animal vaccines have limitations. There is a need to develop a safe and effective vaccine against human and animal brucellosis. In the present study, we generated recombinant cysteine synthase A (rCysK) of Brucella abortus in Escherichia coli and purified it up to homogeneity by metal affinity chromatography. The immunogenicity and protective efficacy of purified rCysK were evaluated in BALB/c mice with Freund's adjuvant, aluminium hydroxide gel or without any adjuvant. High titres of anti-rCysK IgG antibody predominated by IgG1 were observed in all immunized mice. After stimulation with rCysK, the spleen lymphocytes of mice immunized with CysK formulated with aluminium hydroxide gel produced significant levels of IFN-γ. Protection against challenge with virulent B. abortus strain 544 was determined in BALB/c mice, and significant protection was observed in all CysK immunized groups when compared with PBS control. Among all the CysK vaccine groups, comparatively better protection was observed in mice immunized with aluminium hydroxide gel (1.064 units of protection). Overall, the results of the study suggest that rCysK induces primarily Th2 type of immune response and provides partial protection against B. abortus challenge.
Collapse
Affiliation(s)
- Shikha Jain
- Division of Microbiology, Defence Research and Development Establishment, Jhansi Road, Gwalior, 474 002 MP, India
| | | | | |
Collapse
|
37
|
YANG X, SKYBERG JA, CAO L, CLAPP B, THORNBURG T, PASCUAL DW. Progress in Brucella vaccine development. FRONTIERS IN BIOLOGY 2013; 8:60-77. [PMID: 23730309 PMCID: PMC3666581 DOI: 10.1007/s11515-012-1196-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 01/16/2012] [Indexed: 01/18/2023]
Abstract
Brucella spp. are zoonotic, facultative intracellular pathogens, which cause animal and human disease. Animal disease results in abortion of fetuses; in humans, it manifests flu-like symptoms with an undulant fever, with osteoarthritis as a common complication of infection. Antibiotic regimens for human brucellosis patients may last several months and are not always completely effective. While there are no vaccines for humans, several licensed live Brucella vaccines are available for use in livestock. The performance of these animal vaccines is dependent upon the host species, dose, and route of immunization. Newly engineered live vaccines, lacking well-defined virulence factors, retain low residual virulence, are highly protective, and may someday replace currently used animal vaccines. These also have possible human applications. Moreover, due to their enhanced safety and efficacy in animal models, subunit vaccines for brucellosis show great promise for their application in livestock and humans. This review summarizes the progress of brucellosis vaccine development and presents an overview of candidate vaccines.
Collapse
Affiliation(s)
- Xinghong YANG
- Department of Immunology & Infectious Diseases, Montana State University, Bozeman, MT 59717-3610, USA
| | - Jerod A. SKYBERG
- Department of Immunology & Infectious Diseases, Montana State University, Bozeman, MT 59717-3610, USA
| | - Ling CAO
- Department of Immunology & Infectious Diseases, Montana State University, Bozeman, MT 59717-3610, USA
| | - Beata CLAPP
- Department of Immunology & Infectious Diseases, Montana State University, Bozeman, MT 59717-3610, USA
| | - Theresa THORNBURG
- Department of Immunology & Infectious Diseases, Montana State University, Bozeman, MT 59717-3610, USA
| | - David W. PASCUAL
- Department of Immunology & Infectious Diseases, Montana State University, Bozeman, MT 59717-3610, USA
| |
Collapse
|
38
|
Akhvlediani T, Chitadze N, Laws TR, Makharadze M, Chubinidze M, Tsanava S, Commander NJ, Perkins SD, Dyson EH, Rivard RG, Hepburn MJ, Simpson AJH, Imnadze P, Trapaidze N. Comparison of total antibody and interferon-γ T-cell responses in patients following infection with brucellosis in Georgia. J Infect Public Health 2012; 5:321-31. [PMID: 23164560 DOI: 10.1016/j.jiph.2012.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 02/13/2012] [Accepted: 03/22/2012] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is an ancient disease that still remains a significant threat to humans and is typically linked to exposure to infected animals and/or consumption of unpasteurized animal products. Despite this history, we have a relatively limited understanding of the host characteristics of this disease; consequently, further research is necessary. In this study, we examined the humoral immune response in 43 Georgian individuals that had been diagnosed with brucellosis 3-12 months before enrollment in the study, many of whom still had symptoms after the completion of antibiotic therapy. In total, 35 of 43 (83%) of the patients had antibodies that bound to Brucella lipopolysaccharide (LPS) by COMPELISA, and 34 of 38 (89%) patients had demonstrable specific antibodies to Brucellergene™ antigens; the results from the two ELISAs were highly correlated (p=0.031, r=0.851). We also studied the cellular immune responses in 15 patients. All of the patients generated interferon (IFN)-γ in response to ex vivo stimulation with Brucella protein antigens, and the majority of the patients maintained measurable humoral responses to both LPS and protein antigens. From this initial study, we conclude that measurement of antibody and of cellular (IFN-γ) responses to brucellergene OCB protein epitopes may be worthy of further investigation as an alternative or adjunct to current diagnostics.
