1
|
Li J, Yuan N, Zhai Y, Wang M, Hao M, Liu X, Zhou D, Liu W, Jin Y, Wang A. Protein disulfide isomerase A4 binds to Brucella BtpB and mediates intracellular NAD +/NADH metabolism in RAW264.7 cells. Int Immunopharmacol 2024; 142:113046. [PMID: 39226825 DOI: 10.1016/j.intimp.2024.113046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
The Toll/interleukin-1 receptor (TIR) signaling domain is distributed widely in mammalian Toll-like receptors and adaptors, plant nucleotide-binding leucine-rich repeat receptors, and specific bacterial virulence proteins. Proteins that possess TIR domain exhibit NADase activity which is distinct from the canonical signaling function of these domains. However, the effects of bacterial TIR domain proteins on host metabolic switches and the underlying mechanism of NADase activity in these proteins remain unclear. Here, we utilized Brucella TIR domain-containing type IV secretion system effector protein, BtpB, to explore the mechanism of NADase activity in host cells. We showed that using ectopic expression BtpB not only generates depletion of NAD+ but also loss of NADH and ATP in RAW264.7 macrophage cells. Moreover, immunoprecipitation-mass spectrometry, co-immunoprecipitation, and confocal microscope assays revealed that BtpB interacted with host protein disulfide isomerase A4 (PDIA4). The Brucella mutant strain deleted the gene for BtpB, significantly decreased PDIA4 expression. Furthermore, our data revealed that PDIA4 played an important role in regulating intracellular NAD+/NADH levels in macrophages, and PDIA4 overexpression restored the decline of intracellular NAD+ and NADH levels induced by Brucella BtpB. The results provide new insights into the metabolic regulatory activity of TIR domain proteins in the critical human and animal pathogen Brucella.
Collapse
Affiliation(s)
- Junmei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Department of Veterinary Medicine, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Ningqiu Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yunyi Zhai
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Minghui Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Mingyue Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Xiaofang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Dong Zhou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Wei Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, China; Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
2
|
Tian T, Zhu Y, Shi J, Shang K, Yin Z, Shi H, He Y, Ding J, Zhang F. The development of a human Brucella mucosal vaccine: What should be considered? Life Sci 2024; 355:122986. [PMID: 39151885 DOI: 10.1016/j.lfs.2024.122986] [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: 07/09/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Brucellosis is a chronic infectious disease that is zoonotic in nature. Brucella can infect humans through interactions with livestock, primarily via the digestive tract, respiratory tract, and oral cavity. This bacterium has the potential to be utilized as a biological weapon and is classified as a Category B pathogen by the Centers for Disease Control and Prevention. Currently, there is no approved vaccine for humans against Brucella, highlighting an urgent need for the development of a vaccine to mitigate the risks posed by this pathogen. Brucella primarily infects its host by adhering to and penetrating mucosal surfaces. Mucosal immunity plays a vital role in preventing local infections, clearing microorganisms from mucosal surfaces, and inhibiting the spread of pathogens. As mucosal vaccine strategies continue to evolve, the development of a safe and effective mucosal vaccine against Brucella appears promising.This paper reviews the immune mechanism of mucosal vaccines, the infection mechanism of Brucella, successful Brucella mucosal vaccines in animals, and mucosal adjuvants. Additionally, it elucidates targeting and optimization strategies for mucosal vaccines to facilitate the development of human vaccines against Brucella.
Collapse
Affiliation(s)
- Tingting Tian
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Yuejie Zhu
- Reproductive Fertility Assistance Center, First Afffliated Hospital of Xinjiang Medical University, China
| | - Juan Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Kaiyu Shang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Zhengwei Yin
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Huidong Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Yueyue He
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Jianbing Ding
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Fengbo Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China; Department of Clinical laboratory, The First Affiliated hospital of Xinjiang Medical University, China.
