1
|
Zhi F, Liu K, Geng H, Su M, Xu J, Fu L, Ma K, Gao P, Yuan L, Chu Y. Copper sensing transcription factor ArsR2 regulates VjbR to sustain virulence in Brucella abortus. Emerg Microbes Infect 2024; 13:2406274. [PMID: 39295505 PMCID: PMC11425708 DOI: 10.1080/22221751.2024.2406274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 09/21/2024]
Abstract
Brucellosis, caused by the intracellular pathogen Brucella, is a major zoonotic infection that promotes reproductive disease in domestic animals and chronic debilitating conditions in humans. The ArsR family of transcriptional regulators plays key roles in diverse cellular processes, including metal ion homeostasis, responding to adverse conditions, and virulence. However, little is known about the function of ArsR family members in Brucella. Here, we identified ArsR2 as a nonclassical member of the family that lacks autoregulatory function, but which nevertheless plays a vital role in maintaining copper homeostasis in B. abortus. ArsR2 is a global regulator of 241 genes, including those involved in the VirB type IV secretion system (T4SS). Significantly, ArsR2 regulates T4SS production in B. abortus by targeting VjbR which encodes a LuxR-type family transcriptional regulator. Moreover, copper modulates transcriptional activity of ArsR2, but not of VjbR. Furthermore, deletion of arsR2 attenuated virulence in a mouse model. Collectively, these findings enhance understanding of the mechanism by which ArsR proteins regulate virulence gene expression in pathogenic Brucella species.
Collapse
Affiliation(s)
- Feijie Zhi
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Kemeng Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Hao Geng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Mengru Su
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Jian Xu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Lei Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Ke Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Pengcheng Gao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - Lvfeng Yuan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| | - YueFeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, People’s Republic of China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, People’s Republic of China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, People’s Republic of China
| |
Collapse
|
2
|
Binnebose AM, Mullis AS, Haughney SL, Narasimhan B, Bellaire BH. Nanotherapeutic delivery of antibiotic cocktail enhances intra-macrophage killing of Mycobacterium marinum. FRONTIERS IN ANTIBIOTICS 2023; 2:1162941. [PMID: 39816663 PMCID: PMC11732124 DOI: 10.3389/frabi.2023.1162941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/26/2023] [Indexed: 01/18/2025]
Abstract
Mycobacterium marinum is a waterborne pathogen responsible for tuberculosis-like infections in cold-blooded animals and is an opportunistic pathogen in humans. M. marinum is the closest genetic relative of the Mycobacterium tuberculosis complex and is a reliable surrogate for drug susceptibility testing. We synthesized and evaluated two nanoparticle (NP) formulations for compatibility with rifampicin, isoniazid, pyrazinamide, and ethambutol (PIRE), the front-line antimycobacterial drugs used in combination against active tuberculosis infections. Improved in vitro antimicrobial activity was observed with encapsulated rifampicin alone or in a cocktail of drugs formulated through co-encapsulation in amphiphilic polyanhydride NPs. Broth antimicrobial testing revealed that the encapsulation of PIRE in NP resulted in a significant increase in antimicrobial activity, with the benefit over soluble formulations at biologically relevant concentrations ranging from >10 to >3,000 fold. M. marinum-infected human macrophages treated with NP-PIRE were cleared of viable bacteria in 48 h following a single treatment, representing a >4 log reduction in colony-forming units and a >2,000-fold increase in antimicrobial activity. The amphiphilic polyanhydride nanoparticles demonstrated the ability to co-encapsulate PIRE antibiotics and enhance their antimicrobial activity against M. marinum in infected macrophages in culture and in vitro. These data suggest that polyanhydride nanoparticles are a promising nanotherapeutic for combatting Mycobacterium infections through improved intracellular targeting of encapsulated antibiotics.
Collapse
Affiliation(s)
- Andrea M. Binnebose
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
- Department of Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
- Cargill Animal Nutrition, Elk River, MN, United States
| | - Adam S. Mullis
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Shannon L. Haughney
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Bryan H. Bellaire
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
- Department of Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
| |
Collapse
|
3
|
Pellegrini JM, Gorvel JP, Mémet S. Immunosuppressive Mechanisms in Brucellosis in Light of Chronic Bacterial Diseases. Microorganisms 2022; 10:1260. [PMID: 35888979 PMCID: PMC9324529 DOI: 10.3390/microorganisms10071260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 01/27/2023] Open
Abstract
Brucellosis is considered one of the major zoonoses worldwide, constituting a critical livestock and human health concern with a huge socio-economic burden. Brucella genus, its etiologic agent, is composed of intracellular bacteria that have evolved a prodigious ability to elude and shape host immunity to establish chronic infection. Brucella's intracellular lifestyle and pathogen-associated molecular patterns, such as its specific lipopolysaccharide (LPS), are key factors for hiding and hampering recognition by the immune system. Here, we will review the current knowledge of evading and immunosuppressive mechanisms elicited by Brucella species to persist stealthily in their hosts, such as those triggered by their LPS and cyclic β-1,2-d-glucan or involved in neutrophil and monocyte avoidance, antigen presentation impairment, the modulation of T cell responses and immunometabolism. Attractive strategies exploited by other successful chronic pathogenic bacteria, including Mycobacteria, Salmonella, and Chlamydia, will be also discussed, with a special emphasis on the mechanisms operating in brucellosis, such as granuloma formation, pyroptosis, and manipulation of type I and III IFNs, B cells, innate lymphoid cells, and host lipids. A better understanding of these stratagems is essential to fighting bacterial chronic infections and designing innovative treatments and vaccines.
Collapse
|
4
|
Mirzaei R, Sholeh M, Jalalifar S, Zafari E, Kazemi S, Rasouli-Saravani A, Karampoor S, Yousefimashouf R. Immunometabolism in human brucellosis: An emerging field of investigation. Microb Pathog 2021; 158:105115. [PMID: 34332069 DOI: 10.1016/j.micpath.2021.105115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 01/16/2023]
Abstract
In recent years, extreme attention has been focused on the role of immunometabolism in the regulation of immune cell responses in healthy individuals during infection, autoimmunity, and cancer. In the infection biology area, it has been shown that there is a close relationship between the immune system and the host metabolic changes. Brucella species is an intracellular coccobacillus that infects humans and mammals, which led to brucellosis. Brucella species with host-specific evolutionary mechanisms allow it to hide from or manipulate cellular immunity and achieve intracellular persistence. Intracellular bacterial pathogens such as Brucella species also employ host cell resources to replicate and persist inside the host. Targeting these host systems is one promising strategy for developing novel antimicrobials to tackle intracellular infections. This study will summarize the role of metabolic reprogramming in immune cells and their relationship to brucellosis.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Mohammad Sholeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saba Jalalifar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Zafari
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sima Kazemi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ashkan Rasouli-Saravani
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
5
|
Roop RM, Barton IS, Hopersberger D, Martin DW. Uncovering the Hidden Credentials of Brucella Virulence. Microbiol Mol Biol Rev 2021; 85:e00021-19. [PMID: 33568459 PMCID: PMC8549849 DOI: 10.1128/mmbr.00021-19] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Bacteria in the genus Brucella are important human and veterinary pathogens. The abortion and infertility they cause in food animals produce economic hardships in areas where the disease has not been controlled, and human brucellosis is one of the world's most common zoonoses. Brucella strains have also been isolated from wildlife, but we know much less about the pathobiology and epidemiology of these infections than we do about brucellosis in domestic animals. The brucellae maintain predominantly an intracellular lifestyle in their mammalian hosts, and their ability to subvert the host immune response and survive and replicate in macrophages and placental trophoblasts underlies their success as pathogens. We are just beginning to understand how these bacteria evolved from a progenitor alphaproteobacterium with an environmental niche and diverged to become highly host-adapted and host-specific pathogens. Two important virulence determinants played critical roles in this evolution: (i) a type IV secretion system that secretes effector molecules into the host cell cytoplasm that direct the intracellular trafficking of the brucellae and modulate host immune responses and (ii) a lipopolysaccharide moiety which poorly stimulates host inflammatory responses. This review highlights what we presently know about how these and other virulence determinants contribute to Brucella pathogenesis. Gaining a better understanding of how the brucellae produce disease will provide us with information that can be used to design better strategies for preventing brucellosis in animals and for preventing and treating this disease in humans.
Collapse
Affiliation(s)
- R Martin Roop
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Ian S Barton
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Dariel Hopersberger
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Daniel W Martin
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
6
|
Jiao H, Luo Y, Zhou Z, Gu G, Li B, Li W, Liu Y, Wang Y, Wang X, Zhao Y, Wu L, Chen J, Shuai X, Huang Q. Integrative Bioinformatics Indentification of the Autophagic Pathway-Associated miRNA-mRNA Networks in RAW264.7 Macrophage Cells Infected with ∆Omp25 Brucella melitensis. Inflammation 2021; 43:532-539. [PMID: 31807961 DOI: 10.1007/s10753-019-01135-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Brucellosis is a zoonotic infectious disease caused by Brucella infection. Outer membrane protein 25 (Omp25) is closely related to the virulence and immunogenicity of Brucella. However, the molecular mechanism of Omp25 affecting Brucella-mediated macrophage autophagy remains unclear. Our previous study reported that four miRNAs (the upregulation of mmu-miR-146a-5p and mmu-miR-155-5p and downregulation of mmu-miR-149-3p and mmu-miR-5126) were confirmed and revealed the differentially expressed genes (DEGs) profile in RAW264.7 macrophage cells infected with Brucella melitensis Omp25 deletion mutant (∆Omp25 B. melitensis). Here, we predicted the target genes of the four miRNAs by TargetScan, miRanda, and PicTar. GO and KEGG were used for functional enrichment analysis of DEGs profile to reveal the autophagic pathway-associated genes. The overlapped genes, which drawn the autophagic pathway-associated miRNA-mRNA networks by cytoscape software, were identified by intersecting with the predicted target genes and autophagic pathway-associated DEGs. qRT-PCR was performed to validate the mRNAs of networks. The results showed that the autophagic pathway-associated networks of mmu-miR-149-3p-Ptpn5, mmu-miR-149-3p-Ppp2r3c, and mmu-miR-146a-5p-Dusp16 were identified in RAW264.7 macrophage cells infected with ∆Omp25 B. melitensis. Our findings are of great significance in elucidating the function of Omp25, revealing the infection mechanism of Brucella and prophylaxising and treating brucellosis.