Collapse
Affiliation(s)
- T Akhvlediani
- Clinical Research Unit-CRU, Technology Management Company-TMC, Tbilisi, Georgia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Pei J, Ding X, Fan Y, Rice-Ficht A, Ficht TA. Toll-like receptors are critical for clearance of Brucella and play different roles in development of adaptive immunity following aerosol challenge in mice. Front Cell Infect Microbiol 2012; 2:115. [PMID: 22973560 PMCID: PMC3435510 DOI: 10.3389/fcimb.2012.00115] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 08/15/2012] [Indexed: 01/18/2023] Open
Abstract
Brucella spp. cause undulant fever in humans and brucellosis in variety of other animals. Both innate and adaptive immunity have been shown to be important in controlling Brucella infection. Toll-like receptors (TLRs) represent a group of pattern recognition receptors (PRRs) that play critical roles in the host innate immune response, as well as development of adaptive immunity. In the current report, we investigated the role of TLR signaling in the clearance of Brucella and development of adaptive immunity in TLR2(-/-), TLR4(-/-), or MyD88(-/-) mice following aerosol exposure to B. melitensis 16 M. Consistent with previous reports, MyD88 is required for efficient clearance of Brucella from all three organs (lung, spleen, and liver). The results reveal Th2-skewed immune responses in TLR2(-/-) mice late in infection and support a TLR2 requirement for efficient clearance of Brucella from the lungs, but not from the spleen or liver. Similarly, TLR4 is required for efficient clearance of Brucella from the lung, but exhibits a minor contribution to clearance from the spleen and no demonstrable contribution to clearance from the liver. Lymphocyte proliferation assays suggest that the TLRs are not involved in the development of cell-mediated memory response to Brucella antigen. Antibody detection reveals that TLR2 and 4 are required to generate early antigen-specific IgG, but not during the late stages of infection. TLR2 and 4 are only transiently required for IgM production and not at all for IgA production. In contrast, MyD88 is essential for antigen specific IgG production late in infection, but is not required for IgM generation over the course of infection. Surprisingly, despite the prominent role for MyD88 in clearance from all tissues, MyD88-knockout mice express significantly higher levels of serum IgA. These results confirm the important role of MyD88 in controlling infection in the spleen while providing evidence of a prominent contribution to protection in other tissues. In addition, although TLR4 and TLR2 contribute little to control of spleen infection, a significant contribution to clearance of lung infection is described.
Collapse
Affiliation(s)
- Jianwu Pei
- Department of Veterinary Pathobiology, Texas A&M University, College Station TX, USA.
