1
|
Pellegrini JM, González-Espinoza G, Shayan RR, Hysenaj L, Rouma T, Arce-Gorvel V, Lelouard H, Popoff D, Zhao Y, Hanniffy S, Castillo-Zeledón A, Loperena-Barber M, Celis-Gutierrez J, Mionnet C, Bosilkovski M, Solera J, Muraille E, Barquero-Calvo E, Moreno E, Conde-Álvarez R, Moriyón I, Gorvel JP, Mémet S. Brucella abortus impairs T lymphocyte responsiveness by mobilizing IL-1RA-secreting omental neutrophils. Nat Commun 2025; 16:862. [PMID: 39833171 DOI: 10.1038/s41467-024-55799-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/30/2024] [Indexed: 01/22/2025] Open
Abstract
Immune evasion strategies of Brucella, the etiologic agent of brucellosis, a global zoonosis, remain partially understood. The omentum, a tertiary lymphoid organ part of visceral adipose tissue, has never been explored as a Brucella reservoir. We report that B. abortus infects and replicates within murine omental macrophages. Throughout the chronic phase of infection, the omentum accumulates macrophages, monocytes and neutrophils. The maintenance of PD-L1+Sca-1+ macrophages, monocytes and neutrophils in the omentum depends on the wadC-encoded determinant of Brucella LPS. We demonstrate that PD-L1+Sca-1+ murine omental neutrophils produce high levels of IL-1RA leading to T cell hyporesponsiveness. These findings corroborate brucellosis patient analysis of whole blood displaying upregulation of PDL1 and Ly6E genes, and of serum exhibiting high levels of IL-1RA. Overall, the omentum, a reservoir for B. abortus, promotes bacterial persistence and causes CD4+ and CD8+ T cell immunosuppression by IL-1RA secreted by PD-L1+Sca-1+ neutrophils.
Collapse
Affiliation(s)
| | | | | | - Lisiena Hysenaj
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Thomas Rouma
- Unité de Recherche en Biologie des Microorganismes (URBM)-Laboratoire d'Immunologie et de Microbiologie, NARILIS, University of Namur, Namur, Belgium
- Laboratoire de de Parasitologie, and ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles, Gosselies, Belgium
| | | | - Hugues Lelouard
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Dimitri Popoff
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Yun Zhao
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Sean Hanniffy
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Amanda Castillo-Zeledón
- Universidad Nacional, Pathology Department, Escuela de Medicina Veterinaria, Heredia, Costa Rica
| | - Maite Loperena-Barber
- Universidad de Navarra, Instituto de Salud Tropical e Departamento de Microbiología y Parasitología, Pamplona, Spain
| | | | - Cyrille Mionnet
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Mile Bosilkovski
- University Clinic for Infectious Diseases and Febrile Conditions, Skopje, Republic of North Macedonia
| | - Javier Solera
- Hospital General Universitario, Facultad de Medicina, Universidad Castilla la Mancha Albacete, Albacete, Spain
| | - Eric Muraille
- Unité de Recherche en Biologie des Microorganismes (URBM)-Laboratoire d'Immunologie et de Microbiologie, NARILIS, University of Namur, Namur, Belgium
- Laboratoire de de Parasitologie, and ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles, Gosselies, Belgium
| | - Elías Barquero-Calvo
- Universidad Nacional, Pathology Department, Escuela de Medicina Veterinaria, Heredia, Costa Rica
| | - Edgardo Moreno
- Universidad Nacional, Pathology Department, Escuela de Medicina Veterinaria, Heredia, Costa Rica
| | - Raquel Conde-Álvarez
- Universidad de Navarra, Instituto de Salud Tropical e Departamento de Microbiología y Parasitología, Pamplona, Spain
| | - Ignacio Moriyón
- Universidad de Navarra, Instituto de Salud Tropical e Departamento de Microbiología y Parasitología, Pamplona, Spain
| | | | - Sylvie Mémet
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France.
