1
|
Li X, Wang B, Li X, He J, Shi Y, Wang R, Li D, Haitao D. Analysis and validation of serum biomarkers in brucellosis patients through proteomics and bioinformatics. Front Cell Infect Microbiol 2025; 14:1446339. [PMID: 39872944 PMCID: PMC11769985 DOI: 10.3389/fcimb.2024.1446339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/24/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction This study aims to utilize proteomics, bioinformatics, and machine learning algorithms to identify diagnostic biomarkers in the serum of patients with acute and chronic brucellosis. Methods Proteomic analysis was conducted on serum samples from patients with acute and chronic brucellosis, as well as from healthy controls. Differential expression analysis was performed to identify proteins with altered expression, while Weighted Gene Co-expression Network Analysis (WGCNA) was applied to detect co-expression modules associated with clinical features of brucellosis. Machine learning algorithms were subsequently used to identify the optimal combination of diagnostic biomarkers. Finally, ELISA was employed to validate the identified proteins. Results A total of 1,494 differentially expressed proteins were identified, revealing two co-expression modules significantly associated with the clinical characteristics of brucellosis. The Gaussian Mixture Model (GMM) algorithm identified six proteins that were concurrently present in both the differentially expressed and co-expression modules, demonstrating promising diagnostic potential. After ELISA validation, five proteins were ultimately selected. Discussion These five proteins are implicated in the innate immune processes of brucellosis, potentially associated with its pathogenic mechanisms and chronicity. Furthermore, we highlighted their potential as diagnostic biomarkers for brucellosis. This study further enhances our understanding of brucellosis at the protein level, paving the way for future research endeavors.
Collapse
Affiliation(s)
- Xiao Li
- Department of Inner Mongolia Clinical Medicine College, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Bo Wang
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Xiaocong Li
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Juan He
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Yue Shi
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Rui Wang
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Dongwei Li
- Department of Inner Mongolia Clinical Medicine College, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Ding Haitao
- Department of Clinical Laboratory Medicine Center, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Hohhot, Inner Mongolia, China
| |
Collapse
|
2
|
Yu H, Gu X, Wang D, Wang Z. Brucella infection and Toll-like receptors. Front Cell Infect Microbiol 2024; 14:1342684. [PMID: 38533384 PMCID: PMC10963510 DOI: 10.3389/fcimb.2024.1342684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/17/2024] [Indexed: 03/28/2024] Open
Abstract
Brucella consists of gram-negative bacteria that have the ability to invade and replicate in professional and non-professional phagocytes, and its prolonged persistence in the host leads to brucellosis, a serious zoonosis. Toll-like receptors (TLRs) are the best-known sensors of microorganisms implicated in the regulation of innate and adaptive immunity. In particular, TLRs are transmembrane proteins with a typical structure of an extracellular leucine-rich repeat (LRR) region and an intracellular Toll/interleukin-1 receptor (TIR) domain. In this review, we discuss Brucella infection and the aspects of host immune responses induced by pathogens. Furthermore, we summarize the roles of TLRs in Brucella infection, with substantial emphasis on the molecular insights into its mechanisms of action.
Collapse
Affiliation(s)
- Hui Yu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Xinyi Gu
- The College of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Danfeng Wang
- The College of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| |
Collapse
|
3
|
Ravi S, Martin LC, Krishnan M, Kumaresan M, Manikandan B, Ramar M. Interactions between macrophage membrane and lipid mediators during cardiovascular diseases with the implications of scavenger receptors. Chem Phys Lipids 2024; 258:105362. [PMID: 38006924 DOI: 10.1016/j.chemphyslip.2023.105362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The onset and progression of cardiovascular diseases with the major underlying cause being atherosclerosis, occur during chronic inflammatory persistence in the vascular system, especially within the arterial wall. Such prolonged maladaptive inflammation is driven by macrophages and their key mediators are generally attributed to a disparity in lipid metabolism. Macrophages are the primary cells of innate immunity, endowed with expansive membrane domains involved in immune responses with their signalling systems. During atherosclerosis, the membrane domains and receptors control various active organisations of macrophages. Their scavenger/endocytic receptors regulate the trafficking of intracellular and extracellular cargo. Corresponding influence on lipid metabolism is mediated by their dynamic interaction with scavenger membrane receptors and their integrated mechanisms such as pinocytosis, phagocytosis, cholesterol export/import, etc. This interaction not only results in the functional differentiation of macrophages but also modifies their structural configurations. Here, we reviewed the association of macrophage membrane biomechanics and their scavenger receptor families with lipid metabolites during the event of atherogenesis. In addition, the membrane structure of macrophages and the signalling pathways involved in endocytosis integrated with lipid metabolism are detailed. This article establishes future insights into the scavenger receptors as potential targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Manikandan Kumaresan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai 600 015, India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
4
|
Razei A, Javanbakht M, Hajizade A, Heiat M, Zhao S, Aghamollaei H, Saadati M, Khafaei M, Asadi M, Cegolon L, Keihan AH. Nano and microparticle drug delivery systems for the treatment of Brucella infections. Biomed Pharmacother 2023; 169:115875. [PMID: 37979375 DOI: 10.1016/j.biopha.2023.115875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023] Open
Abstract
Nano-based drug delivery systems are increasingly used for diagnosis, prevention and treatment of several diseases, thanks to several beneficial properties, including the ability to target specific cells or organs, allowing to reduce treatment costs and side effects frequently associated with chemotherapeutic medications, thereby improving treatment compliance of patients. In the field of communicable diseases, especially those caused by intracellular bacteria, the delivery of antibiotics targeting specific cells is of critical importance to maximize their treatment efficacy. Brucella melitensis, an intracellular obligate bacterium surviving and replicating inside macrophages is hard to be eradicated, mainly because of the low ability of antibiotics to enter these phagocityc cells . Although different antibiotics regimens including gentamicin, doxycycline and rifampicin are in fact used against the Brucellosis, no efficient treatment has been attained yet, due to the intracellular life of the respective pathogen. Nano-medicines responding to environmental stimuli allow to maximize drug delivery targeting macropages, thereby boosting treatment efficacy. Several drug delivery nano-technologies, including solid lipid nanoparticles, liposomes, chitosan, niosomes, and their combinations with chitosan sodium alginate can be employed in combination of antibiotics to successfully eradicate Brucellosis infection from patients.
Collapse
Affiliation(s)
- Ali Razei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center,Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Abbas Hajizade
- Biology Research Centre, Faculty of Basic Sciences, Imam Hossain University, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shi Zhao
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China
| | - Hossien Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mojtaba Saadati
- Biology Research Centre, Faculty of Basic Sciences, Imam Hossain University, Tehran, Iran
| | - Mostafa Khafaei
- Human Genetics Research Center, Baqiyatallah Medical Science University, Tehran, Iran
| | - Mosa Asadi
- Nephrology and Urology Research Center,Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Luca Cegolon
- University of Trieste, Department of Medical, Surgical & Health Sciences, Trieste, Italy; University Health Agency Giuliano-Isontina (ASUGI), Public Health Department, Trieste, Italy
| | - Amir Homayoun Keihan
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Guo X, Zeng H, Li M, Xiao Y, Gu G, Song Z, Shuai X, Guo J, Huang Q, Zhou B, Chu Y, Jiao H. The mechanism of chronic intracellular infection with Brucella spp. Front Cell Infect Microbiol 2023; 13:1129172. [PMID: 37143745 PMCID: PMC10151771 DOI: 10.3389/fcimb.2023.1129172] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Globally, brucellosis is a widespread zoonotic disease. It is prevalent in more than 170 countries and regions. It mostly damages an animal's reproductive system and causes extreme economic losses to the animal husbandry industry. Once inside cells, Brucella resides in a vacuole, designated the BCV, which interacts with components of the endocytic and secretory pathways to ensure bacterial survival. Numerous studies conducted recently have revealed that Brucella's ability to cause a chronic infection depends on how it interacts with the host. This paper describes the immune system, apoptosis, and metabolic control of host cells as part of the mechanism of Brucella survival in host cells. Brucella contributes to both the body's non-specific and specific immunity during chronic infection, and it can aid in its survival by causing the body's immune system to become suppressed. In addition, Brucella regulates apoptosis to avoid being detected by the host immune system. The BvrR/BvrS, VjbR, BlxR, and BPE123 proteins enable Brucella to fine-tune its metabolism while also ensuring its survival and replication and improving its ability to adapt to the intracellular environment.
Collapse
Affiliation(s)
- Xiaoyi Guo
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Hui Zeng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Mengjuan Li
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yu Xiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Guojing Gu
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenhui Song
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xuehong Shuai
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jianhua Guo
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Qingzhou Huang
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Bo Zhou
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
- *Correspondence: Bo Zhou, ; Yuefeng Chu, ; Hanwei Jiao,
| | - Yuefeng Chu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- *Correspondence: Bo Zhou, ; Yuefeng Chu, ; Hanwei Jiao,
| | - Hanwei Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
- The Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
- *Correspondence: Bo Zhou, ; Yuefeng Chu, ; Hanwei Jiao,
| |
Collapse
|
6
|
Linares-Alcántara E, Mendlovic F. Scavenger Receptor A1 Signaling Pathways Affecting Macrophage Functions in Innate and Adaptive Immunity. Immunol Invest 2022; 51:1725-1755. [PMID: 34986758 DOI: 10.1080/08820139.2021.2020812] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
First discovered on macrophages by Goldstein and Brown in 1979, Scavenger Receptors have since been shown to participate in a diverse number of cell functions; equally diverse are their structures and the ligands they bind. Macrophage activation is crucial in the outcome of an immune response. SR-A1 is highly abundant on macrophages and recognizes both host- and microorganism-derived molecules that impact processes that are initiated, perpetuated, or modified. This review summarizes the involvement of SR-A1 in both inflammatory and anti-inflammatory responses, the multiple-ligand internalization mechanisms and the diversity of signaling pathways that impact macrophage function and activation. Engagement of SR-A1 results in the stimulation of differential signaling pathways and patterns of cytokine expression, kinetics, magnitude of response and activation status. SR-A1 plays essential roles in phagocytosis and efferocytosis, interacting with other receptors and promoting tolerance in response to apoptotic cell uptake. In cell adhesion, tissue remodeling, and cell migration, SR-A1 signals through different pathways engaging different cytoplasmic motifs. We describe the role of SR-A1 during innate and adaptive immune responses, such as participation in macrophage polarization and interaction with other innate receptors, as well as in antigen uptake, processing, and presentation, regulating T and B cell activation. The dichotomous contribution of SR-A1 on macrophage functions is discussed. A better understanding of the role SR-A1 plays through molecular mechanisms and crosstalk with other receptors may provide insights into developing novel therapeutic strategies to modulate immune responses and immunopathologies.