| | | | | | | | | |
Collapse
|
40
|
Antunes JMADP, Allendorf SD, Appolinário CM, Cagnini DQ, Figueiredo PR, Júnior JB, Baños JV, Kocan KM, de la Fuente J, Megid J. Rough virulent strain of Brucella ovis induces pro- and anti-inflammatory cytokines in reproductive tissues in experimentally infected rams. Vet Microbiol 2012; 161:339-43. [PMID: 23041076 DOI: 10.1016/j.vetmic.2012.07.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 07/14/2012] [Accepted: 07/16/2012] [Indexed: 01/18/2023]
Abstract
The ovine brucellosis caused by Brucella ovis has tropism for reproductive tissues but until now the mechanism of bacterial persistence is not understood. Cytokine expression profiles were studied for 8 months in rams after being experimentally infected with the rough virulent strain of B. ovis (R-B. ovis) to study the pathogenesis of B. ovis and immune mechanism possibly associated to bacteria tropism and persistence. The messenger RNA (mRNA) expression levels of interleukin-1α (IL-1α), IL-1β, IL-6, IL-10, IL-12, interferon-γ (INF-γ) and tumour necrosis factor-α (TNF-α) cytokines were quantified by real-time quantitative RT-PCR (qRT-PCR) in reproductive tissues (epididymus, testicles, ampolae, vesicular glands and bulbourethral glands), and non-reproductive (liver, spleen and kidneys) tissues at 30, 60, 120 and 240 days post infection (dpi). During the acute phase of infection at 30 dpi, the host immune response was most notable demonstrating an up-regulation of several cytokines in reproductive tissues, including the epididymus (IL-6, IL-1β and IL-1α), testicles (INF-γ and IL-12), bulbourethral glands (IL-6 and TNF-α) and ampolae (INF-γ, IL-10, IL-1β and IL-1α). During the development of infection, cytokine gene expression levels decreased, providing evidence of immunosuppression and evidence of immune evasion that favoured persistence of chronic R-B. ovis infection. During the chronic phase of R-B. ovis infection (120 and 240 dpi), cytokine production was down-regulated in the epididymus (IL-1β and IL-1α), testicles (INF-γ and IL-12), and ampolae (INF-γ, IL-10, IL-1β and IL-1α), with the exception of the bulbourethral glands (IL-6 and TNF-α) and epididymus (IL-6); in these tissues, R-B. ovis infection resulted in up-regulation of the pro-inflammatory cytokine IL-6. Herein, we report cytokine expression profiles in tissues of rams experimentally infected with the rough strain of B. ovis, which are associated with bacterial persistence and macrophage activation.
Collapse
|
41
|
Velásquez LN, Delpino MV, Ibañez AE, Coria LM, Miraglia MC, Scian R, Cassataro J, Giambartolomei GH, Barrionuevo P. Brucella abortus induces apoptosis of human T lymphocytes. Microbes Infect 2012; 14:639-650. [PMID: 22387699 DOI: 10.1016/j.micinf.2012.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/16/2012] [Accepted: 02/15/2012] [Indexed: 01/18/2023]
Abstract
Immune evasion is essential for Brucella abortus to survive in the face of robust adaptive CD4+ T cell response. We have previously demonstrated that B. abortus can indirectly inhibit CD4+ T cells by down-regulating MHC-II expression and antigen presentation on macrophages. However, whether B. abortus is able to directly interfere with T lymphocytes is not known. We report here that B. abortus induces apoptosis of human T lymphocytes, even though invasion of T lymphocytes was low and non-replicative. The ability of heat-killed B. abortus to reproduce the same phenomenon suggested that there was a bacterial structural component involved. We demonstrated that a prototypical B. abortus outer membrane lipoprotein (l-Omp19), but not its unlipidated form, induced T lymphocyte apoptosis. Moreover, a synthetic lipohexapeptide that mimics the structure of the protein lipid moiety also induced an increase in T lymphocyte cell death, indicating that the structural component implicated in the phenomenon could be any B. abortus lipoprotein. B. abortus-induced T lymphocyte apoptosis was dependent on the secretion of TNF-α since pre-incubation of T lymphocytes with anti-TNF-α mAb inhibited the apoptosis of the cells. Overall, these results represent a new mechanism whereby B. abortus by directly inhibiting T cell-mediated responses may evade adaptive immune responses.