| |
Collapse
|
2
|
Ketterer M, Chiquet P, Esposito M, Sedzicki J, Québatte M, Dehio C. The putative type 4 secretion system effector BspD is involved in maintaining envelope integrity of the pathogen Brucella. mSphere 2024; 9:e0023224. [PMID: 39387552 PMCID: PMC11580434 DOI: 10.1128/msphere.00232-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/05/2024] [Indexed: 10/15/2024] Open
Abstract
Brucellosis is a debilitating disease caused by the Gram-negative, facultative intracellular zoonotic pathogen Brucella. En route to its intracellular replicative niche, Brucella encounters various stressful environments that may compromise envelope integrity. Here we show that the proposed type 4 secretion system (T4SS) effector BspD is a conserved protein of the Rhizobiales, which does not show signs of co-evolution with the presence of a T4SS or a certain lifestyle. We further present data indicating that BspD is critical for the envelope integrity of Brucella abortus in the stationary phase and in the presence of EDTA, a compound known to destabilize the outer membrane. Deletion of bspD resulted in abnormal bacterial morphologies, indicating its involvement in maintaining envelope integrity. Additionally, the absence of BspD led to the formation of fewer and smaller intracellular microcolonies in a macrophage infection model. From our observations, we propose that BspD of B. abortus is critical for preserving the integrity of the bacterial envelope, particularly under stressful conditions, which may enhance Brucella's ability to survive within host cells. IMPORTANCE Brucellosis, caused by the intracellular pathogen Brucella, poses a significant health threat. Understanding how Brucella adapts to stressful environments is crucial. This study unveils BspD, a conserved protein within the Rhizobiales order, as a key player in maintaining Brucella's envelope integrity. Remarkably, BspD's presence within the Rizobiales appears independent of the presence of a T4SS or a specific lifestyle. Deletion of bspD resulted in compromised envelope integrity, abnormal bacterial morphologies, and reduced intracellular microcolony formation. These findings underscore BspD's critical role, particularly in stressful conditions like the stationary phase and EDTA exposure, and highlight its significance for the survival of Brucella within host cells. This elucidation deepens our understanding of Brucella pathogenesis and may inform future therapeutic strategies against brucellosis.
Collapse
|
3
|
Molina RE, Osorio A, Flores-Concha M, Gómez LA, Alvarado I, Ferrari I, Oñate A. Immunoinformatic design of a multivalent vaccine against Brucella abortus and its evaluation in a murine model using a DNA prime-protein boost strategy. Front Immunol 2024; 15:1456078. [PMID: 39640259 PMCID: PMC11617539 DOI: 10.3389/fimmu.2024.1456078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction The development of effective vaccines against Brucella abortus is critical due to its significant impact on human and animal health. The objective of this study was to design and evaluate in silico and in vivo a multivalent vaccine based on the immunogenic potential of three selected open reading frames (ORFs) of Brucella. Methods The designed construct, named S22, was analyzed in silico to evaluate its physicochemical properties, antigenicity, allergenicity and toxicity. This construct was modeled and subjected to molecular dynamics analysis. Additionally, the antigenicity and protection induced by this construct was evaluated through In vivo assays immunizing BALB/c mice with protein (S22), DNA (pVS22) and combining both vaccine formats using a prime boost immunization strategy. Results All bioinformatics analyses showed safe and high quality structural features, revealing favorable interactions between S22 and the TLR4/MD2 complex. Moreover, results from in vivo assays indicated that the S22 protein induced robust levels of IgG1 and IgG2a, suggesting a balanced Th1 and Th2 immune response. The DNA construct (pVS22) elicited primarily a Th1 response, whereas the use of a prime boost strategy, which combines both formats resulted in a balanced immune response with significant induction of lymphoproliferation and elevated. Discussion Although our assays did not demonstrate the induction of a substantial protective response against B. abortus, this construct was capable of inducing immunogenicity. This study highlights the utility of in silico design for predicting and optimizing candidate vaccines and underscores the potential of using strategies such as prime boost, which incorporate antigens of different biological nature to modulate the immune response, while balancing parameters such as stability of the antigens and the cost of production.