Collapse
Affiliation(s)
- Elizabeth Linares-Alcántara
- Facultad de Ciencias, UNAM, Av. Universidad 3000, Col. Copilco-Universidad, Ciudad de Mexico, Mexico.,Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Av. Universidad 3000, Col. Copilco-Universidad, Ciudad de Mexico, Mexico
| | - Fela Mendlovic
- Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Av. Universidad 3000, Col. Copilco-Universidad, Ciudad de Mexico, Mexico.,Facultad de Ciencias de la Salud, Universidad Anahuac Mexico Norte, Huixquilucan, Mexico
| |
Collapse
|
7
|
Ouyang A, Wang H, Su J, Liu X. Mannose Receptor Mediates the Activation of Chitooligosaccharides on Blunt Snout Bream ( Megalobrama amblycephala) Macrophages. Front Immunol 2021; 12:686846. [PMID: 34408745 PMCID: PMC8365301 DOI: 10.3389/fimmu.2021.686846] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/16/2021] [Indexed: 01/23/2023] Open
Abstract
Chitooligosaccharide (COS) is an important immune enhancer and has been proven to have a variety of biological activities. Our previous research has established an M1 polarization mode by COS in blunt snout bream (Megalobrama amblycephala) macrophages, but the mechanism of COS activation of blunt snout bream macrophages remains unclear. In this study, we further explored the internalization mechanism and signal transduction pathway of chitooligosaccharide hexamer (COS6) in blunt snout bream macrophages. The results showed that mannose receptor C-type lectin-like domain 4-8 of M. amblycephala (MaMR CTLD4-8) could recognize and bind to COS6 and mediate COS6 into macrophages by both clathrin-dependent and caveolin-dependent pathways. In the inflammatory response of macrophages activated by COS6, the gene expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and nitric oxide synthase 2 (NOS2) was significantly inhibited after MaMR CTLD4-8-specific antibody blockade. However, even if it was blocked, the expression of these inflammation-related genes was still relatively upregulated, which suggested that there are other receptors involved in immune regulation. Further studies indicated that MaMR CTLD4-8 and Toll-like receptor 4 (TLR4) cooperated to regulate the pro-inflammatory response of macrophages caused by COS6. Taken together, these results revealed that mannose receptor (MR) CTLD4-8 is indispensable in the process of recognition, binding, internalization, and immunoregulation of COS in macrophages of blunt snout bream.
Collapse
Affiliation(s)
- Aotian Ouyang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Huabing Wang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jianguo Su
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Xiaoling Liu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
| |
Collapse
|
8
|
Stranahan LW, Arenas-Gamboa AM. When the Going Gets Rough: The Significance of Brucella Lipopolysaccharide Phenotype in Host-Pathogen Interactions. Front Microbiol 2021; 12:713157. [PMID: 34335551 PMCID: PMC8319746 DOI: 10.3389/fmicb.2021.713157] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/22/2021] [Indexed: 01/18/2023] Open
Abstract
Brucella is a facultatively intracellular bacterial pathogen and the cause of worldwide zoonotic infections, infamous for its ability to evade the immune system and persist chronically within host cells. Despite the frequent association with attenuation in other Gram-negative bacteria, a rough lipopolysaccharide phenotype is retained by Brucella canis and Brucella ovis, which remain fully virulent in their natural canine and ovine hosts, respectively. While these natural rough strains lack the O-polysaccharide they, like their smooth counterparts, are able to evade and manipulate the host immune system by exhibiting low endotoxic activity, resisting destruction by complement and antimicrobial peptides, entering and trafficking within host cells along a similar pathway, and interfering with MHC-II antigen presentation. B. canis and B. ovis appear to have compensated for their roughness by alterations to their outer membrane, especially in regards to outer membrane proteins. B. canis, in particular, also shows evidence of being less proinflammatory in vivo, suggesting that the rough phenotype may be associated with an enhanced level of stealth that could allow these pathogens to persist for longer periods of time undetected. Nevertheless, much additional work is required to understand the correlates of immune protection against the natural rough Brucella spp., a critical step toward development of much-needed vaccines. This review will highlight the significance of rough lipopolysaccharide in the context of both natural disease and host–pathogen interactions with an emphasis on natural rough Brucella spp. and the implications for vaccine development.
Collapse
Affiliation(s)
- Lauren W Stranahan
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Angela M Arenas-Gamboa
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
9
|
Oishi K, Morise M, Vo LK, Tran NT, Sahashi D, Ueda-Wakamatsu R, Nishimura W, Komatsu M, Shiozaki K. Host lactosylceramide enhances Edwardsiella tarda infection. Cell Microbiol 2021; 23:e13365. [PMID: 33988901 DOI: 10.1111/cmi.13365] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/01/2021] [Accepted: 05/10/2021] [Indexed: 12/01/2022]
Abstract
Edwardsiella tarda is a Gram-negative bacterium causing economic damage in aquaculture. The interaction of E. tarda with microdomains is an important step in the invasion, but the target molecules in microdomains remain undefined. Here, we found that intraperitoneal injection of E. tarda altered splenic glycosphingolipid patterns in the model host medaka (Oryzias latipes) accompanied by alteration of glycosphingolipid metabolism-related gene expressions, suggesting that glycosphingolipid levels are involved in E. tarda infection. To ascertain the significance of glycosphingolipids in the infection, fish cell lines, DIT29 cells with a high amount of lactosylceramide (LacCer) and glucosylceramide (GlcCer), and GAKS cells with a low amount of these lipids, were treated with methyl-β-cyclodextrin to disrupt the microdomain. E. tarda infection was suppressed in DIT29 cells, but not in GAKS cells, suggesting the involvement of microdomain LacCer and GlcCer in the infection. DL-threo-1-phenyl-2-palmitoylamino-3-morpholino-1-propanol, an inhibitor of glycosphingolipid-synthesis, attenuated the infection in DIT29 cells, while Neu3-overexpressing GAKS cells, which accumulated LacCer, enhanced the infection. E. tarda possessed binding ability towards LacCer, but not GlcCer, and LacCer preincubation declined the infection towards fish cells, possibly due to the masking of binding sites. The present study suggests that LacCer may be a positive regulator of E. tarda invasion.
Collapse
Affiliation(s)
- Kazuki Oishi
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Moeri Morise
- Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Linh Khanh Vo
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Nhung Thi Tran
- Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Daichi Sahashi
- Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | | | | | - Masaharu Komatsu
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan.,Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Kazuhiro Shiozaki
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan.,Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
10
|
Jiao H, Zhou Z, Li B, Xiao Y, Li M, Zeng H, Guo X, Gu G. The Mechanism of Facultative Intracellular Parasitism of Brucella. Int J Mol Sci 2021; 22:ijms22073673. [PMID: 33916050 PMCID: PMC8036852 DOI: 10.3390/ijms22073673] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/22/2021] [Accepted: 03/30/2021] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is a highly prevalent zoonotic disease characterized by abortion and reproductive dysfunction in pregnant animals. Although the mortality rate of Brucellosis is low, it is harmful to human health, and also seriously affects the development of animal husbandry, tourism and international trade. Brucellosis is caused by Brucella, which is a facultative intracellular parasitic bacteria. It mainly forms Brucella-containing vacuoles (BCV) in the host cell to avoid the combination with lysosome (Lys), so as to avoid the elimination of it by the host immune system. Brucella not only has the ability to resist the phagocytic bactericidal effect, but also can make the host cells form a microenvironment which is conducive to its survival, reproduction and replication, and survive in the host cells for a long time, which eventually leads to the formation of chronic persistent infection. Brucella can proliferate and replicate in cells, evade host immune response and induce persistent infection, which are difficult problems in the treatment and prevention of Brucellosis. Therefore, the paper provides a preliminary overview of the facultative intracellular parasitic and immune escape mechanisms of Brucella, which provides a theoretical basis for the later study on the pathogenesis of Brucella.
Collapse
Affiliation(s)
- Hanwei Jiao
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, China
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Z.Z.); (B.L.); (Y.X.); (M.L.); (H.Z.); (X.G.); (G.G.)
- Veterinary Scientific Engineering Research Center, Chongqing 402460, China
- Correspondence:
| | - Zhixiong Zhou
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Z.Z.); (B.L.); (Y.X.); (M.L.); (H.Z.); (X.G.); (G.G.)
| | - Bowen Li
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Z.Z.); (B.L.); (Y.X.); (M.L.); (H.Z.); (X.G.); (G.G.)
| | - Yu Xiao
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Z.Z.); (B.L.); (Y.X.); (M.L.); (H.Z.); (X.G.); (G.G.)
| | - Mengjuan Li
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Z.Z.); (B.L.); (Y.X.); (M.L.); (H.Z.); (X.G.); (G.G.)
| | - Hui Zeng
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Z.Z.); (B.L.); (Y.X.); (M.L.); (H.Z.); (X.G.); (G.G.)
| | - Xiaoyi Guo
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Z.Z.); (B.L.); (Y.X.); (M.L.); (H.Z.); (X.G.); (G.G.)
| | - Guojing Gu
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Z.Z.); (B.L.); (Y.X.); (M.L.); (H.Z.); (X.G.); (G.G.)
| |
Collapse
|
11
|
González-Espinoza G, Arce-Gorvel V, Mémet S, Gorvel JP. Brucella: Reservoirs and Niches in Animals and Humans. Pathogens 2021; 10:pathogens10020186. [PMID: 33572264 PMCID: PMC7915599 DOI: 10.3390/pathogens10020186] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 01/18/2023] Open
Abstract
Brucella is an intracellular bacterium that causes abortion, reproduction failure in livestock and leads to a debilitating flu-like illness with serious chronic complications if untreated in humans. As a successful intracellular pathogen, Brucella has developed strategies to avoid recognition by the immune system of the host and promote its survival and replication. In vivo, Brucellae reside mostly within phagocytes and other cells including trophoblasts, where they establish a preferred replicative niche inside the endoplasmic reticulum. This process is central as it gives Brucella the ability to maintain replicating-surviving cycles for long periods of time, even at low bacterial numbers, in its cellular niches. In this review, we propose that Brucella takes advantage of the environment provided by the cellular niches in which it resides to generate reservoirs and disseminate to other organs. We will discuss how the favored cellular niches for Brucella infection in the host give rise to anatomical reservoirs that may lead to chronic infections or persistence in asymptomatic subjects, and which may be considered as a threat for further contamination. A special emphasis will be put on bone marrow, lymph nodes, reproductive and for the first time adipose tissues, as well as wildlife reservoirs.