Collapse
Affiliation(s)
- Lis N Velásquez
- Instituto de Estudios de la Inmunidad Humoral, CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Skyberg JA, Thornburg T, Kochetkova I, Layton W, Callis G, Rollins MF, Riccardi C, Becker T, Golden S, Pascual DW. IFN-γ-deficient mice develop IL-1-dependent cutaneous and musculoskeletal inflammation during experimental brucellosis. J Leukoc Biol 2012; 92:375-87. [PMID: 22636321 DOI: 10.1189/jlb.1211626] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Human brucellosis exhibits diverse pathological manifestations that can affect almost any organ. In particular, osteoarticular complications are the most common focal manifestation of brucellosis and occur in 40-80% of patients. In immunocompetent mice, Brucella replication is generally restricted to the spleen, liver, and to a lesser extent, LNs, thereby limiting their use for study of focal inflammation often found in brucellosis. Here, we report that nasal, oral, or peritoneal infection of IFN-γ(-/-) mice with WT Brucella melitensis or Brucella abortus results in joint and periarticular tissue inflammation. Histological analysis of the affected joints revealed inflammatory infiltrates and debris within the joint space colocalizing with Brucella antigen. Osteoarthritis, necrosis, periarticular soft tissue inflammation, and substantial brucellae burdens were observed. Oral rifampicin was effective in clearing infection and halting further progression of focal inflammation from infected IFN-γ(-/-) mice, although some symptoms and swelling remained. Elevated IL-1 β, but not TNF-α, IL-6, or IL-17, was detected in joint homogenates from infected IFN-γ(-/-) mice. Whereas more susceptible to systemic infection, IL-1R(-/-) mice depleted of IFN-γ were more resistant to focal inflammation than WT mice similarly depleted of IFN-γ. Collectively, these results show IFN-γ(-/-) mice represent a potential model for study of focal inflammation attributed to Brucella infection and will allow evaluation of intervention strategies targeting IL-1, IL-1R, or other inflammatory mediators, with the potential to complement antibiotic-based therapies.
Collapse
Affiliation(s)
- Jerod A Skyberg
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Gupta VK, Radhakrishnan G, Harms J, Splitter G. Invasive Escherichia coli vaccines expressing Brucella melitensis outer membrane proteins 31 or 16 or periplasmic protein BP26 confer protection in mice challenged with B. melitensis. Vaccine 2012; 30:4017-22. [PMID: 22546330 DOI: 10.1016/j.vaccine.2012.04.036] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 04/05/2012] [Accepted: 04/09/2012] [Indexed: 11/30/2022]
Abstract
Because of the serious economic and medical consequences of brucellosis, efforts are to prevent infection of domestic animals through vaccines. Many disadvantages are associated with the current Brucella melitensis Rev.1 vaccine prompting development of alternative vaccines and delivery. Escherichia coli (DH5α) was engineered to express a plasmid containing the inv gene from Yersinia pseudotuberculosis and the hly gene from Listeria monocytogenes. These recombinant invasive E. coli expressing B. melitensis outer membrane proteins (Omp31 or 16) or the periplasmic protein BP26 were evaluated for protection of mice against virulent B. melitensis. Importantly, these invasive E. coli vaccines induced significant protection against B. melitensis challenged mice. Invasive E. coli may be an ideal vaccine platform with natural adjuvant properties for application against B. melitensis since the E. coli delivery system is non-pathogenic and can deliver antigens to antigen-presenting cells promoting cellular immune responses.
Collapse
Affiliation(s)
- V K Gupta
- Central Institute for Research On Goats, Makhdoom, PO Farah, Mathura 281 122, India.
| | | | | | | |
Collapse
|
44
|
Active evasion of CTL mediated killing and low quality responding CD8+ T cells contribute to persistence of brucellosis. PLoS One 2012; 7:e34925. [PMID: 22558103 PMCID: PMC3338818 DOI: 10.1371/journal.pone.0034925] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/09/2012] [Indexed: 02/02/2023] Open
Abstract
Brucellosis is a common zoonotic disease that remains endemic in many parts of the world. Dissecting the host immune response during this disease provides insight as to why brucellosis is often difficult to resolve. We used a Brucella epitope specific in vivo killing assay to investigate the ability of CD8+ T cells to kill targets treated with purified pathogenic protein. Importantly, we found the pathogenic protein TcpB to be a novel effector of adaptive immune evasion by inhibiting CD8+ T cell killing of Brucella epitope specific target cells in mice. Further, BALB/c mice show active Brucella melitensis infection beyond one year, many with previously unreported focal infection of the urogenital area. A fraction of CD8+ T cells show a CD8+ Tmem phenotype of LFA-1hi, CD127hi, KLRG-1lo during the course of chronic brucellosis, while the CD8+ T cell pool as a whole had a very weak polyfunctional cytokine response with diminished co-expression of IFN-γ with TNFα and/or IL-2, a hallmark of exhaustion. When investigating the expression of these 3 cytokines individually, we observed significant IFN-γ expression at 90 and 180 days post-infection. TNFα expression did not significantly exceed or fall below background levels at any time. IL-2 expression did not significantly exceeded background, but, interestingly, did fall significantly below that of uninfected mice at 180 days post-infection. Brucella melitensis evades and blunts adaptive immunity during acute infection and our findings provide potential mechanisms for the deficit observed in responding CD8+ T cells during chronic brucellosis.