Collapse
Affiliation(s)
- Hanwei Jiao
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China.
| | - Yichen Luo
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Zhixiong Zhou
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Guojing Gu
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Bowen Li
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Wenjie Li
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Yuxuan Liu
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Yidan Wang
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Xinglong Wang
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Yu Zhao
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Li Wu
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Jixuan Chen
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Xuehong Shuai
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| | - Qingzhou Huang
- Veterinary Scientific Engineering Research Center, College of Animal Science, Southwest University, Chongqing, 402460, People's Republic of China
| |
Collapse
|
7
|
Szulc-Dąbrowska L, Bossowska-Nowicka M, Struzik J, Toka FN. Cathepsins in Bacteria-Macrophage Interaction: Defenders or Victims of Circumstance? Front Cell Infect Microbiol 2020; 10:601072. [PMID: 33344265 PMCID: PMC7746538 DOI: 10.3389/fcimb.2020.601072] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Macrophages are the first encounters of invading bacteria and are responsible for engulfing and digesting pathogens through phagocytosis leading to initiation of the innate inflammatory response. Intracellular digestion occurs through a close relationship between phagocytic/endocytic and lysosomal pathways, in which proteolytic enzymes, such as cathepsins, are involved. The presence of cathepsins in the endo-lysosomal compartment permits direct interaction with and killing of bacteria, and may contribute to processing of bacterial antigens for presentation, an event necessary for the induction of antibacterial adaptive immune response. Therefore, it is not surprising that bacteria can control the expression and proteolytic activity of cathepsins, including their inhibitors – cystatins, to favor their own intracellular survival in macrophages. In this review, we summarize recent developments in defining the role of cathepsins in bacteria-macrophage interaction and describe important strategies engaged by bacteria to manipulate cathepsin expression and activity in macrophages. Particularly, we focus on specific bacterial species due to their clinical relevance to humans and animal health, i.e., Mycobacterium, Mycoplasma, Staphylococcus, Streptococcus, Salmonella, Shigella, Francisella, Chlamydia, Listeria, Brucella, Helicobacter, Neisseria, and other genera.
Collapse
Affiliation(s)
- Lidia Szulc-Dąbrowska
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland
| | - Magdalena Bossowska-Nowicka
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland
| | - Justyna Struzik
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland
| | - Felix N Toka
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland.,Center for Integrative Mammalian Research, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| |
Collapse
|
8
|
Kornspan D, Zahavi T, Salmon-Divon M. The Acidic Stress Response of the Intracellular Pathogen Brucella melitensis: New Insights from a Comparative, Genome-Wide Transcriptome Analysis. Genes (Basel) 2020; 11:genes11091016. [PMID: 32872264 PMCID: PMC7563570 DOI: 10.3390/genes11091016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 11/30/2022] Open
Abstract
The intracellular pathogenic bacteria belonging to the genus Brucella must cope with acidic stress as they penetrate the host via the gastrointestinal route, and again during the initial stages of intracellular infection. A transcription-level regulation has been proposed to explain this but the specific molecular mechanisms are yet to be determined. We recently reported a comparative transcriptomic analysis of the attenuated vaccine Brucella melitensis strain Rev.1 against the virulent strain 16M in cultures grown under either neutral or acidic conditions. Here, we re-analyze the RNA-seq data of 16M from our previous study and compare it to published transcriptomic data of this strain from both an in cellulo and an in vivo model. We identify 588 genes that are exclusively differentially expressed in 16M grown under acidic versus neutral pH conditions, including 286 upregulated genes and 302 downregulated genes that are not differentially expressed in either the in cellulo or the in vivo model. Of these, we highlight 13 key genes that are known to be associated with a bacterial response to acidic stress and, in our study, were highly upregulated under acidic conditions. These genes provide new molecular insights into the mechanisms underlying the acid-resistance of Brucella within its host.
Collapse
Affiliation(s)
- David Kornspan
- Department of Bacteriology, Kimron Veterinary Institute, Bet Dagan 50250, Israel
- Correspondence: ; Tel.: +972-3-968-1745
| | - Tamar Zahavi
- Genomic Bioinformatics Laboratory, Department of Molecular Biology, Ariel University, Ariel 40700, Israel; (T.Z.); (M.S.-D.)
| | - Mali Salmon-Divon
- Genomic Bioinformatics Laboratory, Department of Molecular Biology, Ariel University, Ariel 40700, Israel; (T.Z.); (M.S.-D.)
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| |
Collapse
|
9
|
Salmon-Divon M, Kornspan D. Transcriptomic analysis of smooth versus rough Brucella melitensis Rev.1 vaccine strains reveals insights into virulence attenuation. Int J Med Microbiol 2019; 310:151363. [PMID: 31699441 DOI: 10.1016/j.ijmm.2019.151363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/10/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022] Open
Abstract
Brucella melitensis Rev.1 is the live attenuated Elberg-originated vaccine strain of the facultative intracellular Brucella species, and is widely used to control brucellosis in small ruminants. However, Rev.1 may cause abortions in small ruminants that have been vaccinated during the last trimester of gestation, it is pathogenic to humans, and it induces antibodies directed at the O-polysaccharide (O-PS) of the smooth lipopolysaccharide, thus making it difficult to distinguish between vaccinated and infected animals. Rough Brucella strains, which lack O-PS and are considered less pathogenic, have been introduced to address these drawbacks; however, as Rev.1 confers a much better immunity than the rough mutants, it is still considered the reference vaccine for the prophylaxis of brucellosis in small ruminants. Therefore, developing an improved vaccine strain, which lacks the Rev.1 drawbacks, is a highly evaluated task, which requires a better understanding of the molecular mechanisms underlying the virulence attenuation of Rev.1 smooth strains and of natural Rev.1 rough strains, which are currently only partly understood. As the acidification of the Brucella-containing vacuole during the initial stages of infection is crucial for their survival, identifying the genes that contribute to their survival in an acidic environment versus a normal environment will greatly assist our understanding of the molecular pathogenic mechanisms and the attenuated virulence of the Rev.1 strain. Here, we compared the transcriptomes of the smooth and natural rough Rev.1 strains, each grown under either normal or acidic conditions. We found 12 key genes that are significantly downregulated in the Rev.1 rough strains under normal pH, as compared with Rev.1 smooth strains, and six highly important genes that are significantly upregulated in the smooth strains under acidic conditions, as compared with Rev.1 rough strains. All 18 differentially expressed genes are associated with bacterial virulence and survival and may explain the attenuated virulence of the rough Rev.1 strains versus smooth Rev.1 strains, thus providing new insights into the virulence attenuation mechanisms of Brucella. These highly important candidate genes may facilitate the design of new and improved brucellosis vaccines.
Collapse
Affiliation(s)
- Mali Salmon-Divon
- Genomic Bioinformatics Laboratory, Department of Molecular Biology, Ariel University, Ariel, Israel; Adelson School of Medicine, Ariel University, Israel.
| | - David Kornspan
- Department of Bacteriology, Kimron Veterinary Institute, Bet Dagan, Israel.
| |
Collapse
|
10
|
A Role for the VPS Retromer in Brucella Intracellular Replication Revealed by Genomewide siRNA Screening. mSphere 2019; 4:4/3/e00380-19. [PMID: 31243080 PMCID: PMC6595151 DOI: 10.1128/msphere.00380-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Brucella, the agent causing brucellosis, is a major zoonotic pathogen with worldwide distribution. Brucella resides and replicates inside infected host cells in membrane-bound compartments called Brucella-containing vacuoles (BCVs). Following uptake, Brucella resides in endosomal BCVs (eBCVs) that gradually mature from early to late endosomal features. Through a poorly understood process that is key to the intracellular lifestyle of Brucella, the eBCV escapes fusion with lysosomes by transitioning to the replicative BCV (rBCV), a replicative niche directly connected to the endoplasmic reticulum (ER). Despite the notion that this complex intracellular lifestyle must depend on a multitude of host factors, a holistic view on which of these components control Brucella cell entry, trafficking, and replication is still missing. Here we used a systematic cell-based small interfering RNA (siRNA) knockdown screen in HeLa cells infected with Brucella abortus and identified 425 components of the human infectome for Brucella infection. These include multiple components of pathways involved in central processes such as the cell cycle, actin cytoskeleton dynamics, or vesicular trafficking. Using assays for pathogen entry, knockdown complementation, and colocalization at single-cell resolution, we identified the requirement of the VPS retromer for Brucella to escape the lysosomal degradative pathway and to establish its intracellular replicative niche. We thus validated the VPS retromer as a novel host factor critical for Brucella intracellular trafficking. Further, our genomewide data shed light on the interplay between central host processes and the biogenesis of the Brucella replicative niche.IMPORTANCE With >300,000 new cases of human brucellosis annually, Brucella is regarded as one of the most important zoonotic bacterial pathogens worldwide. The agent causing brucellosis resides inside host cells within vacuoles termed Brucella-containing vacuoles (BCVs). Although a few host components required to escape the degradative lysosomal pathway and to establish the ER-derived replicative BCV (rBCV) have already been identified, the global understanding of this highly coordinated process is still partial, and many factors remain unknown. To gain deeper insight into these fundamental questions, we performed a genomewide RNA interference (RNAi) screen aiming at discovering novel host factors involved in the Brucella intracellular cycle. We identified 425 host proteins that contribute to Brucella cellular entry, intracellular trafficking, and replication. Together, this study sheds light on previously unknown host pathways required for the Brucella infection cycle and highlights the VPS retromer components as critical factors for the establishment of the Brucella intracellular replicative niche.