Collapse
|
12
|
Bialer MG, Sycz G, Muñoz González F, Ferrero MC, Baldi PC, Zorreguieta A. Adhesins of Brucella: Their Roles in the Interaction with the Host. Pathogens 2020; 9:E942. [PMID: 33198223 PMCID: PMC7697752 DOI: 10.3390/pathogens9110942] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 01/30/2023] Open
Abstract
A central aspect of Brucella pathogenicity is its ability to invade, survive, and replicate in diverse phagocytic and non-phagocytic cell types, leading to chronic infections and chronic inflammatory phenomena. Adhesion to the target cell is a critical first step in the invasion process. Several Brucella adhesins have been shown to mediate adhesion to cells, extracellular matrix components (ECM), or both. These include the sialic acid-binding proteins SP29 and SP41 (binding to erythrocytes and epithelial cells, respectively), the BigA and BigB proteins that contain an Ig-like domain (binding to cell adhesion molecules in epithelial cells), the monomeric autotransporters BmaA, BmaB, and BmaC (binding to ECM components, epithelial cells, osteoblasts, synoviocytes, and trophoblasts), the trimeric autotransporters BtaE and BtaF (binding to ECM components and epithelial cells) and Bp26 (binding to ECM components). An in vivo role has also been shown for the trimeric autotransporters, as deletion mutants display decreased colonization after oral and/or respiratory infection in mice, and it has also been suggested for BigA and BigB. Several adhesins have shown unipolar localization, suggesting that Brucella would express an adhesive pole. Adhesin-based vaccines may be useful to prevent brucellosis, as intranasal immunization in mice with BtaF conferred high levels of protection against oral challenge with B. suis.
Collapse
Affiliation(s)
- Magalí G. Bialer
- Fundación Instituto Leloir (FIL), IIBBA (CONICET-FIL), Buenos Aires 1405, Argentina; (M.G.B.); (G.S.)
| | - Gabriela Sycz
- Fundación Instituto Leloir (FIL), IIBBA (CONICET-FIL), Buenos Aires 1405, Argentina; (M.G.B.); (G.S.)
| | - Florencia Muñoz González
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (F.M.G.); (M.C.F.)
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Buenos Aires 1113, Argentina
| | - Mariana C. Ferrero
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (F.M.G.); (M.C.F.)
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Buenos Aires 1113, Argentina
| | - Pablo C. Baldi
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (F.M.G.); (M.C.F.)
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Buenos Aires 1113, Argentina
| | - Angeles Zorreguieta
- Fundación Instituto Leloir (FIL), IIBBA (CONICET-FIL), Buenos Aires 1405, Argentina; (M.G.B.); (G.S.)
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires 1428, Argentina
| |
Collapse
|
13
|
Abstract
Brucellosis is a bacterial disease of domestic animals and humans. The pathogenic ability of Brucella organisms relies on their stealthy strategy and their capacity to replicate within host cells and to induce long-lasting infections. Brucella organisms barely induce neutrophil activation and survive within these leukocytes by resisting microbicidal mechanisms. Very few Brucella-infected neutrophils are found in the target organs, except for the bone marrow, early in infection. Still, Brucella induces a mild reactive oxygen species formation and, through its lipopolysaccharide, promotes the premature death of neutrophils, which release chemokines and express "eat me" signals. This effect drives the phagocytosis of infected neutrophils by mononuclear cells that become thoroughly susceptible to Brucella replication and vehicles for bacterial dispersion. The premature death of the infected neutrophils proceeds without NETosis, necrosis/oncosis, or classical apoptosis morphology. In the absence of neutrophils, the Th1 response exacerbates and promotes bacterial removal, indicating that Brucella-infected neutrophils dampen adaptive immunity. This modulatory effect opens a window for bacterial dispersion in host tissues before adaptive immunity becomes fully activated. However, the hyperactivation of immunity is not without a price, since neutropenic Brucella-infected animals develop cachexia in the early phases of the disease. The delay in the immunological response seems a sine qua non requirement for the development of long-lasting brucellosis. This property may be shared with other pathogenic alphaproteobacteria closely related to Brucella We propose a model in which Brucella-infected polymorphonuclear neutrophils (PMNs) function as "Trojan horse" vehicles for bacterial dispersal and as modulators of the Th1 adaptive immunity in infection.
Collapse
|
14
|
Gómez LA, Alvarez FI, Molina RE, Soto-Shara R, Daza-Castro C, Flores MR, León Y, Oñate AA. A Zinc-Dependent Metalloproteinase of Brucella abortus Is Required in the Intracellular Adaptation of Macrophages. Front Microbiol 2020; 11:1586. [PMID: 32765455 PMCID: PMC7379133 DOI: 10.3389/fmicb.2020.01586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/17/2020] [Indexed: 01/19/2023] Open
Abstract
Brucella abortus is a pathogen that survives in macrophages. Several virulence factors participate in this process, including the open reading frame (ORF) BAB1_0270 codifying for a zinc-dependent metalloproteinase (ZnMP). Here, its contribution in the intracellular adaptation of B. abortus was analyzed by infecting RAW264.7 macrophages with the mutant B. abortus Δ270 strain. Results showed that this ZnMP did not participated in either the adherence or the initial intracellular traffic of B. abortus in macrophages. Nevertheless, its deletion significantly increased the co-localization of B. abortus Δ270 with phagolysosomal cathepsin D and reduced its co-localization with calnexin present in endoplasmic reticulum (RE)-derived vesicles. Although B. abortus Δ270 showed an upregulated expression of genes involved in virulence (vjbR, hutC, bvrR, virB1), it was insufficient to reach a successful intracellular replication within macrophages. Furthermore, its attenuation favored in macrophages infected the production of high levels of cytokines (TNF-α and IL-6) and co-stimulatory proteins (CD80 and CD86), signals required in T cell activation. Finally, its deletion significantly reduced the ability of B. abortus Δ270 to adapt, grow and express several virulence factors under acidic conditions. Based on these results, and considering that this ZnMP has homology with ImmA/IrrE proteases, we discuss its role in the virulence of this pathogen, concluding that ZnMP is required in the intracellular adaptation of B. abortus 2308 during the infection of macrophages.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Angel A. Oñate
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| |
Collapse
|
15
|
Otopathogenic Staphylococcus aureus Invades Human Middle Ear Epithelial Cells Primarily through Cholesterol Dependent Pathway. Sci Rep 2019; 9:10777. [PMID: 31346200 PMCID: PMC6658548 DOI: 10.1038/s41598-019-47079-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/26/2019] [Indexed: 01/20/2023] Open
Abstract
Chronic suppurative otitis media (CSOM) is one of the most common infectious diseases of the middle ear especially affecting children, leading to delay in language development and communication. Although Staphylococcus aureus is the most common pathogen associated with CSOM, its interaction with middle ear epithelial cells is not well known. In the present study, we observed that otopathogenic S. aureus has the ability to invade human middle ear epithelial cells (HMEECs) in a dose and time dependent manner. Scanning electron microscopy demonstrated time dependent increase in the number of S. aureus on the surface of HMEECs. We observed that otopathogenic S. aureus primarily employs a cholesterol dependent pathway to colonize HMEECs. In agreement with these findings, confocal microscopy showed that S. aureus colocalized with lipid rafts in HMEECs. The results of the present study provide new insights into the pathogenesis of S. aureus induced CSOM. The availability of in vitro cell culture model will pave the way to develop novel effective treatment modalities for CSOM beyond antibiotic therapy.
Collapse
|
16
|
Abstract
The entry of pathogens into nonphagocytic host cells has received much attention in the past three decades, revealing a vast array of strategies employed by bacteria and viruses. A method of internalization that has been extensively studied in the context of viral infections is the use of the clathrin-mediated pathway. More recently, a role for clathrin in the entry of some intracellular bacterial pathogens was discovered. Classically, clathrin-mediated endocytosis was thought to accommodate internalization only of particles smaller than 150 nm; however, this was challenged upon the discovery that Listeria monocytogenes requires clathrin to enter eukaryotic cells. Now, with discoveries that clathrin is required during other stages of some bacterial infections, another paradigm shift is occurring. There is a more diverse impact of clathrin during infection than previously thought. Much of the recent data describing clathrin utilization in processes such as bacterial attachment, cell-to-cell spread and intracellular growth may be due to newly discovered divergent roles of clathrin in the cell. Not only does clathrin act to facilitate endocytosis from the plasma membrane, but it also participates in budding from endosomes and the Golgi apparatus and in mitosis. Here, the manipulation of clathrin processes by bacterial pathogens, including its traditional role during invasion and alternative ways in which clathrin supports bacterial infection, is discussed. Researching clathrin in the context of bacterial infections will reveal new insights that inform our understanding of host-pathogen interactions and allow researchers to fully appreciate the diverse roles of clathrin in the eukaryotic cell.
Collapse
Affiliation(s)
- Eleanor A Latomanski
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hayley J Newton
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
17
|
Pombinho R, Sousa S, Cabanes D. Scavenger Receptors: Promiscuous Players during Microbial Pathogenesis. Crit Rev Microbiol 2018; 44:685-700. [PMID: 30318962 DOI: 10.1080/1040841x.2018.1493716] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Innate immunity is the most broadly effective host defense, being essential to clear the majority of microbial infections. Scavenger Receptors comprise a family of sensors expressed in a multitude of host cells, whose dual role during microbial pathogenesis gained importance over recent years. SRs regulate the recruitment of immune cells and control both host inflammatory response and bacterial load. In turn, pathogens have evolved different strategies to overcome immune response, avoid recognition by SRs and exploit them to favor infection. Here, we discuss the most relevant findings regarding the interplay between SRs and pathogens, discussing how these multifunctional proteins recognize a panoply of ligands and act as bacterial phagocytic receptors.
Collapse
Affiliation(s)
- Rita Pombinho
- a Instituto de Investigação e Inovação em Saúde (i3S), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal.,b Instituto de Biologia Molecular e Celular (IBMC), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal
| | - Sandra Sousa
- a Instituto de Investigação e Inovação em Saúde (i3S), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal.,b Instituto de Biologia Molecular e Celular (IBMC), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal
| | - Didier Cabanes
- a Instituto de Investigação e Inovação em Saúde (i3S), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal.,b Instituto de Biologia Molecular e Celular (IBMC), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal
| |
Collapse
|
18
|
PRR Function of Innate Immune Receptors in Recognition of Bacteria or Bacterial Ligands. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1112:255-280. [DOI: 10.1007/978-981-13-3065-0_18] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
19
|
Abstract
Coxiella burnetii is an intracellular bacterial pathogen and a significant cause of culture-negative endocarditis in the United States. Upon infection, the nascent Coxiella phagosome fuses with the host endocytic pathway to form a large lysosome-like vacuole called the parasitophorous vacuole (PV). The PV membrane is rich in sterols, and drugs perturbing host cell cholesterol homeostasis inhibit PV formation and bacterial growth. Using cholesterol supplementation of a cholesterol-free cell model system, we found smaller PVs and reduced Coxiella growth as cellular cholesterol concentration increased. Further, we observed in cells with cholesterol a significant number of nonfusogenic PVs that contained degraded bacteria, a phenotype not observed in cholesterol-free cells. Cholesterol had no effect on axenic Coxiella cultures, indicating that only intracellular bacteria are sensitive to cholesterol. Live-cell microscopy revealed that both plasma membrane-derived cholesterol and the exogenous cholesterol carrier protein low-density lipoprotein (LDL) traffic to the PV. To test the possibility that increasing PV cholesterol levels affects bacterial survival, infected cells were treated with U18666A, a drug that traps cholesterol in lysosomes and PVs. U18666A treatment led to PVs containing degraded bacteria and a significant loss in bacterial viability. The PV pH was significantly more acidic in cells with cholesterol or cells treated with U18666A, and the vacuolar ATPase inhibitor bafilomycin blocked cholesterol-induced PV acidification and bacterial death. Additionally, treatment of infected HeLa cells with several FDA-approved cholesterol-altering drugs led to a loss of bacterial viability, a phenotype also rescued by bafilomycin. Collectively, these data suggest that increasing PV cholesterol further acidifies the PV, leading to Coxiella death. The intracellular Gram-negative bacterium Coxiella burnetii is a significant cause of culture-negative infectious endocarditis, which can be fatal if untreated. The existing treatment strategy requires prolonged antibiotic treatment, with a 10-year mortality rate of 19% in treated patients. Therefore, new clinical therapies are needed and can be achieved by better understanding C. burnetii pathogenesis. Upon infection of host cells, C. burnetii grows within a specialized replication niche, the parasitophorous vacuole (PV). Recent data have linked cholesterol to intracellular C. burnetii growth and PV formation, leading us to further decipher the role of cholesterol during C. burnetii-host interaction. We observed that increasing PV cholesterol concentration leads to increased acidification of the PV and bacterial death. Further, treatment with FDA-approved drugs that alter host cholesterol homeostasis also killed C. burnetii through PV acidification. Our findings suggest that targeting host cholesterol metabolism might prove clinically efficacious in controlling C. burnetii infection.