| |
Collapse
|
3
|
Alonso Paiva IM, A. Santos R, Brito CB, Ferrero MC, Ortiz Wilczyñski JM, Silva EAC, C. Oliveira S, Baldi PC. Role of the cGAS/STING pathway in the control of Brucella abortus infection acquired through the respiratory route. Front Immunol 2023; 14:1116811. [PMID: 37261352 PMCID: PMC10227575 DOI: 10.3389/fimmu.2023.1116811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/28/2023] [Indexed: 06/02/2023] Open
Abstract
Despite the importance of the respiratory route for Brucella transmission, the lung immune response to this pathogen is scarcely characterized. We investigated the role of the cGAS/STING pathway of microbial DNA recognition in the control of respiratory Brucella infection. After in vitro B. abortus infection, CFU numbers were significantly higher in alveolar macrophages (AM) and lung explants from STING KO mice than in samples from wild type (WT) mice, but no difference was observed for cGAS KO samples. CFU were also increased in WT AM and lung epithelial cells preincubated with the STING inhibitor H151. Several proinflammatory cytokines (TNF-α, IL-1β, IL-6, IP-10/CXCL10) were diminished in Brucella-infected lung explants and/or AM from STING KO mice and cGAS KO mice. These cytokines were also reduced in infected AM and lung epithelial cells pretreated with H151. After intratracheal infection with B. abortus, STING KO mice exhibited increased CFU in lungs, spleen and liver, a reduced expression of IFN-β mRNA in lungs and spleen, and reduced levels of proinflammatory cytokines and chemokines in bronchoalveolar lavage fluid (BALF) and lung homogenates. Increased lung CFU and reduced BALF cytokines were also observed in cGAS KO mice. In summary, the cGAS/STING pathway induces the production of proinflammatory cytokines after respiratory Brucella infection, which may contribute to the STING-dependent control of airborne brucellosis.
Collapse
Affiliation(s)
- Iván M. Alonso Paiva
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Raiany A. Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Camila B. Brito
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mariana C. Ferrero
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Manuel Ortiz Wilczyñski
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental (IMEX, CONICET-Academia Nacional de Medicina (ANM)), Buenos Aires, Argentina
| | - Eugenio A. Carrera Silva
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental (IMEX, CONICET-Academia Nacional de Medicina (ANM)), Buenos Aires, Argentina
| | - Sergio C. Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Pablo C. Baldi
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
4
|
Hedges JF, Snyder DT, Robison A, Thompson MA, Aspelin K, Plewa J, Baldridge J, Jutila MA. A TLR4 agonist liposome formulation effectively stimulates innate immunity and enhances protection from bacterial infection. Innate Immun 2023:17534259231168725. [PMID: 37083049 DOI: 10.1177/17534259231168725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
Stimulation of innate immunity can protect against infectious insult and could be used in combination with other therapies. Since antibiotic resistance is an increasing concern, strategies to reduce the dose or eliminate the need for these drugs are warranted. Lipo-CRX is a formulation in which the TLR4 agonist CRX-527 is incorporated into lipid membranes in liposomes. Lipo-CRX is less inflammatory than either CRX-527 or LPS, but retains unique capacity to enhance host defense responses. We compared lipo-CRX to other agonists in vitro using mammalian cells and in vivo in mice, and assessed indicators of innate immune responses and protection from bacterial infection. Lipo-CRX is similar to E. coli LPS in its capacity to activate bovine γδ T cells and to recruit neutrophils into mouse lungs, but with less reactivity in the LAL assay. However, lipo-CRX uniquely induced the production of systemic innate immune cytokines. In the mouse model of brucellosis, delivery of lipo-CRX to the lungs reduced the dissemination of B. abortus. While lipo-CRX or the antibiotic ampicillin alone did not alter B. abortus burdens in the lung, the combination had a synergistic beneficial effect. Our data suggest that stimulating the innate immune system with lipo-CRX, either alone or when combined with antibiotics, can enhance bacterial clearance in the mouse model of brucellosis.