Collapse
|
11
|
Głowacka P, Żakowska D, Naylor K, Niemcewicz M, Bielawska-Drózd A. Brucella - Virulence Factors, Pathogenesis and Treatment. Pol J Microbiol 2019; 67:151-161. [PMID: 30015453 PMCID: PMC7256693 DOI: 10.21307/pjm-2018-029] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2018] [Indexed: 12/27/2022] Open
Abstract
Brucellae are Gram-negative, small rods infecting mammals and capable of causing disease called brucellosis. The infection results in abortion and sterility in domestic animals (sheeps, pigs, rams etc). Especially dangerous for humans are: Brucella melitensis, Brucella suis, Brucella abortus, and Brucella canis that trigger unspecific symptoms (flu-like manifestation). Brucella rods are introduced via host cells, by inhalation, skin abrasions, ingestion or mucosal membranes. The most important feature of Brucella is the ability to survive and multiply within both phagocytic and non-phagocytic cells. Brucella does not produce classical virulence factors: exotoxin, cytolisins, exoenzymes, plasmids, fimbria, and drug resistant forms. Major virulence factors are: lipopolysaccharide (LPS), T4SS secretion system and BvrR/BvrS system, which allow interaction with host cell surface, formation of an early, late BCV (Brucella Containing Vacuole) and interaction with endoplasmic reticulum (ER) when the bacteria multiply. The treatment of brucellosis is based on two-drug therapy, the most common combinations of antibiotics are: doxycycline with rifampicin or fluoroquinolones with rifampicin. Currently, also other methods are used to disrupt Brucella intracellular replication (tauroursodeoxycholic acid or ginseng saponin fraction A).
Collapse
Affiliation(s)
- Patrycja Głowacka
- Biological Threats Identification and Countermeasure Center of the General Karol Kaczkowski Military Institute of Hygiene and Epidemiology,Puławy,Poland
| | - Dorota Żakowska
- Biological Threats Identification and Countermeasure Center of the General Karol Kaczkowski Military Institute of Hygiene and Epidemiology,Puławy,Poland
| | - Katarzyna Naylor
- Lublin Medical University, Department of Didactics and Medical Simulation,Lublin,Poland
| | - Marcin Niemcewicz
- Biological Threats Identification and Countermeasure Center of the General Karol Kaczkowski Military Institute of Hygiene and Epidemiology,Puławy,Poland
| | - Agata Bielawska-Drózd
- Biological Threats Identification and Countermeasure Center of the General Karol Kaczkowski Military Institute of Hygiene and Epidemiology,Puławy,Poland
| |
Collapse
|
12
|
Omotade TO, Roy CR. Manipulation of Host Cell Organelles by Intracellular Pathogens. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0022-2019. [PMID: 31025623 PMCID: PMC11590418 DOI: 10.1128/microbiolspec.bai-0022-2019] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Indexed: 12/24/2022] Open
Abstract
In this article, we explore the unique adaptations of intracellular bacterial pathogens that manipulate conserved cellular pathways, organelles, and cargo to convert the phagosome into a pathogen-containing vacuole (PCV). The phagosome is a degradative organelle that rapidly acidifies as it delivers cargo to the lysosome to destroy microbes and cellular debris. However, to avoid this fate, intracellular bacterial pathogens hijack the key molecular modulators of intracellular traffic: small GTPases, phospholipids, SNAREs, and their associated effectors. Following uptake, pathogens that reside in the phagosome either remain associated with the endocytic pathway or rapidly diverge from the preprogrammed route to the lysosome. Both groups rely on effector-mediated mechanisms to meet the common challenges of intracellular life, such as nutrient acquisition, vacuole expansion, and evasion of the host immune response. Mycobacteria, Salmonella, and Coxiella serve as a lens through which we explore regulators of the canonical endocytic route and pathogens that seek to subvert it. On the other hand, pathogens such as Chlamydia, Legionella, and Brucella disconnect from the canonical endocytic route. This bifurcation is linked to extensive hijacking of the secretory pathway and repurposing of the PCV into specialized compartments that resemble organelles in the secretory network. Finally, each pathogen devises specific strategies to counteract host immune responses, such as autophagy, which aim to destroy these aberrant organelles. Collectively, each unique intracellular niche and the pathogens that construct them reflect the outcome of an aggressive and ongoing molecular arms race at the host-pathogen interface. Improving our understanding of these well-adapted pathogens can help us refine our knowledge of conserved cell biological processes.
Collapse
Affiliation(s)
| | - Craig R Roy
- Department of Microbial Pathogenesis, Yale University, New Haven, CT
| |
Collapse
|
13
|
Amjadi O, Rafiei A, Mardani M, Zafari P, Zarifian A. A review of the immunopathogenesis of Brucellosis. Infect Dis (Lond) 2019; 51:321-333. [PMID: 30773082 DOI: 10.1080/23744235.2019.1568545] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Brucellosis, caused by the intracellular pathogens Brucella, is one of the major zoonotic infections. Considering the economic burden, its prevalence has been a health concern especially in endemic regions. Brucella is able to survive and replicate within host cells by expressing different virulence factors and using various strategies to avoid the host's immune response. This leads to progression of the disease from an acute phase to chronic brucellosis. Exploration of genetic variations has confirmed the expected influence of gene polymorphisms on susceptibility and resistance to brucellosis of humans. Since there is no approved human vaccine and treatment is uncertain with risk of relapse, it is important to increase knowledge about pathogenesis, diagnosis and treatment of brucellosis in order to manage and control this infection, especially in endemic regions.
Collapse
Affiliation(s)
- Omolbanin Amjadi
- a Student Research Committee, Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran
| | - Alireza Rafiei
- b Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran
| | - Masoud Mardani
- c Infectious Diseases and Tropical Medicine Research Center , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Parisa Zafari
- a Student Research Committee, Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran.,b Department of Immunology, School of Medicine , Mazandaran University of Medical Sciences , Sari , Iran
| | - Ahmadreza Zarifian
- d Infectious Disease Research Group, Student Research Committee, Medical School , Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
14
|
Kumar DR, Sivalingam J, Mishra SK, Kumar A, Vineeth MR, Chaudhuri P, Kataria RS, Niranjan SK. Differential expression of cytokines in PBMC of Bos indicus and Bos taurus × Bos indicus cattle due to Brucella abortus S19 antigen. Anim Biotechnol 2019; 31:148-154. [PMID: 30717621 DOI: 10.1080/10495398.2018.1555167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Brucellosis is the most dreadful disease among bovines, although breed differences have been observed in prevalence of disease, worldwide. In present study, antibody response and relative expression of proinflammatory cytokines was compared in Bos indicus (zebu) and Bos taurus × Bos indicus (crossbred) cattle vaccinated by live attenuated Brucella abortus S19 antigen. Six female calves (4-6 months age) of both groups were vaccinated with B.abortus S19 strain. Blood samples were collected before vaccination (0d) and 7th (7d), 14th (14d) and 28th (28d) days after vaccination. Indirect ELISA showed high (p < .05) anti-Brucella antibody level after vaccination; with no significant difference between the groups. During Real-time expression, IFNγ, TNFα, IL6 and IL10 genes initially showed down regulation followed by upregulation in both the groups; however, the trend was much prominent in crossbreds. The expressions of IFNγ, TNFα and IL6, proinflammatory molecules important for initial containment of the Brucella were significantly (p < .01) higher in crossbred. The study showed that the Sahiwal cattle were less responsive to B.abortus S19 antigen than crossbreds, indicating its lower sensitivity to the Brucella, comparatively. In contrary, higher expression of the proinflammatory molecules in crossbreds could be important for containment of the organism during initial stage of infection.
Collapse
Affiliation(s)
- D Ravi Kumar
- Animal Genetics Division, ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India.,ICAR-Animal Genetics and Breeding National Dairy Research Institute, Karnal, Haryana, India
| | - Jayakumar Sivalingam
- Animal Genetics Division, ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Shailendra K Mishra
- Animal Genetics Division, ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Anshuman Kumar
- ICAR-Animal Genetics and Breeding National Dairy Research Institute, Karnal, Haryana, India
| | - M R Vineeth
- ICAR-Animal Genetics and Breeding National Dairy Research Institute, Karnal, Haryana, India
| | - Pallab Chaudhuri
- Division of Bacteriology and Mycology, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - R S Kataria
- Animal Genetics Division, ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - S K Niranjan
- Animal Genetics Division, ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| |
Collapse
|
15
|
Nanotherapeutic provides dose sparing and improved antimicrobial activity against Brucella melitensis infections. J Control Release 2019; 294:288-297. [DOI: 10.1016/j.jconrel.2018.12.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 11/19/2022]
|
16
|
Poncin K, Gillet S, De Bolle X. Learning from the master: targets and functions of the CtrA response regulator in Brucella abortus and other alpha-proteobacteria. FEMS Microbiol Rev 2018; 42:500-513. [PMID: 29733367 DOI: 10.1093/femsre/fuy019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 05/02/2018] [Indexed: 12/27/2022] Open
Abstract
The α-proteobacteria are a fascinating group of free-living, symbiotic and pathogenic organisms, including the Brucella genus, which is responsible for a worldwide zoonosis. One common feature of α-proteobacteria is the presence of a conserved response regulator called CtrA, first described in the model bacterium Caulobacter crescentus, where it controls gene expression at different stages of the cell cycle. Here, we focus on Brucella abortus and other intracellular α-proteobacteria in order to better assess the potential role of CtrA in the infectious context. Comparative genomic analyses of the CtrA control pathway revealed the conservation of specific modules, as well as the acquisition of new factors during evolution. The comparison of CtrA regulons also suggests that specific clades of α-proteobacteria acquired distinct functions under its control, depending on the essentiality of the transcription factor. Other CtrA-controlled functions, for instance motility and DNA repair, are proposed to be more ancestral. Altogether, these analyses provide an interesting example of the plasticity of a regulation network, subject to the constraints of inherent imperatives such as cell division and the adaptations to diversified environmental niches.
Collapse
Affiliation(s)
- Katy Poncin
- URBM-Biology, Université de Namur, Unité de recherche en biologie moléculaire, Belgium
| | - Sébastien Gillet
- URBM-Biology, Université de Namur, Unité de recherche en biologie moléculaire, Belgium
| | - Xavier De Bolle
- URBM-Biology, Université de Namur, Unité de recherche en biologie moléculaire, Belgium
| |
Collapse
|
17
|
Gheibi A, Khanahmad H, Kashfi K, Sarmadi M, Khorramizadeh MR. Development of new generation of vaccines for Brucella abortus. Heliyon 2018; 4:e01079. [PMID: 30603712 PMCID: PMC6307385 DOI: 10.1016/j.heliyon.2018.e01079] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 01/18/2023] Open
Abstract
Brucella abortus is a Gram-negative facultative and intracellular bacteria, it causes bovine brucellosis, a zoonotic disease that is responsible for considerable economic loss to owners of domesticated animals and can cause problems in otherwise healthy humans. There are a few available live attenuated vaccines for animal immunization against brucellosis; however, these have significant side effects and offer insufficient protective efficacy. Thus, the need for more research into the Molecular pathobiology and immunological properties of B. abortus that would lead to the development of better and safer vaccines. In this paper we have reviewed the main aspects of the pathology and the responsive immunological mechanisms, we have also covered current and new prospective vaccines against B. abortus.