Collapse
|
20
|
The Bacterial Second Messenger Cyclic di-GMP Regulates Brucella Pathogenesis and Leads to Altered Host Immune Response. Infect Immun 2016; 84:3458-3470. [PMID: 27672085 DOI: 10.1128/iai.00531-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/19/2016] [Indexed: 11/20/2022] Open
Abstract
Brucella species are facultative intracellular bacteria that cause brucellosis, a chronic debilitating disease significantly impacting global health and prosperity. Much remains to be learned about how Brucella spp. succeed in sabotaging immune host cells and how Brucella spp. respond to environmental challenges. Multiple types of bacteria employ the prokaryotic second messenger cyclic di-GMP (c-di-GMP) to coordinate responses to shifting environments. To determine the role of c-di-GMP in Brucella physiology and in shaping host-Brucella interactions, we utilized c-di-GMP regulatory enzyme deletion mutants. Our results show that a ΔbpdA phosphodiesterase mutant producing excess c-di-GMP displays marked attenuation in vitro and in vivo during later infections. Although c-di-GMP is known to stimulate the innate sensor STING, surprisingly, the ΔbpdA mutant induced a weaker host immune response than did wild-type Brucella or the low-c-di-GMP guanylate cyclase ΔcgsB mutant. Proteomics analysis revealed that c-di-GMP regulates several processes critical for virulence, including cell wall and biofilm formation, nutrient acquisition, and the type IV secretion system. Finally, ΔbpdA mutants exhibited altered morphology and were hypersensitive to nutrient-limiting conditions. In summary, our results indicate a vital role for c-di-GMP in allowing Brucella to successfully navigate stressful and shifting environments to establish intracellular infection.
Collapse
|
21
|
Peruń A, Biedroń R, Konopiński MK, Białecka A, Marcinkiewicz J, Józefowski S. Phagocytosis of live versus killed or fluorescently labeled bacteria by macrophages differ in both magnitude and receptor specificity. Immunol Cell Biol 2016; 95:424-435. [PMID: 27826145 DOI: 10.1038/icb.2016.112] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 01/23/2023]
Abstract
Scavenger receptor (SR)-mediated opsonin-independent phagocytosis of bacteria by macrophages has been suggested to represent an important, early mechanism of anti-bacterial host defense. However, although the ability to bind bacteria has been demonstrated to be a shared feature of all types of SRs, in many cases the evidence is limited to the demonstration of increased binding of killed, fluorescently labeled bacteria to non-phagocytic cells transfected with these receptors. We sought to verify the ability of SRs to mediate non-opsonic phagocytosis of live Escherichia coli (Ec) and Staphylococcus aureus (Sa), model species of Gram-negative and -positive bacteria, respectively, and to assess the relative contributions of different SRs expressed on murine macrophages in this process. We found that the class A SR SR-A/CD204 was the major receptor mediating phagocytosis of fluorescently labeled Sa, whereas different SRs had highly redundant roles in the phagocytosis of live Sa. Conversely, different SRs contributed to the phagocytosis of fluorescently labeled Ec. In comparison, phagocytosis of live Ec was of much lower magnitude and was selectively mediated by SR-A. These results question the use of fluorescently labeled bacteria as valid replacements for live bacteria. The low magnitude of opsonin-independent phagocytosis of Ec and unimpaired phagocytosis of Sa in SR-A- or CD36-deficient macrophages indicate that the defect in this process might not be responsible for the reported impaired bacteria clearance in mice deficient in these receptors. We postulate that this impairment might result to a larger extent from inhibition of intracellular bacteria killing caused by pro-inflammatory cytokines, produced in excessive amounts by SR-deficient cells in response to bacterial products.
Collapse
Affiliation(s)
- Angelika Peruń
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| | - Rafał Biedroń
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| | - Maciej K Konopiński
- Institute of Nature Conservation, Polish Academy of Sciences, Cracow, Poland
| | - Anna Białecka
- Centre of Microbiological Research and Autovaccines, Cracow, Poland
| | - Janusz Marcinkiewicz
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| | - Szczepan Józefowski
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
22
|
Ahmed W, Zheng K, Liu ZF. Establishment of Chronic Infection: Brucella's Stealth Strategy. Front Cell Infect Microbiol 2016; 6:30. [PMID: 27014640 PMCID: PMC4791395 DOI: 10.3389/fcimb.2016.00030] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/29/2016] [Indexed: 01/18/2023] Open
Abstract
Brucella is a facultative intracellular pathogen that causes zoonotic infection known as brucellosis which results in abortion and infertility in natural host. Humans, especially in low income countries, can acquire infection by direct contact with infected animal or by consumption of animal products and show high morbidity, severe economic losses and public health problems. However for survival, host cells develop complex immune mechanisms to defeat and battle against attacking pathogens and maintain a balance between host resistance and Brucella virulence. On the other hand as a successful intracellular pathogen, Brucella has evolved multiple strategies to evade immune response mechanisms to establish persistent infection and replication within host. In this review, we mainly summarize the "Stealth" strategies employed by Brucella to modulate innate and the adaptive immune systems, autophagy, apoptosis and possible role of small noncoding RNA in the establishment of chronic infection. The purpose of this review is to give an overview for recent understanding how this pathogen evades immune response mechanisms of host, which will facilitate to understanding the pathogenesis of brucellosis and the development of novel, more effective therapeutic approaches to treat brucellosis.
Collapse
Affiliation(s)
- Waqas Ahmed
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| | - Ke Zheng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| | - Zheng-Fei Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University Wuhan, China
| |
Collapse
|
23
|
Toledo A, Benach JL. Hijacking and Use of Host Lipids by Intracellular Pathogens. Microbiol Spectr 2015; 3:10.1128/microbiolspec.VMBF-0001-2014. [PMID: 27337282 PMCID: PMC5790186 DOI: 10.1128/microbiolspec.vmbf-0001-2014] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Indexed: 12/14/2022] Open
Abstract
Intracellular bacteria use a number of strategies to survive, grow, multiply, and disseminate within the host. One of the most striking adaptations that intracellular pathogens have developed is the ability to utilize host lipids and their metabolism. Bacteria such as Anaplasma, Chlamydia, or Mycobacterium can use host lipids for different purposes, such as a means of entry through lipid rafts, building blocks for bacteria membrane formation, energy sources, camouflage to avoid the fusion of phagosomes and lysosomes, and dissemination. One of the most extreme examples of lipid exploitation is Mycobacterium, which not only utilizes the host lipid as a carbon and energy source but is also able to reprogram the host lipid metabolism. Likewise, Chlamydia spp. have also developed numerous mechanisms to reprogram lipids onto their intracellular inclusions. Finally, while the ability to exploit host lipids is important in intracellular bacteria, it is not an exclusive trait. Extracellular pathogens, including Helicobacter, Mycoplasma, and Borrelia, can recruit and metabolize host lipids that are important for their growth and survival.Throughout this chapter we will review how intracellular and extracellular bacterial pathogens utilize host lipids to enter, survive, multiply, and disseminate in the host.
Collapse
Affiliation(s)
- Alvaro Toledo
- Department of Molecular Genetics and Microbiology, Stony Brook University, Center for Infectious Diseases at the Center for Molecular Medicine, Stony Brook, NY 11794
| | - Jorge L Benach
- Department of Molecular Genetics and Microbiology, Stony Brook University, Center for Infectious Diseases at the Center for Molecular Medicine, Stony Brook, NY 11794
| |
Collapse
|
24
|
de Figueiredo P, Ficht TA, Rice-Ficht A, Rossetti CA, Adams LG. Pathogenesis and immunobiology of brucellosis: review of Brucella-host interactions. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1505-17. [PMID: 25892682 DOI: 10.1016/j.ajpath.2015.03.003] [Citation(s) in RCA: 310] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 02/10/2015] [Accepted: 03/02/2015] [Indexed: 01/18/2023]
Abstract
This review of Brucella-host interactions and immunobiology discusses recent discoveries as the basis for pathogenesis-informed rationales to prevent or treat brucellosis. Brucella spp., as animal pathogens, cause human brucellosis, a zoonosis that results in worldwide economic losses, human morbidity, and poverty. Although Brucella spp. infect humans as an incidental host, 500,000 new human infections occur annually, and no patient-friendly treatments or approved human vaccines are reported. Brucellae display strong tissue tropism for lymphoreticular and reproductive systems with an intracellular lifestyle that limits exposure to innate and adaptive immune responses, sequesters the organism from the effects of antibiotics, and drives clinical disease manifestations and pathology. Stealthy brucellae exploit strategies to establish infection, including i) evasion of intracellular destruction by restricting fusion of type IV secretion system-dependent Brucella-containing vacuoles with lysosomal compartments, ii) inhibition of apoptosis of infected mononuclear cells, and iii) prevention of dendritic cell maturation, antigen presentation, and activation of naive T cells, pathogenesis lessons that may be informative for other intracellular pathogens. Data sets of next-generation sequences of Brucella and host time-series global expression fused with proteomics and metabolomics data from in vitro and in vivo experiments now inform interactive cellular pathways and gene regulatory networks enabling full-scale systems biology analysis. The newly identified effector proteins of Brucella may represent targets for improved, safer brucellosis vaccines and therapeutics.