Collapse
Affiliation(s)
- Jodi F Hedges
- Department of Microbiology and Cell Biology, Montana State University, P.O. Box 173610, Bozeman, MT, USA
| | - Deann T Snyder
- Department of Microbiology and Cell Biology, Montana State University, P.O. Box 173610, Bozeman, MT, USA
| | - Amanda Robison
- Department of Microbiology and Cell Biology, Montana State University, P.O. Box 173610, Bozeman, MT, USA
| | - Macy A Thompson
- Department of Microbiology and Cell Biology, Montana State University, P.O. Box 173610, Bozeman, MT, USA
| | - Klara Aspelin
- Department of Microbiology and Cell Biology, Montana State University, P.O. Box 173610, Bozeman, MT, USA
| | - Jack Plewa
- Department of Microbiology and Cell Biology, Montana State University, P.O. Box 173610, Bozeman, MT, USA
| | - Jory Baldridge
- Totem BioSciences, 240 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Mark A Jutila
- Department of Microbiology and Cell Biology, Montana State University, P.O. Box 173610, Bozeman, MT, USA
| |
Collapse
|
5
|
Li J, Qi L, Diao Z, Zhang M, Li B, Zhai Y, Hao M, Zhou D, Liu W, Jin Y, Wang A. Brucella BtpB Manipulates Apoptosis and Autophagic Flux in RAW264.7 Cells. Int J Mol Sci 2022; 23:ijms232214439. [PMID: 36430916 PMCID: PMC9693124 DOI: 10.3390/ijms232214439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/02/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Brucella transfers effectors into host cells, manipulating cellular processes to its advantage; however, the mechanism by which effectors regulate cellular processes during infection is poorly understood. A growing number of studies have shown that apoptosis and autophagy are critical mechanisms for target cells to cope with pathogens and maintain cellular homeostasis. BtpB is a Brucella type IV secretion system effector with a complex mechanism for manipulating host infection. Here, we show that the ectopic expression of BtpB promoted DNA fragmentation. In contrast, an isogenic mutant strain, ΔbtpB, inhibited apoptosis compared to the wild-type strain B. suis S2 in RAW264.7 cells. In addition, BtpB inhibited autophagy, as determined by LC3-II protein levels, the number of LC3 puncta, and p62 degradation. We also found that BtpB reduced autophagolysosome formation and blocked the complete autophagic flux. Moreover, our results revealed that the autophagy inhibitor, chloroquine, reduces Brucella's intracellular survival. Overall, our data unveil new mechanisms of virulence implicating the effector BtpB in regulating host intracellular infection.
Collapse
Affiliation(s)
- Junmei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Lin Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Ziyang Diao
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Mengyu Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Bin Li
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Yunyi Zhai
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Mingyue Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Dong Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Wei Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Yaping Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Aihua Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
- Correspondence: or
| |
Collapse
|
6
|
Stranahan LW, Garcia-Gonzalez DG, Hensel ME, Arenas-Gamboa AM. Primary and memory immune responses against rough Brucella canis are less robust compared to smooth B. abortus and B. melitensis following intratracheal infection in mice. Front Immunol 2022; 13:959328. [PMID: 36032120 PMCID: PMC9402402 DOI: 10.3389/fimmu.2022.959328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/25/2022] [Indexed: 11/20/2022] Open
Abstract
Brucella canis is the cause of canine brucellosis, a globally distributed, zoonotic pathogen which primarily causes disease in dogs. B. canis is unique amongst the zoonotic Brucella spp. with its rough lipopolysaccharide, a trait typically associated with attenuation in gram-negative bacteria. Unfortunately, no vaccine is available against B. canis, and vaccine development is hampered by a limited understanding of the immune response required to combat it and the course of infection following a physiologically relevant, mucosal route of inoculation. To address these concerns and analyze the impact of the rough phenotype on the immune response, we infected mice intratracheally with rough B. canis or smooth B. melitensis or B. abortus. Bacterial colonization and histologic lesions were assessed in systemic target organs as well as locally in the lungs and draining mediastinal lymph node. Mice were also reinfected with Brucella following antibiotic treatment and cytokine production by T lymphocytes in the lung and spleen was assessed by flow cytometry to investigate the memory immune response. Despite its rough phenotype, B. canis established a persistent infection at the same level of colonization as the smooth strains. However, B. canis induced significantly less granulomatous inflammation in the spleen as well as a lack of bronchial-associated lymphoid tissue (BALT) hyperplasia in the lungs. These differences coincided with increased IL-10 and decreased IFN-γ in the spleen of B. canis-infected mice. Previous exposure to all Brucella strains provided protection against colonization following secondary challenge, although induction of IFN-γ by T lymphocytes was seen only in the lungs during B. canis infection while the smooth strains induced this cytokine in the spleen as well. Neither Brucella strain induced significant polyfunctional T lymphocytes, a potential immunomodulatory mechanism that appears to be independent of lipopolysaccharide phenotype.