Collapse
Affiliation(s)
- Azam Gheibi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA
| | - Mahdieh Sarmadi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Reza Khorramizadeh
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Sedzicki J, Tschon T, Low SH, Willemart K, Goldie KN, Letesson JJ, Stahlberg H, Dehio C. 3D correlative electron microscopy reveals continuity of Brucella-containing vacuoles with the endoplasmic reticulum. J Cell Sci 2018; 131:jcs.210799. [PMID: 29361547 DOI: 10.1242/jcs.210799] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/04/2018] [Indexed: 01/17/2023] Open
Abstract
Entry of the facultative intracellular pathogen Brucella into host cells results in the formation of endosomal Brucella-containing vacuoles (eBCVs) that initially traffic along the endocytic pathway. eBCV acidification triggers the expression of a type IV secretion system that translocates bacterial effector proteins into host cells. This interferes with lysosomal fusion of eBCVs and supports their maturation to replicative Brucella-containing vacuoles (rBCVs). Bacteria replicate in rBCVs to large numbers, eventually occupying most of the cytoplasmic volume. As rBCV membranes tightly wrap each individual bacterium, they are constantly being expanded and remodeled during exponential bacterial growth. rBCVs are known to carry endoplasmic reticulum (ER) markers; however, the relationship of the vacuole to the genuine ER has remained elusive. Here, we have reconstructed the 3-dimensional ultrastructure of rBCVs and associated ER by correlative structured illumination microscopy (SIM) and focused ion beam/scanning electron microscopic tomography (FIB/SEM). Studying B. abortus-infected HeLa cells and trophoblasts derived from B. melitensis-infected mice, we demonstrate that rBCVs are complex and interconnected compartments that are continuous with neighboring ER cisternae, thus supporting a model that rBCVs are extensions of genuine ER.
Collapse
Affiliation(s)
- Jaroslaw Sedzicki
- Focal Area Infection Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland.,Center for Cellular Imaging and NanoAnalytics (C-CINA), Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Therese Tschon
- Focal Area Infection Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Shyan Huey Low
- Focal Area Infection Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Kevin Willemart
- Microorganisms Biology Research Unit (URBM, Unité de Recherche en Biologie des Microorganismes), University of Namur, 5000 Namur, Belgium
| | - Kenneth N Goldie
- Center for Cellular Imaging and NanoAnalytics (C-CINA), Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Jean-Jacques Letesson
- Microorganisms Biology Research Unit (URBM, Unité de Recherche en Biologie des Microorganismes), University of Namur, 5000 Namur, Belgium
| | - Henning Stahlberg
- Center for Cellular Imaging and NanoAnalytics (C-CINA), Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Christoph Dehio
- Focal Area Infection Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
19
|
Alves-Silva J, Tavares IP, Guimarães ES, Costa Franco MM, Figueiredo BC, Marques JT, Splitter G, Oliveira SC. Modulation of Microtubule Dynamics Affects Brucella abortus Intracellular Survival, Pathogen-Containing Vacuole Maturation, and Pro-inflammatory Cytokine Production in Infected Macrophages. Front Microbiol 2017; 8:2217. [PMID: 29184543 PMCID: PMC5694624 DOI: 10.3389/fmicb.2017.02217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/27/2017] [Indexed: 01/18/2023] Open
Abstract
The microtubule (MT) cytoskeleton regulates several cellular processes related to the immune system. For instance, an intricate intracellular transport mediated by MTs is responsible for the proper localization of vesicular receptors of innate immunity and its adaptor proteins. In the present study, we used nocodazole to induce MTs depolymerization and paclitaxel or recombinant (r) TIR (Toll/interleukin-1 receptor) domain containing protein (TcpB) to induce MT stabilization in bone marrow-derived macrophages infected with Brucella abortus. Following treatment of the cells, we evaluated their effects on pathogen intracellular replication and survival, and in pro-inflammatory cytokine production. First, we observed that intracellular trafficking and maturation of Brucella-containing vesicles (BCVs) is affected by partial destabilization or stabilization of the MTs network. A typical marker of early BCVs, LAMP-1, is retained in late BCVs even 24 h after infection in the presence of low doses of nocodazole or paclitaxel and in the presence of different amounts of rTcpB. Second, microscopy and colony forming unit analysis revealed that bacterial load was increased in infected macrophages treated with lower doses of nocodazole or paclitaxel and with rTcpB compared to untreated cells. Third, innate immune responses were also affected by disturbing MT dynamics. MT depolymerization by nocodazole reduced IL-12 production in infected macrophages. Conversely, rTcpB-treated cells augmented IL-12 and IL-1β secretion in infected cells. In summary, these findings demonstrate that modulation of MTs affects several crucial steps of B. abortus pathogenesis, including BCV maturation, intracellular survival and IL-12 secretion in infected macrophages.
Collapse
Affiliation(s)
- Juliana Alves-Silva
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabela P Tavares
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Erika S Guimarães
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Miriam M Costa Franco
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Barbara C Figueiredo
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - João T Marques
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gary Splitter
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
20
|
Li S, Li P, Zhang L, Hu W, Wang M, Liu Y, Tang G, Wang D, Zhou B, Yan J. The role of reactive oxygen intermediates in the intracellular fate of Leptospira interrogans in the macrophages of different hosts. PLoS One 2017; 12:e0178618. [PMID: 28575082 PMCID: PMC5456347 DOI: 10.1371/journal.pone.0178618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/16/2017] [Indexed: 12/20/2022] Open
Abstract
Background Pathogenic species of Leptospira cause leptospirosis, a global zoonotic disease. Our previous work showed that leptospires survive and replicate in human macrophages but are killed in murine macrophages. However, the mechanism responsible for the different intracellular fates of leptospires within the macrophages of different hosts remains unclear. Results The present study demonstrates that infection with Leptospira interrogans caused significant up-regulation of reactive oxygen species (ROS) and superoxide in J774A.1 cells but did so to a lesser extent in THP-1 cells. The up-regulation of ROS and superoxide was significantly inhibited by the NADPH oxidase inhibitor apocynin. The damaged leptospires and remnants of leptospires within membrane-bound vacuoles were significantly inhibited by apocynin in J774A.1 cells but were less inhibited in THP-1 cells. In addition, apocynin significantly prevented damage to leptospires and the co-localization of L. interrogans with lysosomes in J774A.1 cells but did so to a lesser extent in THP-1 cells. Furthermore, the relative fluorescence intensity levels of intracellular leptospires and the viability of the intracellular leptospires increased in apocynin pretreated J774A.1 and THP-1 cells after 2 h of infection. Conclusions The present study, based on our previous findings, further demonstrated that ROS contributed substantially to the bactericidal ability of mouse macrophages to kill intracellular leptospires. However, ROS did not contribute as much in human macrophages, which partially explains the different intracellular fates of L. interrogans in human and mouse macrophages.
Collapse
Affiliation(s)
- Shijun Li
- Institute of Communicable Disease Control and Prevention, Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou, P.R. China
| | - Peili Li
- College of Animal Science, Guizhou University, Huaxi District, Guiyang, Guizhou, P.R. China
| | - Lei Zhang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, P.R. China
| | - Weilin Hu
- Department of Medical Microbiology and Parasitology, College of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Ming Wang
- College of Animal Science, Guizhou University, Huaxi District, Guiyang, Guizhou, P.R. China
| | - Ying Liu
- Institute of Communicable Disease Control and Prevention, Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou, P.R. China
| | - Guangpeng Tang
- Institute of Communicable Disease Control and Prevention, Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou, P.R. China
| | - Dingming Wang
- Institute of Communicable Disease Control and Prevention, Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou, P.R. China
| | - Bijun Zhou
- College of Animal Science, Guizhou University, Huaxi District, Guiyang, Guizhou, P.R. China
| | - Jie Yan
- Department of Medical Microbiology and Parasitology, College of Medicine, Zhejiang University, Hangzhou, P.R. China
| |
Collapse
|
21
|
Mohan A, Saxena HM, Malhotra P. A comparison of titers of anti-Brucella antibodies of naturally infected and healthy vaccinated cattle by standard tube agglutination test, microtiter plate agglutination test, indirect hemagglutination assay, and indirect enzyme-linked immunosorbent assay. Vet World 2016; 9:717-22. [PMID: 27536032 PMCID: PMC4983122 DOI: 10.14202/vetworld.2016.717-722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/08/2016] [Indexed: 02/05/2023] Open
Abstract
AIM We determined the antibody response in cattle naturally infected with brucellosis and normal healthy adult cattle vaccinated during calf hood with strain 19. MATERIALS AND METHODS The antibody titers were measured by standard tube agglutination test (STAT), microtiter plate agglutination test (MAT), indirect hemagglutination assay (IHA), and indirect enzyme-linked immunosorbent assay (iELISA) as per standard protocols. RESULTS The mean STAT titers were 1.963±0.345 in infected cattle and 1.200±0.155 in healthy vaccinated cattle. The difference was extremely significant (p<0.0001). The mean MAT titers were 2.244±0.727 in infected cattle and 1.200±0.155 in healthy vaccinated cattle. The difference was very significant (p<0.005). The mean IHA titers in infected cattle were 2.284±0.574, and those in healthy vaccinated cattle were 1.200±0.155. The difference was extremely significant (p=0.0002). However, the difference in mean iELISA titers of infected cattle (1.3678±0.014) and healthy vaccinated cattle (1.367±0.014) was non-significant. The infected animals showed very high titers of agglutinating antibodies compared to the vaccinated animals. However, it cannot be ascertained whether these antibodies are due to vaccine or response to infection. Since the infected animals had been vaccinated earlier, the current infection may suggest that vaccination was unable to induce protective levels of antibody. The heightened antibody response after infection may also indicate a secondary immune response to the antigens common to the vaccine strain and wild Brucella organisms. CONCLUSION The brucellosis infected animals showed very high titers of agglutinating antibodies compared to the vaccinated animals.