Collapse
Affiliation(s)
- Paul de Figueiredo
- Department of Veterinary Pathobiology, Texas A&M University and Texas AgriLife Research, College Station, Texas; Norman Borlaug Center, Texas A&M University, College Station, Texas; Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, Texas
| | - Thomas A Ficht
- Department of Veterinary Pathobiology, Texas A&M University and Texas AgriLife Research, College Station, Texas
| | - Allison Rice-Ficht
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - Carlos A Rossetti
- Institute of Pathobiology, CICVyA-CNIA, National Institute of Animal Agriculture Technology (INTA), Buenos Aires, Argentina
| | - L Garry Adams
- Department of Veterinary Pathobiology, Texas A&M University and Texas AgriLife Research, College Station, Texas.
| |
Collapse
|
25
|
Ben J, Zhu X, Zhang H, Chen Q. Class A1 scavenger receptors in cardiovascular diseases. Br J Pharmacol 2015; 172:5523-30. [PMID: 25651870 DOI: 10.1111/bph.13105] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 01/15/2015] [Accepted: 02/02/2015] [Indexed: 01/03/2023] Open
Abstract
Class A1 scavenger receptors (SR-A1) are membrane glycoproteins that can form homotrimers. This receptor was originally defined by its ability to mediate the accumulation of lipids in macrophages. Subsequent studies reveal that SR-A1 plays critical roles in innate immunity, cell apoptosis and proliferation. This review highlights recent advances in understanding the structure, receptor pathway and regulation of SR-A1. Although its role in atherosclerosis is disputable, recent discoveries suggest that SR-A1 function in anti-inflammatory responses by promoting an M2 macrophage phenotype in cardiovascular diseases. Therefore, SR-A1 may be a potential target for therapeutic intervention of cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjing Ben
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, 210029, China
| | - Xudong Zhu
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, 210029, China
| | - Hanwen Zhang
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, 210029, China
| | - Qi Chen
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
26
|
Vadali S, Post SR. Lipid rafts couple class A scavenger receptors to phospholipase A2 activation during macrophage adhesion. J Leukoc Biol 2014; 96:873-81. [PMID: 25070949 DOI: 10.1189/jlb.2a0414-214r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
SR-A mediated macrophage adhesion to modified ECM proteins in a process that involves physical attachment of SR-A to modified ECM and activation of Lyn-PI3K and PLA2-12/15-lipoxygenase signaling pathways. Structurally, SR-A-mediated cell adhesion requires a 6-aa membrane-proximal cytoplasmic motif. However, the mechanism that couples SR-A-mediated adhesion to activation of these distinct signaling pathways is not known. For other adhesion receptors, including integrins, localization in cholesterol-rich LRs is an important mechanism for coupling the receptor with the activation of specific signaling pathways. We hypothesized that SR-A-mediated macrophage adhesion might also involve LRs. Our results demonstrate that SR-A is enriched in LRs in HEK cells that heterologously express SR-A and in macrophages that endogenously expressed the receptor. We further show that a truncated SR-A construct (SR-A(Δ1-49)), which mediates cell adhesion but not ligand internalization, is also enriched in LRs, suggesting an association between LRs and SR-A-dependent cell adhesion. To examine this association more directly, we used the cholesterol chelator MβCD to deplete cholesterol and disrupt LR function. We found that cholesterol depletion significantly decreased SR-A-mediated macrophage adhesion. We further show that decreased SR-A-dependent macrophage adhesion following cholesterol depletion results from the inhibition of PLA2 but not PI3K activation. Overall, our results demonstrate an important role for LRs in selectively coupling SR-A with PLA2 activation during macrophage adhesion.
Collapse
Affiliation(s)
| | - Steven R Post
- Departments of Pharmacology and Toxicology and Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
27
|
Affiliation(s)
- Jean-Pierre Gorvel
- Centre d'Immunologie de Marseille-Luminy; Aix-Marseille University; Marseille, France; CNRS UMR7280; Marseille, France; INSERM U1104; Marseille, France
| |
Collapse
|
28
|
Mechanism of Asp24 upregulation in Brucella abortus rough mutant with a disrupted O-antigen export system and effect of Asp24 in bacterial intracellular survival. Infect Immun 2014; 82:2840-50. [PMID: 24752516 DOI: 10.1128/iai.01765-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We previously showed that Brucella abortus rough mutant strain 2308 ΔATP (called the ΔrfbE mutant in this study) exhibits reduced intracellular survival in RAW264.7 cells and attenuated persistence in BALB/c mice. In this study, we performed microarray analysis to detect genes with differential expression between the ΔrfbE mutant and wild-type strain S2308. Interestingly, acid shock protein 24 gene (asp24) expression was significantly upregulated in the ΔrfbE mutant compared to S2308, as confirmed by quantitative reverse transcription-PCR (qRT-PCR) and Western blotting. Further studies using additional strains indicated that the upregulation of asp24 occurred only in rough mutants with disrupted O-antigen export system components, including the ATP-binding protein gene rfbE (bab1_0542) and the permease gene rfbD (bab1_0543), while the ΔwboA rough mutant (which lacks an O-antigen synthesis-related glycosyltransferase) and the RB51 strain (a vaccine strain with the rough phenotype) showed no significant changes in asp24 expression compared to S2308. In addition, abolishing the intracellular O-antigen synthesis of the ΔrfbE mutant by deleting the wboA gene (thereby creating the ΔrfbE ΔwboA double-knockout strain) recovered asp24 expression. These results indicated that asp24 upregulation is associated with intracellular O-antigen synthesis and accumulation but not with the bacterial rough phenotype. Further studies indicated that asp24 upregulation in the ΔrfbE mutant was associated neither with bacterial adherence and invasion nor with cellular necrosis on RAW264.7 macrophages. However, proper expression of the asp24 gene favors intracellular survival of Brucella in RAW264.7 cells and HeLa cells during an infection. This study reveals a novel mechanism for asp24 upregulation in B. abortus mutants.
Collapse
|
29
|
Proteomic analysis of detergent resistant membrane domains during early interaction of macrophages with rough and smooth Brucella melitensis. PLoS One 2014; 9:e91706. [PMID: 24643124 PMCID: PMC3958395 DOI: 10.1371/journal.pone.0091706] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 02/13/2014] [Indexed: 12/20/2022] Open
Abstract
The plasma membrane contains discrete nanometer-sized domains that are resistant to non-ionic detergents, and which are called detergent resistant membrane domains (DRMDs) or lipid rafts. Exposure of host cells to pathogenic bacteria has been shown to induce the re-distribution of specific host proteins between DRMDs and detergent soluble membranes, which leads to the initiation of cell signaling that enable pathogens to access host cells. DRMDs have been shown to play a role in the invasion of Brucella into host macrophages and the formation of replicative phagosomes called Brucella-containing vacuoles (BCVs). In this study we sought to characterize changes to the protein expression profiles in DRMDs and to respective cellular pathways and networks of Mono Mac 6 cells in response to the adherence of rough VTRM1 and smooth 16 M B. melitensis strains. DRMDs were extracted from Mono Mac 6 cells exposed for 2 minutes at 4°C to Brucella (no infection occurs) and from unexposed control cells. Protein expression was determined using the non-gel based quantitative iTRAQ (Isobaric Tags for Relative and Absolute Quantitation) mass spectrometry technique. Using the identified iTRAQ proteins we performed enrichment analyses and probed constructed human biochemical networks for interactions and metabolic reactions. We identified 149 proteins, which either became enriched, depleted or whose amounts did not change in DRMDs upon Brucella exposure. Several of these proteins were distinctly enriched or depleted in DRMDs upon exposure to rough and smooth B. melitensis strains which results in the differential engagement of cellular pathways and networks immediately upon Brucella encounter. For some of the proteins such as myosin 9, small G protein signaling modulator 3, lysine-specific demethylase 5D, erlin-2, and voltage-dependent anion-selective channel protein 2, we observed extreme differential depletion or enrichment in DRMDs. The identified proteins and pathways could provide the basis for novel ways of treating or diagnosing Brucellosis.
Collapse
|
30
|
Larsen AK, Nymo IH, Briquemont B, Sørensen KK, Godfroid J. Entrance and survival of Brucella pinnipedialis hooded seal strain in human macrophages and epithelial cells. PLoS One 2013; 8:e84861. [PMID: 24376851 PMCID: PMC3869908 DOI: 10.1371/journal.pone.0084861] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 11/19/2013] [Indexed: 11/19/2022] Open
Abstract
Marine mammal Brucella spp. have been isolated from pinnipeds (B. pinnipedialis) and cetaceans (B. ceti) from around the world. Although the zoonotic potential of marine mammal brucellae is largely unknown, reports of human disease exist. There are few studies of the mechanisms of bacterial intracellular invasion and multiplication involving the marine mammal Brucella spp. We examined the infective capacity of two genetically different B. pinnipedialis strains (reference strain; NTCT 12890 and a hooded seal isolate; B17) by measuring the ability of the bacteria to enter and replicate in cultured phagocytes and epithelial cells. Human macrophage-like cells (THP-1), two murine macrophage cell lines (RAW264.7 and J774A.1), and a human malignant epithelial cell line (HeLa S3) were challenged with bacteria in a gentamicin protection assay. Our results show that B. pinnipedialis is internalized, but is then gradually eliminated during the next 72 – 96 hours. Confocal microscopy revealed that intracellular B. pinnipedialis hooded seal strain colocalized with lysosomal compartments at 1.5 and 24 hours after infection. Intracellular presence of B. pinnipedialis hooded seal strain was verified by transmission electron microscopy. By using a cholesterol-scavenging lipid inhibitor, entrance of B. pinnipedialis hooded seal strain in human macrophages was significantly reduced by 65.8 % (± 17.3), suggesting involvement of lipid-rafts in intracellular entry. Murine macrophages invaded by B. pinnipedialis do not release nitric oxide (NO) and intracellular bacterial presence does not induce cell death. In summary, B. pinnipedialis hooded seal strain can enter human and murine macrophages, as well as human epithelial cells. Intracellular entry of B. pinnipedialis hooded seal strain involves, but seems not to be limited to, lipid-rafts in human macrophages. Brucella pinnipedialis does not multiply or survive for prolonged periods intracellulary.