Collapse
|
7
|
Baldi PC. Role of Btp proteins in the pathogenesis of Brucella infection acquired through the airways. Microb Pathog 2022; 167:105567. [DOI: 10.1016/j.micpath.2022.105567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
8
|
Pierce CF, Brown VR, Olsen SC, Boggiatto P, Pedersen K, Miller RS, Speidel SE, Smyser TJ. Loci Associated With Antibody Response in Feral Swine ( Sus scrofa) Infected With Brucella suis. Front Vet Sci 2020; 7:554674. [PMID: 33324693 PMCID: PMC7724110 DOI: 10.3389/fvets.2020.554674] [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: 04/22/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022] Open
Abstract
Feral swine (Sus scrofa) are a destructive invasive species widespread throughout the United States that disrupt ecosystems, damage crops, and carry pathogens of concern for the health of domestic stock and humans including Brucella suis-the causative organism for swine brucellosis. In domestic swine, brucellosis results in reproductive failure due to abortions and infertility. Contact with infected feral swine poses spillover risks to domestic pigs as well as humans, companion animals, wildlife, and other livestock. Genetic factors influence the outcome of infectious diseases; therefore, genome wide association studies (GWAS) of differential immune responses among feral swine can provide an understanding of disease dynamics and inform management to prevent the spillover of brucellosis from feral swine to domestic pigs. We sought to identify loci associated with differential antibody responses among feral swine naturally infected with B. suis using a case-control GWAS. Tissue, serum, and genotype data (68,516 bi-allelic single nucleotide polymorphisms) collected from 47 feral swine were analyzed in this study. The 47 feral swine were culture positive for Brucella spp. Of these 47, 16 were antibody positive (cases) whereas 31 were antibody negative (controls). Single-locus GWAS were performed using efficient mixed-model association eXpedited (EMMAX) methodology with three genetic models: additive, dominant, and recessive. Eight loci associated with seroconversion were identified on chromosome 4, 8, 9, 10, 12, and 18. Subsequent bioinformatic analyses revealed nine putative candidate genes related to immune function, most notably phagocytosis and induction of an inflammatory response. Identified loci and putative candidate genes may play an important role in host immune responses to B. suis infection, characterized by a detectable bacterial presence yet a differential antibody response. Given that antibody tests are used to evaluate brucellosis infection in domestic pigs and for disease surveillance in invasive feral swine, additional studies are needed to fully understand the genetic component of the response to B. suis infection and to more effectively translate estimates of Brucella spp. antibody prevalence among feral swine to disease control management action.
Collapse
Affiliation(s)
- Courtney F. Pierce
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Research Center, Fort Collins, CO, United States
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, United States
| | - Vienna R. Brown
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Feral Swine Damage Management Program, Fort Collins, CO, United States
| | - Steven C. Olsen
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Diseases, National Animal Disease Center, Ames, IA, United States
| | - Paola Boggiatto
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Diseases, National Animal Disease Center, Ames, IA, United States
| | - Kerri Pedersen
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, Raleigh, NC, United States
| | - Ryan S. Miller
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Veterinary Services, Center for Epidemiology and Animal Health, Fort Collins, CO, United States
| | - Scott E. Speidel