Collapse
Affiliation(s)
- Anju Mohan
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana - 141 004, Punjab, India
| | - Hari Mohan Saxena
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana - 141 004, Punjab, India
- Corresponding author: Hari Mohan Saxena, e-mail: , AM: , PM:
| | - Puneet Malhotra
- Department of Animal Genetics and Breeding, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana - 141 004, Punjab, India
| |
Collapse
|
22
|
Pesce Viglietti AI, Arriola Benitez PC, Giambartolomei GH, Delpino MV. Brucella abortus-infected B cells induce osteoclastogenesis. Microbes Infect 2016; 18:529-35. [PMID: 27109230 DOI: 10.1016/j.micinf.2016.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 03/07/2016] [Accepted: 04/13/2016] [Indexed: 01/18/2023]
Abstract
Brucella abortus is an intracellular bacterium that establishes lifelong infections in livestock and humans although the mechanisms of its chronicity are poorly understood. Activated B cells have long lifespan and B. abortus infection activates B cells. Our results indicate that the direct infection of B cells with B. abortus induced matrix metalloproteinase-9 (MMP-9), receptor activator for NF κB ligand (RANKL), tumor necrosis factor (TNF)-α and interleukin (IL)-6 secretion. In addition, supernatants from B. abortus-infected B cells induced bone marrow-derived monocytes to undergo osteoclastogenesis. Using osteoprotegerin, RANKL's decoy receptor, we determined that RANKL is involved in osteoclastogenesis induced by supernatants from B. abortus-infected B cells. The results presented here shed light on how the interactions of B. abortus with B cells may have a role in the pathogenesis of brucellar osteoarticular disease.
Collapse
Affiliation(s)
- Ayelén Ivana Pesce Viglietti
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Hospital de Clínicas "José de San Martín", Facultad de Medicina, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Paula Constanza Arriola Benitez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Hospital de Clínicas "José de San Martín", Facultad de Medicina, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Guillermo Hernán Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Hospital de Clínicas "José de San Martín", Facultad de Medicina, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Hospital de Clínicas "José de San Martín", Facultad de Medicina, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
23
|
De Bolle X, Crosson S, Matroule JY, Letesson JJ. Brucella abortus Cell Cycle and Infection Are Coordinated. Trends Microbiol 2015; 23:812-821. [PMID: 26497941 DOI: 10.1016/j.tim.2015.09.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 09/09/2015] [Accepted: 09/24/2015] [Indexed: 12/29/2022]
Abstract
Brucellae are facultative intracellular pathogens. The recent development of methods and genetically engineered strains allowed the description of cell-cycle progression of Brucella abortus, including unipolar growth and the ordered initiation of chromosomal replication. B. abortus cell-cycle progression is coordinated with intracellular trafficking in the endosomal compartments. Bacteria are first blocked at the G1 stage, growth and chromosome replication being resumed shortly before reaching the intracellular proliferation compartment. The control mechanisms of cell cycle are similar to those reported for the bacterium Caulobacter crescentus, and they are crucial for survival in the host cell. The development of single-cell analyses could also be applied to other bacterial pathogens to investigate their cell-cycle progression during infection.
Collapse
Affiliation(s)
- Xavier De Bolle
- University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium.
| | - Sean Crosson
- University of Chicago, Gordon Center for Integrative Science W125, 929 E. 57th Street, Chicago, IL 60637, USA
| | | | | |
Collapse
|
24
|
Celli J. The changing nature of the Brucella-containing vacuole. Cell Microbiol 2015; 17:951-8. [PMID: 25916795 DOI: 10.1111/cmi.12452] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/01/2015] [Accepted: 04/21/2015] [Indexed: 01/18/2023]
Abstract
Bacteria of the genus Brucella are intracellular vacuolar pathogens of mammals that cause the worldwide zoonosis brucellosis, and reside within phagocytes of infected hosts to promote their survival, persistence and proliferation. These traits are essential to the bacterium's ability to cause disease and have been the subject of much investigation to gain an understanding of Brucella pathogenic mechanisms. Although the endoplasmic reticulum-derived nature of the Brucella replicative niche has been long known, major strides have recently been made in deciphering the molecular mechanisms of its biogenesis, including the identification of bacterial determinants and host cellular pathways involved in this process. Here I will review and discuss the most recent advances in our knowledge of Brucella intracellular pathogenesis, with an emphasis on bacterial exploitation of the host endoplasmic reticulum-associated functions, and how autophagy-related processes contribute to the bacterium's intracellular cycle.
Collapse
Affiliation(s)
- Jean Celli
- Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
25
|
Mol JPS, Costa EA, Carvalho AF, Sun YH, Tsolis RM, Paixão TA, Santos RL. Early transcriptional responses of bovine chorioallantoic membrane explants to wild type, ΔvirB2 or ΔbtpB Brucella abortus infection. PLoS One 2014; 9:e108606. [PMID: 25259715 PMCID: PMC4178178 DOI: 10.1371/journal.pone.0108606] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 08/29/2014] [Indexed: 12/23/2022] Open
Abstract
The pathogenesis of the Brucella-induced inflammatory response in the bovine placenta is not completely understood. In this study we evaluated the role of the B. abortus Type IV secretion system and the anti-inflammatory factor BtpB in early interactions with bovine placental tissues. Transcription profiles of chorioallantoic membrane (CAM) explants inoculated with wild type (strain 2308), ΔvirB2 or ΔbtpB Brucella abortus were compared by microarray analysis at 4 hours post infection. Transcripts with significant variation (>2 fold change; P<0.05) were functionally classified, and transcripts related to defense and inflammation were assessed by quantitative real time RT-PCR. Infection with wild type B. abortus resulted in slightly more genes with decreased than increased transcription levels. Conversely, infection of trophoblastic cells with the ΔvirB2 or the ΔbtpB mutant strains, that lack a functional T4SS or that has impaired inhibition of TLR signaling, respectively, induced more upregulated than downregulated genes. Wild type Brucella abortus impaired transcription of host genes related to immune response when compared to ΔvirB and ΔbtpB mutants. Our findings suggest that proinflammatory genes are negatively modulated in bovine trophoblastic cells at early stages of infection. The virB operon and btpB are directly or indirectly related to modulation of these host genes. These results shed light on the early interactions between B. abortus and placental tissue that ultimately culminate in inflammatory pathology and abortion.
Collapse
Affiliation(s)
- Juliana P. S. Mol
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Erica A. Costa
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alex F. Carvalho
- Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Yao-Hui Sun
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Reneé M. Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Tatiane A. Paixão
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Renato L. Santos
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- * E-mail:
| |
Collapse
|
26
|
Immune evasion, stress resistance, and efficient nutrient acquisition are crucial for intracellular survival of Candida glabrata within macrophages. EUKARYOTIC CELL 2013; 13:170-83. [PMID: 24363366 DOI: 10.1128/ec.00262-13] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Candida glabrata is both a human fungal commensal and an opportunistic pathogen which can withstand activities of the immune system. For example, C. glabrata can survive phagocytosis and replicates within macrophages. However, the mechanisms underlying intracellular survival remain unclear. In this work, we used a functional genomic approach to identify C. glabrata determinants necessary for survival within human monocyte-derived macrophages by screening a set of 433 deletion mutants. We identified 23 genes which are required to resist killing by macrophages. Based on homologies to Saccharomyces cerevisiae orthologs, these genes are putatively involved in cell wall biosynthesis, calcium homeostasis, nutritional and stress response, protein glycosylation, or iron homeostasis. Mutants were further characterized using a series of in vitro assays to elucidate the genes' functions in survival. We investigated different parameters of C. glabrata-phagocyte interactions: uptake by macrophages, replication within macrophages, phagosomal pH, and recognition of mutant cells by macrophages as indicated by production of reactive oxygen species and tumor necrosis factor alpha (TNF-α). We further studied the cell surface integrity of mutant cells, their ability to grow under nutrient-limited conditions, and their susceptibility to stress conditions mirroring the harsh environment inside a phagosome. Additionally, resistance to killing by neutrophils was analyzed. Our data support the view that immune evasion is a key aspect of C. glabrata virulence and that increased immune recognition causes increased antifungal activities by macrophages. Furthermore, stress resistance and efficient nutrient acquisition, in particular, iron uptake, are crucial for intraphagosomal survival of C. glabrata.
Collapse
|
27
|
Salcedo SP, Chevrier N, Lacerda TLS, Ben Amara A, Gerart S, Gorvel VA, de Chastellier C, Blasco JM, Mege JL, Gorvel JP. Pathogenic brucellae replicate in human trophoblasts. J Infect Dis 2013; 207:1075-83. [PMID: 23303808 DOI: 10.1093/infdis/jit007] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Brucellae replicate in a vacuole derived from the endoplasmic reticulum (ER) in epithelial cells, macrophages, and dendritic cells. In animals, trophoblasts are also key cellular targets where brucellae efficiently replicate in association with the ER. Therefore, we investigated the ability of Brucella spp. to infect human trophoblasts using both immortalized and primary trophoblasts. Brucella extensively proliferated within different subpopulations of trophoblasts, suggesting that they constitute an important niche in cases where the fetal-maternal barrier is breached. In extravillous trophoblasts (EVTs), B. abortus and B. suis replicated within single-membrane acidic lysosomal membrane-associated protein 1-positive inclusions, whereas B. melitensis replicated in the ER-derived compartment. Furthermore, B. melitensis but not B. abortus nor B. suis interfered with the invasive capacity of EVT-like cells in vitro. Because EVTs are essential for implantation during early stages of pregnancy, the nature of the replication niche may have a central role during Brucella-associated abortion in infected women.