Collapse
Affiliation(s)
- Anett K. Larsen
- Section for Arctic Veterinary Medicine, Department of Food Safety and Infection Biology, Norwegian School of Veterinary Science, Tromsø, Norway
- The Fram Centre, High North Research Centre for Climate and the Environment, Tromsø, Norway
- * E-mail:
| | - Ingebjørg H. Nymo
- Section for Arctic Veterinary Medicine, Department of Food Safety and Infection Biology, Norwegian School of Veterinary Science, Tromsø, Norway
- The Fram Centre, High North Research Centre for Climate and the Environment, Tromsø, Norway
| | - Benjamin Briquemont
- Faculty of Science, Catholic University of Louvain, Louvain-la-Neuve, Belgium
| | - Karen K. Sørensen
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Jacques Godfroid
- Section for Arctic Veterinary Medicine, Department of Food Safety and Infection Biology, Norwegian School of Veterinary Science, Tromsø, Norway
- The Fram Centre, High North Research Centre for Climate and the Environment, Tromsø, Norway
| |
Collapse
|
31
|
Smith JA, Khan M, Magnani DD, Harms JS, Durward M, Radhakrishnan GK, Liu YP, Splitter GA. Brucella induces an unfolded protein response via TcpB that supports intracellular replication in macrophages. PLoS Pathog 2013; 9:e1003785. [PMID: 24339776 PMCID: PMC3855547 DOI: 10.1371/journal.ppat.1003785] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 10/08/2013] [Indexed: 01/18/2023] Open
Abstract
Brucella melitensis is a facultative intracellular bacterium that causes brucellosis, the most prevalent zoonosis worldwide. The Brucella intracellular replicative niche in macrophages and dendritic cells thwarts immune surveillance and complicates both therapy and vaccine development. Currently, host-pathogen interactions supporting Brucella replication are poorly understood. Brucella fuses with the endoplasmic reticulum (ER) to replicate, resulting in dramatic restructuring of the ER. This ER disruption raises the possibility that Brucella provokes an ER stress response called the Unfolded Protein Response (UPR). In this study, B. melitensis infection up regulated expression of the UPR target genes BiP, CHOP, and ERdj4, and induced XBP1 mRNA splicing in murine macrophages. These data implicate activation of all 3 major signaling pathways of the UPR. Consistent with previous reports, XBP1 mRNA splicing was largely MyD88-dependent. However, up regulation of CHOP, and ERdj4 was completely MyD88 independent. Heat killed Brucella stimulated significantly less BiP, CHOP, and ERdj4 expression, but induced XBP1 splicing. Although a Brucella VirB mutant showed relatively intact UPR induction, a TcpB mutant had significantly compromised BiP, CHOP and ERdj4 expression. Purified TcpB, a protein recently identified to modulate microtubules in a manner similar to paclitaxel, also induced UPR target gene expression and resulted in dramatic restructuring of the ER. In contrast, infection with the TcpB mutant resulted in much less ER structural disruption. Finally, tauroursodeoxycholic acid, a pharmacologic chaperone that ameliorates the UPR, significantly impaired Brucella replication in macrophages. Together, these results suggest Brucella induces a UPR, via TcpB and potentially other factors, that enables its intracellular replication. Thus, the UPR may provide a novel therapeutic target for the treatment of brucellosis. These results also have implications for other intracellular bacteria that rely on host physiologic stress responses for replication.
Collapse
Affiliation(s)
- Judith A. Smith
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Mike Khan
- Cellular and Molecular Pathology Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Diogo D. Magnani
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States of America
| | - Jerome S. Harms
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States of America
| | - Marina Durward
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States of America
| | | | - Yi-Ping Liu
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Gary A. Splitter
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States of America
| |
Collapse
|
32
|
Lee JJ, Kim DG, Kim DH, Simborio HL, Min W, Lee HJ, Her M, Jung SC, Watarai M, Kim S. Interplay between clathrin and Rab5 controls the early phagocytic trafficking and intracellular survival of Brucella abortus within HeLa cells. J Biol Chem 2013; 288:28049-57. [PMID: 23940042 DOI: 10.1074/jbc.m113.491555] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lipid raft-associated clathrin is essential for host-pathogen interactions during infection. Brucella abortus is an intracellular pathogen that circumvents host defenses, but little is known about the precise infection mechanisms that involve interaction with lipid raft-associated mediators. The aim of this study was to elucidate the clathrin-mediated phagocytic mechanisms of B. abortus. The clathrin dependence of B. abortus infection in HeLa cells was investigated using an infection assay and immunofluorescence microscopy. The redistribution of clathrin in the membrane and in phagosomes was investigated using sucrose gradient fractionation of lipid rafts and the isolation of B. abortus-containing vacuoles, respectively. Clathrin and dynamin were concentrated into lipid rafts during B. abortus infection, and the entry and intracellular survival of B. abortus within HeLa cells were abrogated by clathrin inhibition. Clathrin disruption decreased actin polymerization and the colocalization of B. abortus-containing vacuoles with clathrin and Rab5 but not lysosome-associated membrane protein 1 (LAMP-1). Thus, our data demonstrate that clathrin plays a fundamental role in the entry and intracellular survival of B. abortus via interaction with lipid rafts and actin rearrangement. This process facilitates the early intracellular trafficking of B. abortus to safe replicative vacuoles.
Collapse
Affiliation(s)
- Jin Ju Lee
- From the College of Veterinary Medicine, Gyeongsang National University, Jinju 660-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Microarray-based identification of differentially expressed genes in intracellular Brucella abortus within RAW264.7 cells. PLoS One 2013; 8:e67014. [PMID: 23950864 PMCID: PMC3737221 DOI: 10.1371/journal.pone.0067014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 05/14/2013] [Indexed: 11/20/2022] Open
Abstract
Brucella spp. is a species of facultative intracellular Gram-negative bacteria that induces abortion and causes sterility in domesticated mammals and chronic undulant fever in humans. Important determinants of Brucella’s virulence and potential for chronic infection include the ability to circumvent the host cell’s internal surveillance system and the capability to proliferate within dedicated and non-dedicated phagocytes. Hence, identifying genes necessary for intracellular survival may hold the key to understanding Brucella infection. In the present study, microarray analysis reveals that 7.82% (244/3334) of all Brucella abortus genes were up-regulated and 5.4% (180/3334) were down-regulated in RAW264.7 cells, compared to free-living cells in TSB. qRT-PCR verification further confirmed a >5-fold up-regulation for fourteen genes. Functional analysis classified araC, ddp, and eryD as to partake in information storage and processing, alp, flgF and virB9 to be involved in cellular processes, hpcd and aldh to play a role in metabolism, mfs and nikC to be involved in both cellular processes and metabolism, and four hypothetical genes (bruAb1_1814, bruAb1_0475, bruAb1_1926, and bruAb1_0292) had unknown functions. Furthermore, we constructed a B. abortus 2308 mutant Δddp where the ddp gene is deleted in order to evaluate the role of ddp in intracellular survival. Infection assay indicated significantly higher adherence and invasion abilities of the Δddp mutant, however it does not survive well in RAW264.7 cells. Brucella may survive in hostile intracellular environment by modulating gene expression.
Collapse
|
34
|
Ben-Tekaya H, Gorvel JP, Dehio C. Bartonella and Brucella--weapons and strategies for stealth attack. Cold Spring Harb Perspect Med 2013; 3:3/8/a010231. [PMID: 23906880 DOI: 10.1101/cshperspect.a010231] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Bartonella spp. and Brucella spp. are closely related α-proteobacterial pathogens that by distinct stealth-attack strategies cause chronic infections in mammals including humans. Human infections manifest by a broad spectrum of clinical symptoms, ranging from mild to fatal disease. Both pathogens establish intracellular replication niches and subvert diverse pathways of the host's immune system. Several virulence factors allow them to adhere to, invade, proliferate, and persist within various host-cell types. In particular, type IV secretion systems (T4SS) represent essential virulence factors that transfer effector proteins tailored to recruit host components and modulate cellular processes to the benefit of the bacterial intruders. This article puts the remarkable features of these two pathogens into perspective, highlighting the mechanisms they use to hijack signaling and trafficking pathways of the host as the basis for their stealthy infection strategies.
Collapse
Affiliation(s)
- Houchaima Ben-Tekaya
- Focal Area Infection Biology, Biozentrum, University of Basel, 4052 Basel, Switzerland
| | | | | |
Collapse
|
35
|
Ciesielski F, Griffin DC, Rittig M, Moriyón I, Bonev BB. Interactions of lipopolysaccharide with lipid membranes, raft models — A solid state NMR study. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:1731-42. [DOI: 10.1016/j.bbamem.2013.03.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/08/2013] [Accepted: 03/28/2013] [Indexed: 01/09/2023]
|
36
|
Global Rsh-dependent transcription profile of Brucella suis during stringent response unravels adaptation to nutrient starvation and cross-talk with other stress responses. BMC Genomics 2013; 14:459. [PMID: 23834488 PMCID: PMC3710219 DOI: 10.1186/1471-2164-14-459] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/18/2013] [Indexed: 01/08/2023] Open
Abstract
Background In the intracellular pathogen Brucella spp., the activation of the stringent response, a global regulatory network providing rapid adaptation to growth-affecting stress conditions such as nutrient deficiency, is essential for replication in the host. A single, bi-functional enzyme Rsh catalyzes synthesis and hydrolysis of the alarmone (p)ppGpp, responsible for differential gene expression under stringent conditions. Results cDNA microarray analysis allowed characterization of the transcriptional profiles of the B. suis 1330 wild-type and Δrsh mutant in a minimal medium, partially mimicking the nutrient-poor intramacrophagic environment. A total of 379 genes (11.6% of the genome) were differentially expressed in a rsh-dependent manner, of which 198 were up-, and 181 were down-regulated. The pleiotropic character of the response was confirmed, as the genes encoded an important number of transcriptional regulators, cell envelope proteins, stress factors, transport systems, and energy metabolism proteins. Virulence genes such as narG and sodC, respectively encoding respiratory nitrate reductase and superoxide dismutase, were under the positive control of (p)ppGpp, as well as expression of the cbb3-type cytochrome c oxidase, essential for chronic murine infection. Methionine was the only amino acid whose biosynthesis was absolutely dependent on stringent response in B. suis. Conclusions The study illustrated the complexity of the processes involved in adaptation to nutrient starvation, and contributed to a better understanding of the correlation between stringent response and Brucella virulence. Most interestingly, it clearly indicated (p)ppGpp-dependent cross-talk between at least three stress responses playing a central role in Brucella adaptation to the host: nutrient, oxidative, and low-oxygen stress.