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, United States
| | - Timothy J. Smyser
- United States Department of Agriculture, Animal and Plant Health Inspection Service, Wildlife Services, National Wildlife Research Center, Fort Collins, CO, United States
| |
Collapse
|
9
|
Pathogenesis and immune response in Brucella infection acquired by the respiratory route. Microbes Infect 2020; 22:407-415. [DOI: 10.1016/j.micinf.2020.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/25/2020] [Accepted: 06/02/2020] [Indexed: 01/18/2023]
|
10
|
Hasenauer FC, Rossi UA, Caffaro ME, Raschia MA, Maurizio E, Poli MA, Rossetti CA. Association of TNF rs668920841 and INRA111 polymorphisms with caprine brucellosis: A case-control study of candidate genes involved in innate immunity. Genomics 2020; 112:3925-3932. [PMID: 32629097 DOI: 10.1016/j.ygeno.2020.06.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/25/2020] [Accepted: 06/28/2020] [Indexed: 01/24/2023]
Abstract
Caprine brucellosis is an infectious, contagious zoonotic disease caused by Brucella melitensis. Multiple factors, including host genetics, can influence the outcome of the exposure to Brucella; and it is expected that genetic variants that affect the host innate immune response could have a key role in Brucella infection and pathogenesis. In this study, we evaluated if polymorphisms in innate immunity-related genes are associated with results of Brucella infection in goats. Nine polymorphisms within interferon gamma (IFNG), tumor necrosis factor (TNF), MyD88 innate immune signal transduction adaptor (MYD88), interleukin 10 (IL10) and IL-10 receptor subunit alpha (IL10RA) genes and two molecular markers (BMS2753 and INRA111) were resolved by PCR-capillary electrophoresis in samples from 81 seronegative and 61 seropositive goats for brucellosis. A heterozygous genotype at INRA111, a microsatellite near the VRK serine/threonine kinase 2 (VRK2) gene, was associated with absence of Brucella-specific antibodies in goats naturally exposed to the pathogen (P = .004). Conversely, variants in the TNF gene (rs668920841) and near the IFN gamma receptor 1 (IFNGR1) gene (microsatellite BMS2753) were significantly associated with presence of Brucella-specific antibodies at allelic (P = .042 and P = .046) and genotypic level (P = .012 and P = .041, respectively). Moreover, an in silico analysis predicted a functional role of the insertion-deletion polymorphism rs668920841 on the transcriptional regulation of the caprine TNF gene. Altogether, these results contribute to the identification of genetic factors that have a putative effect on the resistance / susceptibility phenotype of goats to Brucella infection.
Collapse
Affiliation(s)
- F C Hasenauer
- Instituto de Patobiología, CICVyA, INTA, Nicolás Repetto y de Los Reseros s/n, Hurlingham, B1686, Buenos Aires, Argentina; CONICET, Godoy Cruz 2290, C1425 CABA, Argentina
| | - U A Rossi
- Instituto de Patobiología, CICVyA, INTA, Nicolás Repetto y de Los Reseros s/n, Hurlingham, B1686, Buenos Aires, Argentina; CONICET, Godoy Cruz 2290, C1425 CABA, Argentina
| | - M E Caffaro
- Instituto de Genética "Ewald A. Favret", CICVyA, INTA, Nicolás Repetto y de Los Reseros s/n, Hurlingham, B1686, Buenos Aires, Argentina
| | - M A Raschia
- Instituto de Genética "Ewald A. Favret", CICVyA, INTA, Nicolás Repetto y de Los Reseros s/n, Hurlingham, B1686, Buenos Aires, Argentina
| | - E Maurizio
- Instituto de Patobiología, CICVyA, INTA, Nicolás Repetto y de Los Reseros s/n, Hurlingham, B1686, Buenos Aires, Argentina; CONICET, Godoy Cruz 2290, C1425 CABA, Argentina
| | - M A Poli
- Instituto de Genética "Ewald A. Favret", CICVyA, INTA, Nicolás Repetto y de Los Reseros s/n, Hurlingham, B1686, Buenos Aires, Argentina
| | - C A Rossetti
- Instituto de Patobiología, CICVyA, INTA, Nicolás Repetto y de Los Reseros s/n, Hurlingham, B1686, Buenos Aires, Argentina.