Collapse
|
28
|
O'Callaghan D. Novel replication profiles of Brucella in human trophoblasts give insights into the pathogenesis of infectious abortion. J Infect Dis 2013; 207:1034-6. [PMID: 23303807 DOI: 10.1093/infdis/jit010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
29
|
Peptide nucleic acids inhibit growth of Brucella suis in pure culture and in infected murine macrophages. Int J Antimicrob Agents 2013; 41:358-62. [PMID: 23305655 DOI: 10.1016/j.ijantimicag.2012.11.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 11/07/2012] [Accepted: 11/09/2012] [Indexed: 01/01/2023]
Abstract
Peptide nucleic acids (PNAs) are single-stranded, synthetic nucleic acid analogues containing a pseudopeptide backbone in place of the phosphodiester sugar-phosphate. When PNAs are covalently linked to cell-penetrating peptides (CPPs) they readily penetrate the bacterial cell envelope, inhibit expression of targeted genes and cause growth inhibition both of Gram-positive and Gram-negative bacteria. However, the effectiveness of PNAs against Brucella, a facultative intracellular bacterial pathogen, was unknown. The susceptibility of a virulent Brucella suis strain to a variety of PNAs was assessed in pure culture as well as in murine macrophages. The studies showed that some of the PNAs targeted to Brucella genes involved in DNA (polA, dnaG, gyrA), RNA (rpoB), cell envelope (asd), fatty acid (kdtA, acpP) and protein (tsf) synthesis inhibit the growth of B. suis in culture and in macrophages after 24 h of treatment. PNA treatment inhibited Brucella growth by interfering with gene expression in a sequence-specific and dose-dependent manner at micromolar concentrations. The most effective PNA in broth culture was that targeting polA at ca. 12 μM. In contrast, in B. suis-infected macrophages, the most effective PNAs were those targeting asd and dnaG at 30 μM; both of these PNAs had little inhibitory effect on Brucella in broth culture. The polA PNA that inhibits wild-type B. suis also inhibits the growth of wild-type Brucella melitensis 16M and Brucella abortus 2308 in culture. This study reveals the potential usefulness of antisense PNA constructs as novel therapeutic agents against intracellular Brucella.
Collapse
|
30
|
Canton J, Kima PE. Interactions of pathogen-containing compartments with the secretory pathway. Cell Microbiol 2012; 14:1676-86. [PMID: 22862745 DOI: 10.1111/cmi.12000] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 07/20/2012] [Accepted: 07/23/2012] [Indexed: 02/03/2023]
Abstract
A subgroup of intracellular pathogens reside and replicate within membrane-bound compartments often termed pathogen-containing compartments (PCC). PCCs navigate around a wide range of host cell vesicles and organelles. In light of the perils of engaging with vesicles of the endocytic pathway, most PCCs modulate their interactions with endocytic vesicles while a few avoid those interactions. The secretory pathway constitutes another important grouping of vesicles and organelles in host cells. Although the negative consequences of engaging with the secretory pathway are not known, there is evidence that PCCs interact differentially with vesicles and organelles in this pathway as well. In this review, we consider three prokaryote pathogens and two protozoan parasites for which there is information on the interactions of their PCCs with the secretory pathway. Current understandings of the molecular interactions as well as the metabolic benefits that accompany those interactions are discussed. Not unexpectedly, our understanding of the extent of these interactions is variable. An underlying theme that is brought to the fore is that PCCs establish preferential interactions with distinct compartments of the secretory pathway.
Collapse
Affiliation(s)
- Johnathan Canton
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | | |
Collapse
|
31
|
de Barsy M, Mirabella A, Letesson JJ, De Bolle X. A Brucella abortus cstA mutant is defective for association with endoplasmic reticulum exit sites and displays altered trafficking in HeLa cells. MICROBIOLOGY-SGM 2012; 158:2610-2618. [PMID: 22820839 DOI: 10.1099/mic.0.060509-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Members of the genus Brucella are facultative intracellular pathogenic bacteria able to control maturation of their vacuoles. In several cell types, Brucella is able to reach a proliferation compartment derived from the endoplasmic reticulum (ER). Since ER exit site (ERES) functions are required for Brucella proliferation, we performed a yeast two-hybrid screen between human ERES-associated proteins and the predicted brucella proteome. This screening led to the identification of CstA, a conserved protein that specifically interacts with Sec24A, a component of the ERES. We found that a tagged CstA is secreted in Brucella abortus culture medium. This secretion is independent of the type IV secretion system VirB and the flagellum, suggesting that CstA is secreted through another system. We also discovered that a B. abortus cstA mutant is impaired for its association with the Sec23 ERES marker. The B. abortus cstA mutant displayed peculiar trafficking, with reduced association with LAMP1 and Calnexin 12 h post-infection in HeLa cells. However, its intracellular proliferation kinetics was not affected. The data reported here suggest that CstA could be directly or indirectly involved in the control of B. abortus intracellular trafficking in HeLa cells.
Collapse
Affiliation(s)
- Marie de Barsy
- Research Unit in Microorganisms Biology (URBM), University of Namur (FUNDP), Namur, Belgium
| | - Aurélie Mirabella
- Research Unit in Microorganisms Biology (URBM), University of Namur (FUNDP), Namur, Belgium
| | - Jean-Jacques Letesson
- Research Unit in Microorganisms Biology (URBM), University of Namur (FUNDP), Namur, Belgium
| | - Xavier De Bolle
- Research Unit in Microorganisms Biology (URBM), University of Namur (FUNDP), Namur, Belgium
| |
Collapse
|
32
|
Goenka R, Guirnalda PD, Black SJ, Baldwin CL. B Lymphocytes provide an infection niche for intracellular bacterium Brucella abortus. J Infect Dis 2012; 206:91-8. [PMID: 22561364 DOI: 10.1093/infdis/jis310] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Brucella spp. are intracellular bacteria that establish lifelong infections whose mechanisms of chronicity are poorly understood. Notably, B cells facilitate the establishment of the high infection plateau that persists for months. METHODS We evaluated the contribution of murine B cells toward providing infection niches for Brucella by using flow cytometry and microscopy and by determining live bacterial counts associated with B cells both in vivo and in vitro. RESULTS Herein we demonstrate that immunoglobulin M and complement-opsonized Brucella abortus infects and survives inside primary murine B cells protected from bactericidal effects of gentamicin. The entry was dependent on microfilaments for internalization and subsequently brucellae reside in a late endosomal/lysosomal compartment. Throughout the infection, 10% of colony-forming units from infected mice was associated with B cells, and these cells transferred disease to naive hosts. Furthermore, Brucella-positive cells were positive for transforming growth factor (TGF) β1, and about 10% of such cells were B cells, similar to rates found for other intracellular pathogens that induce their hosts cells to produce TGF-β1. CONCLUSIONS To conclude, infected B cells contribute to chronic bacterial infections by providing an intracellular niche that may exert an immunoregulatory role. Although professional phagocytic cells harbor intracellular bacteria including Brucella, infection of lymphocytes by bacteria has not been previously appreciated.
Collapse
Affiliation(s)
- Radhika Goenka
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | | | | | | |
Collapse
|
33
|
Ritchie JA, Rupper A, Cardelli JA, Bellaire BH. Host interferon-γ inducible protein contributes to Brucella survival. Front Cell Infect Microbiol 2012; 2:55. [PMID: 22919646 PMCID: PMC3417648 DOI: 10.3389/fcimb.2012.00055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 04/07/2012] [Indexed: 11/13/2022] Open
Abstract
Brucella spp. are highly adapted intracellular pathogens of mammals that cause chronic infections while surving and replicating in host monocytes and macrophages. Although monocytes are normally susceptible to infection, pretreatment with pro-inflammatory cytokine interferon-γ (IFN-γ) activates cellular defense mechanisms that increase intracellular killing of Brucella and prevents bacterial replication. We examined the contribution of the IFN-γ inducible GTPase, LRG-47, to B. abortus 2308 infection in in vitro and in vivo murine models. Infecting non-activated macrophages from LRG-47(-/-) mice revealed that loss of this host protein negatively effected the intracellular survival and replication of IgG opsonized B. abortus. In contrast, survival and replication of non-opsonized B. abortus was the same in both C57/B6 and LRG-47(-/-) peritoneal macrophages. Following IFN-γ activation of LRG-47(-/-) monocytes, IgG opsonized B. abortus survived better than non-opsonized bacteria. The differential fate of opsonized and non-opsonized B. abortus was only observed in macrophages collected from LRG-47(-/-) mice. Given the specific nature of the relationship between this host protein and the mechanism of Brucella internalization, LRG-47(-/-) mice were infected with B. abortus to assess whether the loss of the lrg47 protein would affect the ability of the bacteria to colonize or persist within the host. B. abortus were able to establish and maintain similar numbers of bacteria in both C57/B6 mice and LRG-47(-/-) through 3 weeks post intraperitoneal infection. By 9 weeks p.i. fewer B. abortus were recovered from LRG-47(-/-) mice than controls, suggesting that the host protein has a positive role in maintaining long term persistence of the bacteria within the host. These observations demonstrating a positive role for a host IFN-γ induced protein defense protein has yet to be reported. These results provide interesting insight into the complex interaction between Brucella and their host.
Collapse
Affiliation(s)
- Jennifer A Ritchie
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport LA, USA
| | | | | | | |
Collapse
|
34
|
A new flow-cytometry-based opsonophagocytosis assay for the rapid measurement of functional antibody levels against Group B Streptococcus. J Immunol Methods 2012; 378:11-9. [DOI: 10.1016/j.jim.2012.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 01/20/2012] [Accepted: 01/23/2012] [Indexed: 11/19/2022]
|
35
|
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
|
36
|
Starr T, Child R, Wehrly TD, Hansen B, Hwang S, López-Otin C, Virgin HW, Celli J. Selective subversion of autophagy complexes facilitates completion of the Brucella intracellular cycle. Cell Host Microbe 2012; 11:33-45. [PMID: 22264511 PMCID: PMC3266535 DOI: 10.1016/j.chom.2011.12.002] [Citation(s) in RCA: 246] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 10/26/2011] [Accepted: 12/01/2011] [Indexed: 12/22/2022]
Abstract
Autophagy is a cellular degradation process that can capture and eliminate intracellular microbes by delivering them to lysosomes for destruction. However, pathogens have evolved mechanisms to subvert this process. The intracellular bacterium Brucella abortus ensures its survival by forming the Brucella-containing vacuole (BCV), which traffics from the endocytic compartment to the endoplasmic reticulum (ER), where the bacterium proliferates. We show that Brucella replication in the ER is followed by BCV conversion into a compartment with autophagic features (aBCV). While Brucella trafficking to the ER was unaffected in autophagy-deficient cells, aBCV formation required the autophagy-initiation proteins ULK1, Beclin 1, and ATG14L and PI3-kinase activity. However, aBCV formation was independent of the autophagy-elongation proteins ATG5, ATG16L1, ATG4B, ATG7, and LC3B. Furthermore, aBCVs were required to complete the intracellular Brucella lifecycle and for cell-to-cell spreading, demonstrating that Brucella selectively co-opts autophagy-initiation complexes to subvert host clearance and promote infection.