Collapse
|
45
|
Cannella AP, Tsolis RM, Liang L, Felgner PL, Saito M, Sette A, Gotuzzo E, Vinetz JM. Antigen-specific acquired immunity in human brucellosis: implications for diagnosis, prognosis, and vaccine development. Front Cell Infect Microbiol 2012; 2:1. [PMID: 22919593 PMCID: PMC3417515 DOI: 10.3389/fcimb.2012.00001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 01/11/2012] [Indexed: 11/13/2022] Open
Abstract
Brucella spp., are Gram negative bacteria that cause disease by growing within monocyte/macrophage lineage cells. Clinical manifestations of brucellosis are immune mediated, not due to bacterial virulence factors. Acquired immunity to brucellosis has been studied through observations of naturally infected hosts (cattle, goats), mouse models (mice), and human infection. Even though Brucella spp. are known for producing mechanisms that evade the immune system, cell-mediated immune responses drive the clinical manifestations of human disease after exposure to Brucella species, as high antibody responses are not associated with protective immunity. The precise mechanisms by which cell-mediated immune responses confer protection or lead to disease manifestations remain undefined. Descriptive studies of immune responses in human brucellosis show that TH1 (interferon-γ-producing T cells) are associated with dominant immune responses, findings consistent with animal studies. Whether these T cell responses are protective, or determine the different clinical responses associated with brucellosis is unknown, especially with regard to undulant fever manifestations, relapsing disease, or are associated with responses to distinct sets of Brucella spp. antigens are unknown. Few data regarding T cell responses in terms of specific recognition of Brucella spp. protein antigens and peptidic epitopes, either by CD4+ or CD8+ T cells, have been identified in human brucellosis patients. Additionally because current attenuated Brucella vaccines used in animals cause human disease, there is a true need for a recombinant protein subunit vaccine for human brucellosis, as well as for improved diagnostics in terms of prognosis and identification of unusual forms of brucellosis. This review will focus on current understandings of antigen-specific immune responses induced Brucella peptidic epitopes that has promise for yielding new insights into vaccine and diagnostics development, and for understanding pathogenetic mechanisms of human brucellosis.
Collapse
Affiliation(s)
- Anthony P Cannella
- Division of Infectious Diseases, Department of Medicine, University of California San Diego La Jolla, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Atluri VL, Xavier MN, de Jong MF, den Hartigh AB, Tsolis RM. Interactions of the human pathogenic Brucella species with their hosts. Annu Rev Microbiol 2012; 65:523-41. [PMID: 21939378 DOI: 10.1146/annurev-micro-090110-102905] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Brucellosis is a zoonotic infection caused primarily by the bacterial pathogens Brucella melitensis and B. abortus. It is acquired by consumption of unpasteurized dairy products or by contact with infected animals. Globally, it is one of the most widespread zoonoses, with 500,000 new cases reported each year. In endemic areas, Brucella infections represent a serious public health problem that results in significant morbidity and economic losses. An important feature of the disease is persistent bacterial colonization of the reticuloendothelial system. In this review we discuss recent insights into mechanisms of intracellular survival and immune evasion that contribute to systemic persistence by the pathogenic Brucella species.