| |
Collapse
|
11
|
Zavattieri L, Ferrero MC, Alonso Paiva IM, Sotelo AD, Canellada AM, Baldi PC. Brucella abortus Proliferates in Decidualized and Non-Decidualized Human Endometrial Cells Inducing a Proinflammatory Response. Pathogens 2020; 9:pathogens9050369. [PMID: 32408491 PMCID: PMC7281465 DOI: 10.3390/pathogens9050369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/17/2020] [Accepted: 04/17/2020] [Indexed: 01/18/2023] Open
Abstract
Brucella spp. have been associated with abortion in humans and animals. Although the mechanisms involved are not well established, it is known that placental Brucella infection is accompanied by inflammatory phenomena. The ability of Brucella abortus to infect and survive in human endometrial stromal cells (T-HESC cell line) and the cytokine response elicited were evaluated. B. abortus was able to infect and proliferate in both non-decidualized and decidualized T-HESC cells. Intracellular proliferation depended on the expression of a functional virB operon in the pathogen. B. abortus internalization was inhibited by cytochalasin D and to a lower extent by colchicine, but was not affected by monodansylcadaverine. The infection did not induce cytotoxicity and did not alter the decidualization status of cells. B. abortus infection elicited the secretion of IL-8 and MCP-1 in either decidualized or non-decidualized T-HESC, a response also induced by heat-killed B. abortus and outer membrane vesicles derived from this bacterium. The stimulation of T-HESC with conditioned media from Brucella-infected macrophages induced the production of IL-6, MCP-1 and IL-8 in a dose-dependent manner, and this effect was shown to depend on IL-1β and TNF-α. The proinflammatory responses of T-HESC to B. abortus and to factors produced by infected macrophages may contribute to the gestational complications of brucellosis.
Collapse
Affiliation(s)
- Lucía Zavattieri
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (L.Z.); (M.C.F.); (I.M.A.P.); (A.D.S.); (A.M.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires 1033, Argentina
| | - Mariana C. Ferrero
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (L.Z.); (M.C.F.); (I.M.A.P.); (A.D.S.); (A.M.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires 1033, Argentina
| | - Iván M. Alonso Paiva
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (L.Z.); (M.C.F.); (I.M.A.P.); (A.D.S.); (A.M.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires 1033, Argentina
| | - Agustina D. Sotelo
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (L.Z.); (M.C.F.); (I.M.A.P.); (A.D.S.); (A.M.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires 1033, Argentina
| | - Andrea M. Canellada
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (L.Z.); (M.C.F.); (I.M.A.P.); (A.D.S.); (A.M.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires 1033, Argentina
| | - Pablo C. Baldi
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (L.Z.); (M.C.F.); (I.M.A.P.); (A.D.S.); (A.M.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires 1033, Argentina
- Correspondence: ; Tel.: +54-11-5287-4419
| |
Collapse
|
12
|
Muñoz González F, Sycz G, Alonso Paiva IM, Linke D, Zorreguieta A, Baldi PC, Ferrero MC. The BtaF Adhesin Is Necessary for Full Virulence During Respiratory Infection by Brucella suis and Is a Novel Immunogen for Nasal Vaccination Against Brucella Infection. Front Immunol 2019; 10:1775. [PMID: 31402921 PMCID: PMC6676368 DOI: 10.3389/fimmu.2019.01775] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/15/2019] [Indexed: 01/18/2023] Open
Abstract
Brucella enters their hosts mostly through mucosae from where it spreads systemically. Adhesion to extracellular matrix (ECM) components or to host cells is important for the infectious process, and is mediated by several adhesins, including the BtaF trimeric autotransporter. Although Th1 responses and gamma interferon (IFN-γ) are important for protection, antibodies able to block adhesions might also contribute to prevent Brucella infection. We evaluated the importance of BtaF for respiratory Brucella infection, and characterized the immune response and protection from mucosal challenge induced by nasal vaccination with recombinant BtaF. While lung CFU numbers did not differ at day 1 p.i. between mice intratracheally inoculated with B. suis M1330 (wild type) and those receiving a ΔbtaF mutant, they were reduced in the latter group at 7 and 30 days p.i. For vaccination studies the BtaF passenger domain was engineered and expressed as a soluble trimeric protein. Mice were immunized by the nasal route with BtaF or saline (control group) plus the mucosal adjuvant c-di-AMP. Specific anti-BtaF antibodies (IgG and IgA) were increased in serum, including a mixed IgG2a/IgG1 response. In vitro, these antibodies reduced bacterial adhesion to A549 alveolar epithelial cells. Specific IgA antibodies were also increased in several mucosae. Spleen cells from BtaF immunized mice significantly increased their IL-2, IL-5, IL-17, and IFN-γ secretion upon antigen stimulation. In cervical draining lymph nodes, antigen-experienced CD4+ T cells were maintained mainly as central memory cells. A BtaF-specific delayed-type hypersensitivity response was detected in BtaF immunized mice. Lung cells from the latter produced high levels of IFN-γ upon antigen stimulation. Although nasal immunization with BtaF did not protect mice against B. suis respiratory challenge, it conferred significant protection from intragastric challenge; the splenic load of B. suis was reduced by 3.28 log CFU in immunized mice. This study shows that nasal vaccination with BtaF+c-di-AMP protects against intragastric challenge with B. suis by inducing local and systemic antibody responses, central memory CD4+ T cells and strong Th1 responses. Therefore, although BtaF vaccination did not protect from B. suis respiratory infection, this adhesin constitutes a promising immunogen against mucosal B. suis infection.