Collapse
|
37
|
Chakraborty M, Lou C, Huan C, Kuo MS, Park TS, Cao G, Jiang XC. Myeloid cell-specific serine palmitoyltransferase subunit 2 haploinsufficiency reduces murine atherosclerosis. J Clin Invest 2013; 123:1784-97. [PMID: 23549085 DOI: 10.1172/jci60415] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/24/2013] [Indexed: 12/15/2022] Open
Abstract
Serine palmitoyltransferase (SPT) is the first and rate-limiting enzyme of the de novo biosynthetic pathway of sphingomyelin (SM). Both SPT and SM have been implicated in the pathogenesis of atherosclerosis, the development of which is driven by macrophages; however, the role of SPT in macrophage-mediated atherogenesis is unknown. To address this issue, we have analyzed macrophage inflammatory responses and reverse cholesterol transport, 2 key mediators of atherogenesis, in SPT subunit 2-haploinsufficient (Sptlc2(+/-)) macrophages. We found that Sptlc2(+/-) macrophages have significantly lower SM levels in plasma membrane and lipid rafts. This reduction not only impaired inflammatory responses triggered by TLR4 and its downstream NF-κB and MAPK pathways, but also enhanced reverse cholesterol transport mediated by ABC transporters. LDL receptor-deficient (Ldlr(-/-)) mice transplanted with Sptlc2(+/-) bone marrow cells exhibited significantly fewer atherosclerotic lesions after high-fat and high-cholesterol diet feeding. Additionally, Ldlr(-/-) mice with myeloid cell-specific Sptlc2 haploinsufficiency exhibited significantly less atherosclerosis than controls. These findings suggest that SPT could be a novel therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Mahua Chakraborty
- Department of Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Redundant effects of ketamine on the pathogenesis and severity of Brucella abortus infection. Comp Immunol Microbiol Infect Dis 2013; 36:71-81. [DOI: 10.1016/j.cimid.2012.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 09/13/2012] [Accepted: 10/03/2012] [Indexed: 01/18/2023]
|
39
|
Abstract
Atherosclerosis is the major cause of mortality in the developed countries. Although presently known risk factors have some predictive value for the disease, a major part of the variability in this process remains unexplained. It is extremely important to find new approaches for better understanding of the disease and for treating it. Exploration of the sphingolipid metabolism is one of these approaches. Sphingolipids are a large class of lipids with structural and signaling functions. Recent researches indicated that these lipids play important roles in the development of atherosclerosis. In this chapter, we summarized the major findings in the field.
Collapse
Affiliation(s)
- Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA.
| | | |
Collapse
|
40
|
Drummond R, Cauvi DM, Hawisher D, Song D, Niño DF, Coimbra R, Bickler S, De Maio A. Deletion of scavenger receptor A gene in mice resulted in protection from septic shock and modulation of TLR4 signaling in isolated peritoneal macrophages. Innate Immun 2012; 19:30-41. [PMID: 22751446 DOI: 10.1177/1753425912449548] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Scavenger receptor A (Sra), also known as macrophage scavenger receptor 1 (Msr1), is a surface glycoprotein preferentially present in macrophages that plays a primary role in innate immunity. Previous studies have shown that Sra is a modifier gene for the response to bacterial LPS in mice at the level of IL-10 production, in particular. In the present study, we found that Sra(-/-) mice are more resistant to septic shock induced by cecal ligation and puncture than wild-type C57BL/6 J (B6) mice. In addition, Sra(-/-) mice displayed initial elevated high density lipoprotein (HDL) circulating levels. Naïve peritoneal macrophages (PMs) were isolated from Sra(-/-) mice to understand the possible protective mechanism. Incubation of these cells with LPS was found to modulate TLR4 signaling, leading to a reduction in IL-10 and IL-6 mRNA levels, but not TNF-α expression, at low concentrations of LPS in comparison with PMs isolated from B6 mice. No differences were found in LPS binding between PMs derived from Sra(-/-) or B6 mice. The lack of Sra binding to LPS was confirmed after transfection of Chinese hamster ovary (CHO) cells with the Sra gene. The contribution of Sra to the outcome of sepsis may be a combination of changes in TLR4 signaling pathway and elevated levels of HDL in circulation, but also LPS toxicity.
Collapse
Affiliation(s)
- Robert Drummond
- MSTP program Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Iwabuchi K, Nakayama H, Masuda H, Kina K, Ogawa H, Takamori K. Membrane microdomains in immunity: glycosphingolipid-enriched domain-mediated innate immune responses. Biofactors 2012; 38:275-83. [PMID: 22488955 DOI: 10.1002/biof.1017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 03/14/2012] [Indexed: 11/06/2022]
Abstract
Over the last 30 years, many studies have indicated that glycosphingolipids (GSLs) expressed on the cell surface may act as binding sites for microorganisms. Based on their physicochemical characteristics, GSLs form membrane microdomains with cholesterol, sphingomyelin, glycosylphosphatidylinositol (GPI)-anchored proteins, and various signaling molecules, and GSL-enriched domains have been shown to be involved in these defense responses. Among the GSLs, lactosylceramide (LacCer, CDw17) can bind to various microorganisms. LacCer is expressed at high levels on the plasma membrane of human neutrophils, and forms membrane microdomains associated with the Src family tyrosine kinase Lyn. LacCer-enriched membrane microdomains mediate superoxide generation, chemotaxis, and non-opsonic phagocytosis. Therefore, LacCer-enriched membrane microdomains are thought to function as pattern recognition receptors (PRRs) to recognize pathogen-associated molecular patterns (PAMPs) expressed on microorganisms. In contrast, several pathogens have developed infection mechanisms using membrane microdomains. In addition, some pathogens have the ability to avoid degradation by escaping from the vacuolar compartment or preventing phagosome maturation, utilizing membrane microdomains, such as LacCer-enriched domains, of host cells. The detailed molecular mechanisms of these membrane microdomain-associated host-pathogen interactions remain to be elucidated.
Collapse
Affiliation(s)
- Kazuhisa Iwabuchi
- Institute for Environmental and Gender-specific Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
42
|
Ciesielski F, Davis B, Rittig M, Bonev BB, O'Shea P. Receptor-independent interaction of bacterial lipopolysaccharide with lipid and lymphocyte membranes; the role of cholesterol. PLoS One 2012; 7:e38677. [PMID: 22685597 PMCID: PMC3369841 DOI: 10.1371/journal.pone.0038677] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 05/10/2012] [Indexed: 01/18/2023] Open
Abstract
Lipopolysaccharide (LPS) is a major constituent of bacterial outer membranes where it makes up the bulk of the outer leaflet and plays a key role as determinant of bacterial interactions with the host. Membrane-free LPS is known to activate T-lymphocytes through interactions with Toll-like receptor 4 via multiprotein complexes. In the present study, we investigate the role of cholesterol and membrane heterogeneities as facilitators of receptor-independent LPS binding and insertion, which underpin bacterial interactions with the host in symbiosis, pathogenesis and cell invasion. We use fluorescence spectroscopy to investigate the interactions of membrane-free LPS from intestinal Gram-negative organisms with cholesterol-containing model membranes and with T-lymphocytes. LPS preparations from Klebsiella pneumoniae and Salmonella enterica were found to bind preferentially to mixed lipid membranes by comparison to pure PC bilayers. The same was observed for LPS from the symbiote Escherichia coli but with an order of magnitude higher dissociation constant. Insertion of LPS into model membranes confirmed the preference for sphimgomyelin/cholesterol-containing systems. LPS insertion into Jurkat T-lymphocyte membranes reveals that they have a significantly greater LPS-binding capacity by comparison to methyl-β-cyclodextrin cholesterol-depleted lymphocyte membranes, albeit at slightly lower binding rates.
Collapse
Affiliation(s)
- Filip Ciesielski
- School of Biomedical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Benjamin Davis
- School of Biology, University of Nottingham, Nottingham, United Kingdom
| | - Michael Rittig
- School of Biomedical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Boyan B. Bonev
- School of Biomedical Sciences, University of Nottingham, Nottingham, United Kingdom
- * E-mail: (BBB); (PS)
| | - Paul O'Shea
- School of Biology, University of Nottingham, Nottingham, United Kingdom
- * E-mail: (BBB); (PS)
| |
Collapse
|
43
|
Garcia-Garcia E, Grayfer L, Stafford JL, Belosevic M. Evidence for the presence of functional lipid rafts in immune cells of ectothermic organisms. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2012; 37:257-269. [PMID: 22450166 DOI: 10.1016/j.dci.2012.03.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 03/12/2012] [Accepted: 03/15/2012] [Indexed: 05/31/2023]
Abstract
The role of lipid rafts in non-mammalian leukocytes has been scarcely investigated. We performed biochemical and functional analysis of lipid rafts in fish leukocytes. Fish Flotillin-1 and a fish GM1-like molecule (fGM1-L) were found in low density detergent-resistant membranes (LD-DRM) in goldfish macrophages and catfish B lymphocytes, similarly to mammals. The presence of flotillin-1 and fGM1-L in LD-DRM was sensitive to increased detergent concentrations, and cholesterol extraction. Confocal microscopy analysis of flotillin-1 and fGM1-L in fish leukocytes showed a distinctive punctuated staining pattern, suggestive of pre-existing rafts. Confocal microscopy analysis of macrophages showed that the membrane of phagosomes containing serum-opsonized zymosan was enriched in fGM1-L, and zymosan phagocytosis was reduced after cholesterol extraction. The presence of flotillin-1 and fGM1-L in LD-DRM, the microscopic evidence of flotillin-1 and fGM1-L on fish macrophages and B-cells, and the sensitivity of phagocytosis to cholesterol extraction, indicate that lipid rafts are biochemically and functionally similar in leukocytes from fish and mammals.
Collapse
Affiliation(s)
- Erick Garcia-Garcia
- Department of Biological Sciences, University of Alberta, Edmonton, Canada AB T6G 2E9
| | | | | | | |
Collapse
|
44
|
Drummond R, Song D, Hawisher D, Wolf PL, Vazquez DE, Nino D, Coimbra R, Cauvi DM, De Maio A. Acute acalculous cholecystitis-like phenotype in scavenger receptor A knock-out mice. J Surg Res 2012; 174:344-51. [PMID: 21474146 PMCID: PMC3173564 DOI: 10.1016/j.jss.2010.12.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 12/06/2010] [Accepted: 12/22/2010] [Indexed: 12/22/2022]
Abstract
BACKGROUND Sepsis is a major health problem in the United States that affects more than three-quarters of a million people every year. Previous studies have shown that scavenger receptor A (Sra), also known as macrophage scavenger receptor 1 (Msr1), is a modifier of interleukin 10 (IL-10) expression after injection of bacterial lipopolysaccharide (LPS). Therefore, we investigated the response to sepsis in Sra knock out mice. MATERIALS AND METHODS C57BL/6J (B6) (n = 88) and Sra (-/-) mice (n = 88) were subjected to cecal ligation and puncture (CLP) using 18G or 16G needles, sham operation, or non-operated controls. At the end, mice were autopsied for the determination of abnormalities after the procedure. Cytokine gene expression was examined in lung and liver samples by quantitative RT-PCR (qRT-PCR), and circulating cholesterol levels were also measured. RESULTS Sra (-/-) mice displayed an enlargement of the gallbladder after CLP that was not detected in sham or non-operated mice or in B6 mice (wild-type) after CLP. The enlarged gallbladder resembles a condition of acute acalculous cholecystitis observed in humans. Sra (-/-) mice presented high cholesterol levels in circulation as opposed to wild type B6 mice. Moreover, Sra (-/-) mice exhibited a reduction in IL-10 mRNA levels in lungs compared to wild-type B6 mice after CLP. CONCLUSIONS The development of acute acalculous cholecystitis may be the combination of pre-existing conditions, such as hypercholesterolemia associated with a defect in Sra (Msr1) and a robust inflammation induced by sepsis.