Collapse
Affiliation(s)
- Tregei Starr
- Laboratory of Intracellular Parasites National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Robert Child
- Laboratory of Intracellular Parasites National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Tara D. Wehrly
- Laboratory of Intracellular Parasites National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Bryan Hansen
- Electron Microscopy Unit, Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Seungmin Hwang
- Department of Pathology and Immunology and Midwest Regional Center of Excellence for Biodefense and Emerging infectious Diseases Research, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Carlos López-Otin
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Herbert W. Virgin
- Department of Pathology and Immunology and Midwest Regional Center of Excellence for Biodefense and Emerging infectious Diseases Research, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jean Celli
- Laboratory of Intracellular Parasites National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| |
Collapse
|
37
|
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: 182] [Impact Index Per Article: 14.0] [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
|
38
|
Seider K, Brunke S, Schild L, Jablonowski N, Wilson D, Majer O, Barz D, Haas A, Kuchler K, Schaller M, Hube B. The facultative intracellular pathogen Candida glabrata subverts macrophage cytokine production and phagolysosome maturation. THE JOURNAL OF IMMUNOLOGY 2011; 187:3072-86. [PMID: 21849684 DOI: 10.4049/jimmunol.1003730] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although Candida glabrata is an important human pathogenic yeast, its pathogenicity mechanisms are largely unknown. Immune evasion strategies seem to play key roles during infection, since very little inflammation is observed in mouse models. Furthermore, C. glabrata multiplies intracellularly after engulfment by macrophages. In this study, we sought to identify the strategies that enable C. glabrata to survive phagosome biogenesis and antimicrobial activities within human monocyte-derived macrophages. We show that, despite significant intracellular proliferation, macrophage damage or apoptosis was not apparent, and production of reactive oxygen species was inhibited. Additionally, with the exception of GM-CSF, levels of pro- and anti-inflammatory cytokines were only marginally increased. We demonstrate that adhesion to and internalization by macrophages occur within minutes, and recruitment of endosomal early endosomal Ag 1 and lysosomal-associated membrane protein 1 indicates phagosome maturation. However, phagosomes containing viable C. glabrata, but not heat-killed yeasts, failed to recruit cathepsin D and were only weakly acidified. This inhibition of acidification did not require fungal viability, but it had a heat-sensitive surface attribute. Therefore, C. glabrata modifies the phagosome into a nonacidified environment and multiplies until the host cells finally lyse and release the fungi. Our results suggest persistence of C. glabrata within macrophages as a possible immune evasion strategy.
Collapse
Affiliation(s)
- Katja Seider
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, 07745 Jena, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Polyanhydride microparticles enhance dendritic cell antigen presentation and activation. Acta Biomater 2011; 7:2857-64. [PMID: 21439412 DOI: 10.1016/j.actbio.2011.03.023] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 03/09/2011] [Accepted: 03/16/2011] [Indexed: 12/20/2022]
Abstract
The present study was designed to evaluate the adjuvant activity of polyanhydride microparticles prepared in the absence of additional stabilizers, excipients or immune modulators. Microparticles composed of varying ratios of either 1,6-bis(p-carboxyphenoxy)hexane (CPH) and sebacic acid or 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane and CPH were added to in vitro cultures of bone marrow-derived dendritic cells (DCs). Microparticles were efficiently and rapidly phagocytosed by DCs in the absence of opsonization and without centrifugation or agitation. Within 2h, internalized particles were rapidly localized to an acidic, phagolysosomal compartment. By 48 h, only a minor reduction in microparticle size was observed in the phagolysosomal compartment, indicating minimal particle erosion consistent with being localized within an intracellular microenvironment favoring particle stability. Polyanhydride microparticles increased DC surface expression of major histocompatability complex class II, the co-stimulatory molecules CD86 and CD40, and the C-type lectin CIRE (murine DC-SIGN; CD209). In addition, microparticle stimulation of DCs also enhanced secretion of the cytokines IL-12p40 and IL-6, a phenomenon found to be dependent on polymer chemistry. DCs cultured with polyanhydride microparticles and ovalbumin induced polymer chemistry-dependent antigen-specific proliferation of both CD4(+) OT-II and CD8(+) OT-I T cells. These data indicate that polyanhydride particles can be tailored to take advantage of the potential plasticity of the immune response, resulting in the ability to induce immune protection against many types of pathogens.
Collapse
|
40
|
Skendros P, Pappas G, Boura P. Cell-mediated immunity in human brucellosis. Microbes Infect 2010; 13:134-42. [PMID: 21034846 DOI: 10.1016/j.micinf.2010.10.015] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 10/13/2010] [Accepted: 10/15/2010] [Indexed: 01/18/2023]
Abstract
Brucella can parasitize within human antigen-presenting cells modifying phagocytosis, phagolysosome fusion, antigen presentation, cytokine secretion, and apoptosis. Subversion of innate immune mechanisms by Brucella leads to defective Th1 immune responses and T-cell anergy in chronic brucellosis patients. This review summarizes the cellular immune responses in brucellosis, based on data derived exclusively from human cells or cell lines.
Collapse
Affiliation(s)
- Panagiotis Skendros
- First Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece.
| | | | | |
Collapse
|
41
|
Bernardo J, Long HJ, Simons ER. Initial cytoplasmic and phagosomal consequences of human neutrophil exposure to Staphylococcus epidermidis. Cytometry A 2010; 77:243-52. [PMID: 19937952 DOI: 10.1002/cyto.a.20827] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Microorganisms are recognized by specific phagocyte surface receptors. Liganded receptors then signal a series of events leading to phagocytosis and destruction of the organism by oxidative, lytic, and associated processes. Some organisms, such as Mycobacterium tuberculosis (Mtb), Cryptococcus neoformans (Cf), and others, evade such destruction, surviving and sometimes multiplying within the phagosome to later cause disease. To study such evasion, we developed protocols which permit simultaneous kinetic measurement of early cytoplasmic signaling and of phagosomal pH (pH(p)) and oxidative burst, on a cell-by-cell basis, of polymorphonuclear (PMN) leukocytes exposed to fluorescently labeled, nonpathogenic Staphylococcus epidermidis (Se). The availability of a new, highly sensitive pH probe, pHrodo, permits observation of increasing pH(p). Simultaneous labeling of the organism, applicable to any phagocyte target, with a probe insensitive to pH and oxidative species, such as AlexaFluor350, permits distinction between binding and functional responses to it by ratioing fluorescences. Addition of an extracellular-specific quencher (Trypan blue) permits distinction between bound and phagosome-enclosed targets, so that conditions within the closed phagosome can be studied. We found that opsonization is required for functional activation of PMN by Se, that the organism causes early alkalinization of the phagosome (in contrast to Cf which hyperacidifies it), and that extracellular Ca(2+) is not required for cytoplasmic Ca(2+) signaling but contributes markedly to binding of Se to PMN and to ensuant bactericidal functions. These findings lead to a new approach to the study of select organisms, like Cf and Mtb, which evade killing by manipulating the phagosomal environment.
Collapse
Affiliation(s)
- John Bernardo
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA.
| | | | | |
Collapse
|
42
|
Abstract
Infection of cattle caused by Brucella abortus (ie, bovine brucellosis) has been of political importance in the United States for many decades. The most common clinical manifestation of brucellosis in natural hosts is reproductive loss resulting from abortion, birth of weak offspring, or infertility. Brucellosis regulatory programs were primarily developed as the most efficient way to prevent human infections. This article discusses cattle vaccination with B abortus strains 19 and RB51. Other reservoir hosts for this organism and other Brucella spp in cattle have also been visited.
Collapse
Affiliation(s)
- Steven Olsen
- Infectious Bacterial Diseases Research Unit, United States Department of Agriculture, Agricultural Research Service, National Animal Disease Center, Ames, IA 50010, USA.
| | | |
Collapse
|
43
|
Simons ER. Measurement of phagocytosis and of the phagosomal environment in polymorphonuclear phagocytes by flow cytometry. ACTA ACUST UNITED AC 2010; Chapter 9:Unit9.31. [PMID: 20069529 DOI: 10.1002/0471142956.cy0931s51] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Phagocytes are the most important early components of the immune response, programmed to recognize, engulf, and destroy immune complexes (formed when antibodies recognize their specific antigens), foreign particles, bacteria, mycobacteria, apoptotic cells, etc. Neutrophils, monocytes, macrophages, and dendritic cells all participate in this process. Flow cytometry permits observation of phagocytes that have responded and, with the appropriate fluorescent probes, of the environment in the phagosome that has enclosed the foreign matter. This unit gives the background and the protocols for performing such studies.
Collapse
|
44
|
Croxatto A, Greub G. Early intracellular trafficking of Waddlia chondrophila in human macrophages. MICROBIOLOGY-SGM 2009; 156:340-355. [PMID: 19926655 DOI: 10.1099/mic.0.034546-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Waddlia chondrophila is an obligate intracellular bacterium considered as a potential agent of abortion in both humans and bovines. This member of the order Chlamydiales multiplies rapidly within human macrophages and induces lysis of the infected cells. To understand how this Chlamydia-like micro-organism invades and proliferates within host cells, we investigated its trafficking within monocyte-derived human macrophages. Vacuoles containing W. chondrophila acquired the early endosomal marker EEA1 during the first 30 min following uptake. However, the live W. chondrophila-containing vacuoles never co-localized with late endosome and lysosome markers. Instead of interacting with the endosomal pathway, W. chondrophila immediately co-localized with mitochondria and, shortly after, with endoplasmic reticulum- (ER-) resident proteins such as calnexin and protein disulfide isomerase. The acquisition of mitochondria and ER markers corresponds to the beginning of bacterial replication. It is noteworthy that mitochondrion recruitment to W. chondrophila inclusions is prevented only by simultaneous treatment with the microtubule and actin cytoskeleton-disrupting agents nocodazole and cytochalasin D. In addition, brefeldin A inhibits the replication of W. chondrophila, supporting a role for COPI-dependent trafficking in the biogenesis of the bacterial replicating vacuole. W. chondrophila probably survives within human macrophages by evading the endocytic pathway and by associating with mitochondria and the ER. The intracellular trafficking of W. chondrophila in human macrophages represents a novel route that differs strongly from that used by other members of the order Chlamydiales.