Collapse
Affiliation(s)
- Raúl E. Molina
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | | | - Manuel Flores-Concha
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Leonardo A. Gómez
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Ilse Alvarado
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Italo Ferrari
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Angel Oñate
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| |
Collapse
|
4
|
Qin Y, Zhou G, Jiao F, Cheng C, Meng C, Wang L, Wu S, Fan C, Li J, Zhou B, Chu Y, Jiao H. Brucella mediates autophagy, inflammation, and apoptosis to escape host killing. Front Cell Infect Microbiol 2024; 14:1408407. [PMID: 39507949 PMCID: PMC11537862 DOI: 10.3389/fcimb.2024.1408407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/26/2024] [Indexed: 11/08/2024] Open
Abstract
Brucellosis is a serious zoonosis caused by Brucella spp. infection, which not only seriously jeopardizes the health of humans and mammals, but also causes huge economic losses to the livestock industry. Brucella is a Gram-negative intracellular bacterium that relies primarily on its virulence factors and a variety of evolved survival strategies to replicate and proliferate within cells. Currently, the mechanisms of autophagy, inflammation, and apoptosis in Brucella-infected hosts are not fully understood and require further research and discussion. This review focuses on the relationship between Brucella and autophagy, inflammation, and apoptosis to provide the scientific basis for revealing the pathogenesis of Brucella.
Collapse
Affiliation(s)
- Yaqiong Qin
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Gengxu Zhou
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Fengyuan Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Chuan Cheng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Chi Meng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Lingjie Wang
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Shengping Wu
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Cailiang Fan
- The College of Veterinary Medicine, Southwest University, Chongqing, China
- Animal Epidemic Prevention and Control Center of Rongchang, Chongqing, China
| | - Jixiang Li
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Bo Zhou
- Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Yuefeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Hanwei Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
5
|
Loperena-Barber M, Elizalde-Bielsa A, Salvador-Bescós M, Ruiz-Rodríguez P, Pellegrini JM, Renau-Mínguez C, Lancaster R, Zúñiga-Ripa A, Iriarte M, Bengoechea JA, Coscollá M, Gorvel JP, Moriyón I, Conde-Álvarez R. "Phylogenomic insights into brucellaceae: The Pseudochrobactrum algeriensis case". INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 123:105625. [PMID: 38906517 DOI: 10.1016/j.meegid.2024.105625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
The genus Pseudochrobactrum encompasses free-living bacteria phylogenetically close to Ochrobactrum opportunistic pathogens and to Brucella, facultative intracellular parasites causing brucellosis, a worldwide-extended and grave zoonosis. Recently, Pseudochrobactrum strains were isolated from Brucella natural hosts on Brucella selective media, potentially causing diagnostic confusions. Strikingly, P. algeriensis was isolated from cattle lymph nodes, organs that are inimical to bacteria. Here, we analyse P. algeriensis potential virulence factors in comparison with Ochrobactrum and Brucella. Consistent with genomic analyses, Western-Blot analyses confirmed that P. algeriensis lacks the ability to synthesize the N-formylperosamine O-polysaccharide characteristic of the lipopolysaccharide (LPS) of smooth Brucella core species. However, unlike other Pseudochrobactrum but similar to some early diverging brucellae, P. algeriensis carries genes potentially synthetizing a rhamnose-based O-polysaccharide LPS. Lipid A analysis by MALDI-TOF demonstrated that P. algeriensis LPS bears a lipid A with a reduced pathogen-associated molecular pattern, a trait shared with Ochrobactrum and Brucella that is essential to generate a highly stable outer membrane and to delay immune activation. Also, although not able to multiply intracellularly in macrophages, the analysis of P. algeriensis cell lipid envelope revealed the presence of large amounts of cationic aminolipids, which may account for the extremely high resistance of P. algeriensis to bactericidal peptides and could favor colonization of mucosae and transient survival in Brucella hosts. However, two traits critical in Brucella pathogenicity are either significantly different (T4SS [VirB]) or absent (erythritol catabolic pathway) in P. algeriensis. This work shows that, while diverging in other characteristics, lipidic envelope features relevant in Brucella pathogenicity are conserved in Brucellaceae. The constant presence of these features strongly suggests that reinforcement of the envelope integrity as an adaptive advantage in soil was maintained in Brucella because of the similarity of some environmental challenges, such as the action of cationic peptide antibiotics and host defense peptides. This information adds knowledge about the evolution of Brucellaceae, and also underlines the taxonomical differences of the three genera compared.