Collapse
Affiliation(s)
- Robert Drummond
- Department of Surgery, University of California San Diego, La Jolla, California
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Donghuan Song
- Department of Surgery, University of California San Diego, La Jolla, California
| | - Dennis Hawisher
- Department of Surgery, University of California San Diego, La Jolla, California
| | - Paul L. Wolf
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Daniel E. Vazquez
- Department of Surgery, University of California San Diego, La Jolla, California
| | - Diego Nino
- Department of Surgery, University of California San Diego, La Jolla, California
| | - Raul Coimbra
- Department of Surgery, University of California San Diego, La Jolla, California
| | - David M. Cauvi
- Department of Surgery, University of California San Diego, La Jolla, California
| | - Antonio De Maio
- Department of Surgery, University of California San Diego, La Jolla, California
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
45
|
von Bargen K, Gorvel JP, Salcedo SP. Internal affairs: investigating the Brucella intracellular lifestyle. FEMS Microbiol Rev 2012; 36:533-62. [PMID: 22373010 DOI: 10.1111/j.1574-6976.2012.00334.x] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 01/10/2012] [Accepted: 02/16/2012] [Indexed: 01/18/2023] Open
Abstract
Bacteria of the genus Brucella are Gram-negative pathogens of several animal species that cause a zoonotic disease in humans known as brucellosis or Malta fever. Within their hosts, brucellae reside within different cell types where they establish a replicative niche and remain protected from the immune response. The aim of this article is to discuss recent advances in the field in the specific context of the Brucella intracellular 'lifestyle'. We initially discuss the different host cell targets and their relevance during infection. As it represents the key to intracellular replication, the focus is then set on the maturation of the Brucella phagosome, with particular emphasis on the Brucella factors that are directly implicated in intracellular trafficking and modulation of host cell signalling pathways. Recent data on the role of the type IV secretion system are discussed, novel effector molecules identified and how some of them impact on trafficking events. Current knowledge on Brucella gene regulation and control of host cell death are summarized, as they directly affect intracellular persistence. Understanding how Brucella molecules interplay with their host cell targets to modulate cellular functions and establish the intracellular niche will help unravel how this pathogen causes disease.
Collapse
Affiliation(s)
- Kristine von Bargen
- Faculté de Sciences de Luminy, Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, UM 2, Marseille Cedex, France
| | | | | |
Collapse
|
46
|
Honda H, Nagai Y, Matsunaga T, Saitoh SI, Akashi-Takamura S, Hayashi H, Fujii I, Miyake K, Muraguchi A, Takatsu K. Glycyrrhizin and isoliquiritigenin suppress the LPS sensor toll-like receptor 4/MD-2 complex signaling in a different manner. J Leukoc Biol 2012; 91:967-76. [PMID: 22422925 DOI: 10.1189/jlb.0112038] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Recent evidences suggest that the extracts of plant products are able to modulate innate immune responses. A saponin GL and a chalcone ILG are representative components of Glycyrrhiza uralensis, which attenuate inflammatory responses mediated by TLRs. Here, we show that GL and ILG suppress different steps of the LPS sensor TLR4/MD-2 complex signaling at the receptor level. Extract of G. uralensis suppressed IL-6 and TNF-α production induced by lipid A moiety of LPS in RAW264.7 cells. Among various G. uralensis-related components of saponins and flavanones/chalcones, GL and ILG could suppress IL-6 production induced by lipid A in dose-dependent manners in RAW264.7 cells. Furthermore, elevation of plasma TNF-α in LPS-injected mice was attenuated by passive administration of GL or ILG. GL and ILG inhibited lipid A-induced NF-κB activation in Ba/F3 cells expressing TLR4/MD-2 and CD14 and BMMs. These components also inhibited activation of MAPKs, including JNK, p38, and ERK in BMMs. In addition, GL and ILG inhibited NF-κB activation and IL-6 production induced by paclitaxel, a nonbacterial TLR4 ligand. Interestingly, GL attenuated the formation of the LPS-TLR4/MD-2 complexes, resulting in inhibition of homodimerization of TLR4. Although ILG did not affect LPS binding to TLR4/MD-2, it could inhibit LPS-induced TLR4 homodimerization. These results imply that GL and ILG modulate the TLR4/MD-2 complex at the receptor level, leading to suppress LPS-induced activation of signaling cascades and cytokine production, but their effects are exerted at different steps of TLR4/MD-2 signaling.
Collapse
Affiliation(s)
- Hiroe Honda
- Toyama Prefectural Institute for Pharmaceutical Research, Toyama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
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
|
48
|
Lee JJ, Bae JH, Kim DH, Lim JJ, Kim DG, Lee HJ, Min W, Rhee MH, Chang HH, Park H, Kim S. Intracellular replication inhibitory effects of Galla Rhois ethanol extract for Brucella abortus infection. JOURNAL OF ETHNOPHARMACOLOGY 2011; 138:602-609. [PMID: 22008879 DOI: 10.1016/j.jep.2011.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 09/23/2011] [Accepted: 10/03/2011] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Galla Rhois (GR) has long been applied in traditional Korean and Oriental medicine. Although GR has an anti-bacterial effect, the anti-bacterial mechanism and therapeutic efficiency of GR for intracellular parasitic Brucella infection are still unclear. AIM OF THE STUDY The objective of this study was to investigate the antibacterial and therapeutic effects of GR ethanol extract (GRE), which is a natural antibacterial component for the treatment of Brucella abortus infection. MATERIALS AND METHODS The antibacterial activity of GRE towards Brucella abortus was evaluated by incubating Brucella abortus with GRE. Following treatment with GRE, Brucella abortus adherence, uptake, intracellular growth, and intracellular trafficking in macrophages were monitored. Mice were infected intraperitoneally with Brucella abortus and treated orally with GRE for 14 days, and then the weight and CFUs from each spleen were monitored. RESULTS The viability of Brucella abortus was markedly decreased in a dose-dependent manner. Moreover, Brucella abortus internalization and intracellular growth within macrophages were reduced in GRE-treated cells. The number of bacteria that adhered to GRE-pretreated cells was significantly lower than that of untreated cells. With regards to intracellular trafficking, treatment with GRE augmented the colocalization of Brucella abortus-containing phagosomes with LAMP-1. GRE-treated mice showed considerably decreased weight and bacterial burdens in the spleen compared to untreated mice. CONCLUSION GRE exhibits antibacterial and protective effects on Brucella abortus in vitro and in vivo. These results highlight the beneficial effects of GRE in the prevention and treatment of brucellosis.
Collapse
Affiliation(s)
- Jin Ju Lee
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 660-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Cortes-Bratti X, Bassères E, Herrera-Rodriguez F, Botero-Kleiven S, Coppotelli G, Andersen JB, Masucci MG, Holmgren A, Chaves-Olarte E, Frisan T, Avila-Cariño J. Thioredoxin 80-activated-monocytes (TAMs) inhibit the replication of intracellular pathogens. PLoS One 2011; 6:e16960. [PMID: 21365006 PMCID: PMC3041819 DOI: 10.1371/journal.pone.0016960] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 01/09/2011] [Indexed: 12/18/2022] Open
Abstract
Background Thioredoxin 80 (Trx80) is an 80 amino acid natural cleavage product of Trx, produced primarily by monocytes. Trx80 induces differentiation of human monocytes into a novel cell type, named Trx80-activated-monocytes (TAMs). Principal Findings In this investigation we present evidence for a role of TAMs in the control of intracellular bacterial infections. As model pathogens we have chosen Listeria monocytogenes and Brucella abortus which replicate in the cytosol and the endoplasmic reticulum respectively. Our data indicate that TAMs efficiently inhibit intracellular growth of both L. monocytogenes and B. abortus. Further analysis shows that Trx80 activation prevents the escape of GFP-tagged L. monocytogenes into the cytosol, and induces accumulation of the bacteria within the lysosomes. Inhibition of the lysosomal activity by chloroquine treatment resulted in higher replication of bacteria in TAMs compared to that observed in control cells 24 h post-infection, indicating that TAMs kill bacteria by preventing their escape from the endosomal compartments, which progress into a highly degradative phagolysosome. Significance Our results show that Trx80 potentiates the bactericidal activities of professional phagocytes, and contributes to the first line of defense against intracellular bacteria.
Collapse
Affiliation(s)
- Ximena Cortes-Bratti
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Eugénie Bassères
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Fabiola Herrera-Rodriguez
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología Universidad de Costa Rica, San José, Costa Rica
| | | | - Giuseppe Coppotelli
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jens B. Andersen
- Department of Microbiology and Risk Assessment, National Food Institute, Soeborg, Denmark
| | - Maria G. Masucci
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Arne Holmgren
- Department of Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Esteban Chaves-Olarte
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología Universidad de Costa Rica, San José, Costa Rica
| | - Teresa Frisan
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (TF); (JA)
| | - Javier Avila-Cariño
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (TF); (JA)
| |
Collapse
|
50
|
Zhu XD, Zhuang Y, Ben JJ, Qian LL, Huang HP, Bai H, Sha JH, He ZG, Chen Q. Caveolae-dependent endocytosis is required for class A macrophage scavenger receptor-mediated apoptosis in macrophages. J Biol Chem 2011; 286:8231-8239. [PMID: 21205827 DOI: 10.1074/jbc.m110.145888] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SR-A (class A macrophage scavenger receptor) is a transmembrane receptor that can bind many different ligands, including modified lipoproteins that are relevant to the development of vascular diseases. However, the precise endocytic pathways of SR-A/mediated ligands internalization are not fully characterized. In this study, we show that the SR-A/ligand complex can be endocytosed by both clathrin- and caveolae-dependent pathways. Internalizations of SR-A-lipoprotein (such as acLDL) complexes primarily go through clathrin-dependent endocytosis. In contrast, macrophage apoptosis triggered by SR-A-fucoidan internalization requires caveolae-dependent endocytosis. The caveolae-dependent process activates p38 kinase and JNK signaling, whereas the clathrin-mediated endocytosis elicits ERK signaling. Our results suggest that different SR-A endocytic pathways have distinct functional consequences due to the activation of different signaling cascades in macrophages.
Collapse
Affiliation(s)
- Xu-Dong Zhu
- From the Institute of Reproductive Medicine and; Atherosclerosis Research Center, Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China and
| | - Yan Zhuang
- Atherosclerosis Research Center, Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China and
| | - Jing-Jing Ben
- Atherosclerosis Research Center, Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China and
| | - Ling-Ling Qian
- Atherosclerosis Research Center, Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China and
| | - Han-Peng Huang
- Atherosclerosis Research Center, Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China and
| | - Hui Bai
- Atherosclerosis Research Center, Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China and
| | - Jia-Hao Sha
- From the Institute of Reproductive Medicine and
| | - Zhi-Gang He
- the Division of Neuroscience, Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Qi Chen
- From the Institute of Reproductive Medicine and; Atherosclerosis Research Center, Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China and.
| |
Collapse
|