Collapse
Affiliation(s)
- Antony Croxatto
- Center for Research on Intracellular Bacteria (CRIB), Institute of Microbiology, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Gilbert Greub
- Center for Research on Intracellular Bacteria (CRIB), Institute of Microbiology, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
45
|
Survival of the fittest: how Brucella strains adapt to their intracellular niche in the host. Med Microbiol Immunol 2009; 198:221-38. [PMID: 19830453 DOI: 10.1007/s00430-009-0123-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Indexed: 02/06/2023]
Abstract
Brucella strains produce abortion and infertility in their natural hosts and a zoonotic disease in humans known as undulant fever. These bacteria do not produce classical virulence factors, and their capacity to successfully survive and replicate within a variety of host cells underlies their pathogenicity. Extensive replication of the brucellae in placental trophoblasts is associated with reproductive tract pathology in natural hosts, and prolonged persistence in macrophages leads to the chronic infections that are a hallmark of brucellosis in both natural hosts and humans. This review describes how Brucella strains have efficiently adapted to their intracellular lifestyle in the host.
Collapse
|
46
|
Anderson ES, Paulley JT, Gaines JM, Valderas MW, Martin DW, Menscher E, Brown TD, Burns CS, Roop RM. The manganese transporter MntH is a critical virulence determinant for Brucella abortus 2308 in experimentally infected mice. Infect Immun 2009; 77:3466-74. [PMID: 19487482 PMCID: PMC2715675 DOI: 10.1128/iai.00444-09] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 05/03/2009] [Accepted: 05/20/2009] [Indexed: 11/20/2022] Open
Abstract
The gene designated BAB1_1460 in the Brucella abortus 2308 genome sequence is predicted to encode the manganese transporter MntH. Phenotypic analysis of an isogenic mntH mutant indicates that MntH is the sole high-affinity manganese transporter in this bacterium but that MntH does not play a detectable role in the transport of Fe(2+), Zn(2+), Co(2+), or Ni(2+). Consistent with the apparent selectivity of the corresponding gene product, the expression of the mntH gene in B. abortus 2308 is repressed by Mn(2+), but not Fe(2+), and this Mn-responsive expression is mediated by a Mur-like repressor. The B. abortus mntH mutant MWV15 exhibits increased susceptibility to oxidative killing in vitro compared to strain 2308, and a comparative analysis of the superoxide dismutase activities present in these two strains indicates that the parental strain requires MntH in order to make wild-type levels of its manganese superoxide dismutase SodA. The B. abortus mntH mutant also exhibits extreme attenuation in both cultured murine macrophages and experimentally infected C57BL/6 mice. These experimental findings indicate that Mn(2+) transport mediated by MntH plays an important role in the physiology of B. abortus 2308, particularly during its intracellular survival and replication in the host.
Collapse
Affiliation(s)
- Eric S Anderson
- Department of Microbiology and Immunology, East Carolina University School of Medicine, Greenville, NC 27834, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Li S, Ojcius DM, Liao S, Li L, Xue F, Dong H, Yan J. Replication or death: distinct fates of pathogenic Leptospira strain Lai within macrophages of human or mouse origin. Innate Immun 2009; 16:80-92. [PMID: 19587003 DOI: 10.1177/1753425909105580] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Pathogenic leptospires evoke severe diseases in humans but only cause mild chronic or asymptomatic infection in many host animals. The reasons for this diversity of infection remain unclear. Here, we demonstrated that Leptospira interrogans serovar Lai strain Lai had a similar ability to adhere to and enter primary and immortal (THP-1 and J774A.1) macrophages from human and mouse, but its intracellular fate in human macrophages differed markedly from that in mouse. The leptospires resided within membrane-bound vacuoles in the murine macrophages, but occurred free in the cytosol of human macrophages, with no surrounding vesicular membrane. Most leptospires in murine macrophages co-localized with the late-endosomal/lysosomal marker LAMP-1 and then were killed by lysosomal hydrolases, while most leptospires in human macrophages did not co-localize with this marker and survived. Enumeration of colony-forming units plus quantitative fluorimetry showed that in human, but not in murine, macrophages, the amounts of leptospires increased with incubation time. The infected human macrophages differed from mouse macrophages by displaying gradually enhanced apoptosis, in parallel with the increase in number of leptospires. These data strongly suggest that the outcome for intracellular leptospires depends on differences among host macrophages, which may account for some of the differences in the severity of leptospirosis in humans and animals.
Collapse
Affiliation(s)
- Shijun Li
- Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital of Zhejiang University, Hangzhou, P.R. China
| | | | | | | | | | | | | |
Collapse
|
48
|
Ulery BD, Phanse Y, Sinha A, Wannemuehler MJ, Narasimhan B, Bellaire BH. Polymer chemistry influences monocytic uptake of polyanhydride nanospheres. Pharm Res 2008; 26:683-90. [PMID: 18987960 DOI: 10.1007/s11095-008-9760-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2008] [Accepted: 10/14/2008] [Indexed: 11/26/2022]
Abstract
PURPOSE To demonstrate that polyanhydride copolymer chemistry affects the uptake and intracellular compartmentalization of nanospheres by THP-1 human monocytic cells. METHODS Polyanhydride nanospheres were prepared by an anti-solvent nanoprecipitation technique. Morphology and particle diameter were confirmed via scanning election microscopy and quasi-elastic light scattering, respectively. The effects of varying polymer chemistry on nanosphere and fluorescently labeled protein uptake by THP-1 cells were monitored by laser scanning confocal microscopy. RESULTS Polyanhydride nanoparticles composed of poly(sebacic anhydride) (SA), and 20:80 and 50:50 copolymers of 1,6-bis-(p-carboxyphenoxy)hexane (CPH) anhydride and SA were fabricated with similar spherical morphology and particle diameter (200 to 800 nm). Exposure of the nanospheres to THP-1 monocytes showed that poly(SA) and 20:80 CPH:SA nanospheres were readily internalized whereas 50:50 CPH:SA nanospheres had limited uptake. The chemistries also differentially enhanced the uptake of a red fluorescent protein-labeled antigen. CONCLUSIONS Nanosphere and antigen uptake by monocytes can be directly correlated to the chemistry of the nanosphere. These results demonstrate the importance of choosing polyanhydride chemistries that facilitate enhanced interactions with antigen presenting cells that are necessary in the initiation of efficacious immune responses.
Collapse
Affiliation(s)
- Bret D Ulery
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IOWA, 50011, USA
| | | | | | | | | | | |
Collapse
|
49
|
|
50
|
RNAi screen of endoplasmic reticulum-associated host factors reveals a role for IRE1alpha in supporting Brucella replication. PLoS Pathog 2008; 4:e1000110. [PMID: 18654626 PMCID: PMC2453327 DOI: 10.1371/journal.ppat.1000110] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Accepted: 06/24/2008] [Indexed: 11/20/2022] Open
Abstract
Brucella species are facultative intracellular bacterial pathogens that cause brucellosis, a global zoonosis of profound importance. Although recent studies have demonstrated that Brucella spp. replicate within an intracellular compartment that contains endoplasmic reticulum (ER) resident proteins, the molecular mechanisms by which the pathogen secures this replicative niche remain obscure. Here, we address this issue by exploiting Drosophila S2 cells and RNA interference (RNAi) technology to develop a genetically tractable system that recapitulates critical aspects of mammalian cell infection. After validating this system by demonstrating a shared requirement for phosphoinositide 3-kinase (PI3K) activities in supporting Brucella infection in both host cell systems, we performed an RNAi screen of 240 genes, including 110 ER-associated genes, for molecules that mediate bacterial interactions with the ER. We uncovered 52 evolutionarily conserved host factors that, when depleted, inhibited or increased Brucella infection. Strikingly, 29 of these factors had not been previously suggested to support bacterial infection of host cells. The most intriguing of these was inositol-requiring enzyme 1 (IRE1), a transmembrane kinase that regulates the eukaryotic unfolded protein response (UPR). We employed IRE1α−/− murine embryonic fibroblasts (MEFs) to demonstrate a role for this protein in supporting Brucella infection of mammalian cells, and thereby, validated the utility of the Drosophila S2 cell system for uncovering novel Brucella host factors. Finally, we propose a model in which IRE1α, and other ER-associated genes uncovered in our screen, mediate Brucella replication by promoting autophagosome biogenesis. Brucella spp. are facultative intracellular pathogens that cause brucellosis in a broad range of hosts, including humans. Brucella melitensis, B. abortus, and B. suis are highly infectious and can be readily transmitted in aerosolized form, and a human vaccine against brucellosis is unavailable. Therefore, these pathogens are recognized as potential bioterror agents. Because genetic systems for studying host–Brucella interactions have been unavailable, little is known about the host factors that mediate infection. Here, we demonstrate that a Drosophila S2 cell system and RNA interference can be exploited to study the role that evolutionarily conserved Brucella host proteins play in these processes. We also show that this system provides for the identification and characterization of host factors that mediate Brucella interactions with the host cell endoplasmic reticulum. In fact, we identified 52 host factors that, when depleted, inhibited or increased Brucella infection. Among the identified Brucella host factors, 29 have not been previously shown to support bacterial infection. Finally, we demonstrate that the novel host factor inositol-requiring enzyme 1 (IRE1) and its mammalian ortholog (IRE1α) are required for Brucella infection of Drosophila S2 and mammalian cells, respectively. Therefore, this work contributes to our understanding of host factors mediating Brucella infection.
Collapse
|