Collapse
Affiliation(s)
- Maite Loperena-Barber
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Aitor Elizalde-Bielsa
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Miriam Salvador-Bescós
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Paula Ruiz-Rodríguez
- Institute for Integrative Systems Biology, Universidad de Valencia-CSIC, Valencia, Spain
| | | | - Chantal Renau-Mínguez
- Institute for Integrative Systems Biology, Universidad de Valencia-CSIC, Valencia, Spain
| | - Rebecca Lancaster
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Amaia Zúñiga-Ripa
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Maite Iriarte
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Jose A Bengoechea
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Mireia Coscollá
- Institute for Integrative Systems Biology, Universidad de Valencia-CSIC, Valencia, Spain
| | - Jean-Pierre Gorvel
- Centre d'Immunologie de Marseille-Luminy, CNRS, INSERM, Aix-Marseille University, Marseille, France
| | - Ignacio Moriyón
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Raquel Conde-Álvarez
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
6
|
Yu H, Gu X, Wang D, Wang Z. Brucella infection and Toll-like receptors. Front Cell Infect Microbiol 2024; 14:1342684. [PMID: 38533384 PMCID: PMC10963510 DOI: 10.3389/fcimb.2024.1342684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/17/2024] [Indexed: 03/28/2024] Open
Abstract
Brucella consists of gram-negative bacteria that have the ability to invade and replicate in professional and non-professional phagocytes, and its prolonged persistence in the host leads to brucellosis, a serious zoonosis. Toll-like receptors (TLRs) are the best-known sensors of microorganisms implicated in the regulation of innate and adaptive immunity. In particular, TLRs are transmembrane proteins with a typical structure of an extracellular leucine-rich repeat (LRR) region and an intracellular Toll/interleukin-1 receptor (TIR) domain. In this review, we discuss Brucella infection and the aspects of host immune responses induced by pathogens. Furthermore, we summarize the roles of TLRs in Brucella infection, with substantial emphasis on the molecular insights into its mechanisms of action.
Collapse
Affiliation(s)
- Hui Yu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Xinyi Gu
- The College of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Danfeng Wang
- The College of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| |
Collapse
|
7
|
Flores-Concha M, Gómez LA, Soto-Shara R, Molina RE, Coloma-Rivero RF, Montero DA, Ferrari Í, Oñate Á. Brucella abortus triggers the differential expression of immunomodulatory lncRNAs in infected murine macrophages. Front Immunol 2024; 15:1352306. [PMID: 38464511 PMCID: PMC10921354 DOI: 10.3389/fimmu.2024.1352306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/30/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction The lncRNAs (long non-coding RNAs) are the most diverse group of non-coding RNAs and are involved in most biological processes including the immune response. While some of them have been recognized for their influence on the regulation of inflammatory activity, little is known in the context of infection by Brucella abortus, a pathogen that presents significant challenges due to its ability to manipulate and evade the host immune system. This study focuses on characterize the expression profile of LincRNA-cox2, Lethe, lincRNA-EPS, Malat1 and Gas5 during infection of macrophages by B. abortus. Methods Using public raw RNA-seq datasets we constructed for a lncRNA expression profile in macrophages Brucella-infected. In addition, from public RNA-seq raw datasets of RAW264.7 cells infected with B. abortus we constructed a transcriptomic profile of lncRNAs in order to know the expression of the five immunomodulating lncRNAs studied here at 8 and 24 h post-infection. Finally, we performed in vitro infection assays in RAW264.7 cells and peritoneal macrophages to detect by qPCR changes in the expression of these lncRNAs at first 12 hours post infection, a key stage in the infection cycle where Brucella modulates the immune response to survive. Results Our results demonstrate that infection of macrophages with Brucella abortus, induces significant changes in the expression of LincRNA-Cox2, Lethe, LincRNA-EPS, Gas5, and Malat1. Discussion The change in the expression profile of these immunomodulatory lncRNAs in response to infection, suggest a potential involvement in the immune evasion strategy employed by Brucella to facilitate its intracellular survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ángel Oñate