Collapse
Affiliation(s)
- Vidya L Atluri
- Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616, USA.
| | | | | | | | | |
Collapse
|
47
|
Protective live oral brucellosis vaccines stimulate Th1 and th17 cell responses. Infect Immun 2011; 79:4165-74. [PMID: 21768283 DOI: 10.1128/iai.05080-11] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Zoonotic transmission of brucellosis often results from exposure to Brucella-infected livestock, feral animals, or wildlife or frequently via consumption of unpasteurized milk products or raw meat. Since natural infection of humans often occurs by the oral route, mucosal vaccination may offer a means to confer protection for both mucosal and systemic tissues. Significant efforts have focused on developing a live brucellosis vaccine, and deletion of the znuA gene involved in zinc transport has been found to attenuate Brucella abortus. A similar mutation has been adapted for Brucella melitensis and tested to determine whether oral administration of ΔznuA B. melitensis can confer protection against nasal B. melitensis challenge. A single oral vaccination with ΔznuA B. melitensis rapidly cleared from mice within 2 weeks and effectively protected mice upon nasal challenge with wild-type B. melitensis 16M. In 83% of the vaccinated mice, no detectable brucellae were found in their spleens, unlike with phosphate-buffered saline (PBS)-dosed mice, and vaccination also enhanced the clearance of brucellae from the lungs. Moreover, vaccinated gamma interferon-deficient (IFN-γ(-/-)) mice also showed protection in both spleens and lungs, albeit protection that was not as effective as in immunocompetent mice. Although IFN-γ, interleukin 17 (IL-17), and IL-22 were stimulated by these live vaccines, only RB51-mediated protection was codependent upon IL-17 in BALB/c mice. These data suggest that oral immunization with the live, attenuated ΔznuA B. melitensis vaccine provides an attractive strategy to protect against inhalational infection with virulent B. melitensis.
Collapse
|
48
|
Martirosyan A, Moreno E, Gorvel JP. An evolutionary strategy for a stealthy intracellular Brucella pathogen. Immunol Rev 2011; 240:211-34. [PMID: 21349096 DOI: 10.1111/j.1600-065x.2010.00982.x] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Brucella is an intracellular bacterial pathogen that causes abortion and infertility in mammals and leads to a debilitating febrile illness that can progress into a long lasting disease with severe complications in humans. Its virulence depends on survival and replication properties in host cells. In this review, we describe the stealthy strategy used by Brucella to escape recognition of the innate immunity and the means by which this bacterium evades intracellular destruction. We also discuss the development of adaptive immunity and its modulation during brucellosis that in course leads to chronic infections. Brucella has developed specific strategies to influence antigen presentation mediated by cells. There is increasing evidence that Brucella also modulates signaling events during host adaptive immune responses.
Collapse
Affiliation(s)
- Anna Martirosyan
- Faculté de Sciences de Luminy, Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Marseille, France
| | | | | |
Collapse
|
49
|
Laboratory animal models for brucellosis research. J Biomed Biotechnol 2011; 2011:518323. [PMID: 21403904 PMCID: PMC3043301 DOI: 10.1155/2011/518323] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 11/25/2010] [Accepted: 01/11/2011] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is a chronic infectious disease caused by Brucella spp., a Gram-negative facultative intracellular pathogen that affects humans and animals, leading to significant impact on public health and animal industry. Human brucellosis is considered the most prevalent bacterial zoonosis in the world and is characterized by fever, weight loss, depression, hepato/splenomegaly, osteoarticular, and genital infections. Relevant aspects of Brucella pathogenesis have been intensively investigated in culture cells and animal models. The mouse is the animal model more commonly used to study chronic infection caused by Brucella. This model is most frequently used to investigate specific pathogenic factors of Brucella spp., to characterize the host immune response, and to evaluate therapeutics and vaccines. Other animal species have been used as models for brucellosis including rats, guinea pigs, and monkeys. This paper discusses the murine and other laboratory animal models for human and animal brucellosis.
Collapse
|
50
|
Skendros P, Pappas G, Boura P. Cell-mediated immunity in human brucellosis. Microbes Infect 2010; 13:134-42. [PMID: 21034846 DOI: 10.1016/j.micinf.2010.10.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 10/13/2010] [Accepted: 10/15/2010] [Indexed: 01/18/2023]
Abstract
Brucella can parasitize within human antigen-presenting cells modifying phagocytosis, phagolysosome fusion, antigen presentation, cytokine secretion, and apoptosis. Subversion of innate immune mechanisms by Brucella leads to defective Th1 immune responses and T-cell anergy in chronic brucellosis patients. This review summarizes the cellular immune responses in brucellosis, based on data derived exclusively from human cells or cell lines.
Collapse
Affiliation(s)
- Panagiotis Skendros
- First Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece.
| | | | | |
Collapse
|