Collapse
Affiliation(s)
- Florencia Muñoz González
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriela Sycz
- Fundación Instituto Leloir, IIBBA-CONICET, Buenos Aires, Argentina
| | - Iván M Alonso Paiva
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dirk Linke
- Centre for Ecological and Evolutionary Synthesis, University of Oslo, Oslo, Norway
| | | | - Pablo C Baldi
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariana C Ferrero
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
13
|
Demars A, Lison A, Machelart A, Van Vyve M, Potemberg G, Vanderwinden JM, De Bolle X, Letesson JJ, Muraille E. Route of Infection Strongly Impacts the Host-Pathogen Relationship. Front Immunol 2019; 10:1589. [PMID: 31354728 PMCID: PMC6637429 DOI: 10.3389/fimmu.2019.01589] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/25/2019] [Indexed: 12/24/2022] Open
Abstract
Live attenuated vaccines play a key role in the control of many human and animal pathogens. Their rational development is usually helped by identification of the reservoir of infection, the lymphoid subpopulations associated with protective immunity as well as the virulence genes involved in pathogen persistence. Here, we compared the course of Brucella melitensis infection in C57BL/6 mice infected via intraperitoneal (i.p.), intranasal (i.n.) and intradermal (i.d.) route and demonstrated that the route of infection strongly impacts all of these parameters. Following i.p. and i.n. infection, most infected cells observed in the spleen or lung were F4/80+ myeloid cells. In striking contrast, infected Ly6G+ neutrophils and CD140a+ fibroblasts were also observed in the skin after i.d. infection. The virB operon encoding for the type IV secretion system is considered essential to deflecting vacuolar trafficking in phagocytic cells and allows Brucella to multiply and persist. Unexpectedly, the ΔvirB Brucella strain, which does not persist in the lung after i.n. infection, persists longer in skin tissues than the wild strain after i.d. infection. While the CD4+ T cell-mediated Th1 response is indispensable to controlling the Brucella challenge in the i.p. model, it is dispensable for the control of Brucella in the i.d. and i.n. models. Similarly, B cells are indispensable in the i.p. and i.d. models but dispensable in the i.n. model. γδ+ T cells appear able to compensate for the absence of αβ+ T cells in the i.d. model but not in the other models. Taken together, our results demonstrate the crucial importance of the route of infection for the host pathogen relationship.
Collapse
Affiliation(s)
- Aurore Demars
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Aurore Lison
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Arnaud Machelart
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Margaux Van Vyve
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Georges Potemberg
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | | | - Xavier De Bolle
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Jean-Jacques Letesson
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Eric Muraille
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Bruxelles, Belgium
| |
Collapse
|
14
|
Hielpos MS, Fernández AG, Falivene J, Alonso Paiva IM, Muñoz González F, Ferrero MC, Campos PC, Vieira AT, Oliveira SC, Baldi PC. IL-1R and Inflammasomes Mediate Early Pulmonary Protective Mechanisms in Respiratory Brucella Abortus Infection. Front Cell Infect Microbiol 2018; 8:391. [PMID: 30456207 PMCID: PMC6231193 DOI: 10.3389/fcimb.2018.00391] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/17/2018] [Indexed: 12/19/2022] Open
Abstract
Brucella spp. infection is frequently acquired through contaminated aerosols. The role of interleukin-1 beta (IL-1β) in the early pulmonary response to respiratory Brucella infection is unknown. As shown here, IL-1β levels in lung homogenates and bronchoalveolar lavage fluid (BALF) of mice intratracheally inoculated with B. abortus were increased at 3 and 7 days p.i. At 7 days p.i., pulmonary CFU numbers were higher in IL-1 receptor (IL-1R) knockout (KO) mice than in wild type (WT) mice. At different times p.i. CFU in lungs and BALF were higher in mice lacking some inflammasome components (caspase-1, AIM2, NLRP3) than in WT mice. At 2 days p.i. pulmonary levels of IL-1β and CXCL1 (neutrophils chemoattractant) were lower in caspase-1/11 KO mice. At day 3 p.i., neutrophils counts in BALF were lower in caspase-1/11 KO mice than in WT mice. During in vitro infections, IL-1β secretion was lower in alveolar macrophages from caspase-1/11, NLRP3 or AIM2 KO mice than in WT controls. Similarly, IL-1β production by B. abortus-infected alveolar epithelial cells was reduced by pretreatment with a specific caspase-1 inhibitor. This study shows that IL-1R, probably through IL-1β action, and the NLRP3 and AIM2 inflammasomes are involved in pulmonary innate immune protective mechanisms against respiratory B. abortus infection.