- Laboratory of Molecular Immunology, Department of Microbiology, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
8
|
Alakavuklar MA, Fiebig A, Crosson S. The Brucella Cell Envelope. Annu Rev Microbiol 2023; 77:233-253. [PMID: 37104660 PMCID: PMC10787603 DOI: 10.1146/annurev-micro-032521-013159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
The cell envelope is a multilayered structure that insulates the interior of bacterial cells from an often chaotic outside world. Common features define the envelope across the bacterial kingdom, but the molecular mechanisms by which cells build and regulate this critical barrier are diverse and reflect the evolutionary histories of bacterial lineages. Intracellular pathogens of the genus Brucella exhibit marked differences in cell envelope structure, regulation, and biogenesis when compared to more commonly studied gram-negative bacteria and therefore provide an excellent comparative model for study of the gram-negative envelope. We review distinct features of the Brucella envelope, highlighting a conserved regulatory system that links cell cycle progression to envelope biogenesis and cell division. We further discuss recently discovered structural features of the Brucella envelope that ensure envelope integrity and that facilitate cell survival in the face of host immune stressors.
Collapse
Affiliation(s)
- Melene A Alakavuklar
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA;
| | - Aretha Fiebig
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA;
| | - Sean Crosson
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA;
| |
Collapse
|
9
|
Hao M, Wang M, Zhao D, Shi Y, Yuan Y, Li J, Zhai Y, Liu X, Zhou D, Chen H, Lin P, Tang K, Liu W, Jin Y, Wang A. Alr Gene in Brucella suis S2: Its Role in Lipopolysaccharide Biosynthesis and Bacterial Virulence in RAW264.7. Int J Mol Sci 2023; 24:10744. [PMID: 37445922 DOI: 10.3390/ijms241310744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/24/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Brucella suis, the causative agent of brucellosis, poses a significant public health and animal husbandry threat. However, the role of the alanine racemase (alr) gene, which encodes alanine racemase in Brucella, remains unclear. Here, we analyzed an alr deletion mutant and a complemented strain of Brucella suis S2. The knockout strain displayed an unaltered, smooth phenotype in acriflavine agglutination tests but lacked the core polysaccharide portion of lipopolysaccharide (LPS). Genes involved in the LPS synthesis were significantly upregulated in the deletion mutant. The alr deletion strain exhibited reduced intracellular viability in the macrophages, increased macrophage-mediated killing, and upregulation of the apoptosis markers. Bcl2, an anti-apoptotic protein, was downregulated, while the pro-apoptotic proteins, Bax, Caspase-9, and Caspase-3, were upregulated in the macrophages infected with the deletion strain. The infected macrophages showed increased mitochondrial membrane permeability, Cytochrome C release, and reactive oxygen species, activating the mitochondrial apoptosis pathway. These findings revealed that alanine racemase was dispensable in B. suis S2 but influenced the strain's rough features and triggered the mitochondrial apoptosis pathway during macrophage invasion. The deletion of the alr gene reduced the intracellular survival and virulence. This study enhances our understanding of the molecular mechanism underlying Brucella's survival and virulence and, specifically, how alr gene affects host immune evasion by regulating bacterial LPS biosynthesis.