Collapse
Affiliation(s)
- M Soledad Hielpos
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral, Buenos Aires, Argentina
| | - Andrea G Fernández
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral, Buenos Aires, Argentina
| | - Juliana Falivene
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral, Buenos Aires, Argentina
| | - Iván M Alonso Paiva
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral, Buenos Aires, Argentina
| | - Florencia Muñoz González
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral, Buenos Aires, Argentina
| | - Mariana C Ferrero
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral, Buenos Aires, Argentina
| | - Priscila C Campos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Angelica T Vieira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Sergio Costa Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Pablo C Baldi
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral, Buenos Aires, Argentina
| |
Collapse
|
15
|
Fu Y, Wang Z, Lu B, Zhao S, Zhang Y, Zhao Z, Zhang C, Li J, Zhou B, Guo Z, Qian J, Liu L. Immune response and differentially expressed proteins in the lung tissue of BALB/c mice challenged by aerosolized Brucella melitensis 5. J Int Med Res 2018; 46:4740-4752. [PMID: 30282518 PMCID: PMC6259401 DOI: 10.1177/0300060518799879] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective This study was performed to develop a murine aerosol infection model of brucellosis to investigate the pathogenicity and immune reactions induced by aerosolized Brucella and to identify key proteins associated with Brucella infection in lung tissue. Methods BALB/c mice were exposed to aerosolized Brucella melitensis 5 (M5) for 30 minutes and killed at 1, 3, 7, and 15 days post-exposure. Clinical observation, pathological analysis of lung tissue, and cytokine expression detection were then performed. Proteomic analysis based on two-dimensional electrophoresis and mass spectrometry was used to identify proteins exhibiting significant changes in expression in lung tissues during Brucella infection. Results Pathological analysis revealed alveolar wall thickening, telangiectasia with hyperemia, inflammatory cell infiltration, large areas of congestion and bleeding, and areas of focal necrosis. The T-helper 1 type immune response played an important role during aerosol infection, and 12 differentially expressed proteins were involved in the infectious process in lung tissue. Conclusion These results contribute to our understanding of the pathogenic process of Brucella in the lung tissue of BALB/c mice challenged with aerosolized Brucella. Some of the identified proteins may be potential targets in future therapeutic strategies.
Collapse
Affiliation(s)
- Yingying Fu
- Academy of Military Medical Sciences, Beijing, China
| | - Zhongyi Wang
- Academy of Military Medical Sciences, Beijing, China
| | - Bing Lu
- Academy of Military Medical Sciences, Beijing, China
| | - Siyan Zhao
- Academy of Military Medical Sciences, Beijing, China
| | - Yi Zhang
- Academy of Military Medical Sciences, Beijing, China
| | | | - Chunmao Zhang
- Academy of Military Medical Sciences, Beijing, China
| | - Jiaming Li
- Academy of Military Medical Sciences, Beijing, China
| | - Bo Zhou
- Academy of Military Medical Sciences, Beijing, China
| | - Zhendong Guo
- Academy of Military Medical Sciences, Beijing, China
| | - Jun Qian
- Academy of Military Medical Sciences, Beijing, China
| | - Linna Liu
- Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|