Collapse
Affiliation(s)
- Mingyue Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Minghui Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Danyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Yong Shi
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Ye Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Junmei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Yunyi Zhai
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Xiaofang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Dong Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Huatao Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Pengfei Lin
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Keqiong Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Wei Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Yaping Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| | - Aihua Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling District, Xianyang 712100, China
| |
Collapse
|
10
|
Bányász B, Antal J, Dénes B. False Positives in Brucellosis Serology: Wrong Bait and Wrong Pond? Trop Med Infect Dis 2023; 8:tropicalmed8050274. [PMID: 37235322 DOI: 10.3390/tropicalmed8050274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
This review summarizes the status of resolving the problem of false positive serologic results (FPSR) in Brucella serology, compiles our knowledge on the molecular background of the problem, and highlights some prospects for its resolution. The molecular basis of the FPSRs is reviewed through analyzing the components of the cell wall of Gram-negative bacteria, especially the surface lipopolysaccharide (LPS) with details related to brucellae. After evaluating the efforts that have been made to solve target specificity problems of serologic tests, the following conclusions can be drawn: (i) resolving the FPSR problem requires a deeper understanding than we currently possess, both of Brucella immunology and of the current serology tests; (ii) the practical solutions will be as expensive as the related research; and (iii) the root cause of FPSRs is the application of the same type of antigen (S-type LPS) in the currently approved tests. Thus, new approaches are necessary to resolve the problems stemming from FPSR. Such approaches suggested by this paper are: (i) the application of antigens from R-type bacteria; or (ii) the further development of specific brucellin-based skin tests; or (iii) the application of microbial cell-free DNA as analyte, whose approach is detailed in this paper.
Collapse
Affiliation(s)
- Borbála Bányász
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine Budapest, 1143 Budapest, Hungary
- Laboratory of Immunology, Veterinary Diagnostic Directorate, National Food Chain Safety Office, 1143 Budapest, Hungary
| | - József Antal
- Omixon Biocomputing Ltd., 1117 Budapest, Hungary
| | - Béla Dénes
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine Budapest, 1143 Budapest, Hungary
| |
Collapse
|
11
|
English BC, Savage HP, Mahan SP, Diaz-Ochoa VE, Young BM, Abuaita BH, Sule G, Knight JS, O’Riordan MX, Bäumler AJ, Tsolis RM. The IRE1α-XBP1 Signaling Axis Promotes Glycolytic Reprogramming in Response to Inflammatory Stimuli. mBio 2023; 14:e0306822. [PMID: 36475773 PMCID: PMC9973330 DOI: 10.1128/mbio.03068-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 12/14/2022] Open
Abstract
Immune cells must be able to adjust their metabolic programs to effectively carry out their effector functions. Here, we show that the endoplasmic reticulum (ER) stress sensor Inositol-requiring enzyme 1 alpha (IRE1α) and its downstream transcription factor X box binding protein 1 (XBP1) enhance the upregulation of glycolysis in classically activated macrophages (CAMs). The IRE1α-XBP1 signaling axis supports this glycolytic switch in macrophages when activated by lipopolysaccharide (LPS) stimulation or infection with the intracellular bacterial pathogen Brucella abortus. Importantly, these different inflammatory stimuli have distinct mechanisms of IRE1α activation; while Toll-like receptor 4 (TLR4) supports glycolysis under both conditions, TLR4 is required for activation of IRE1α in response to LPS treatment but not B. abortus infection. Though IRE1α and XBP1 are necessary for maximal induction of glycolysis in CAMs, activation of this pathway is not sufficient to increase the glycolytic rate of macrophages, indicating that the cellular context in which this pathway is activated ultimately dictates the cell's metabolic response and that IRE1α activation may be a way to fine-tune metabolic reprogramming. IMPORTANCE The immune system must be able to tailor its response to different types of pathogens in order to eliminate them and protect the host. When confronted with bacterial pathogens, macrophages, frontline defenders in the immune system, switch to a glycolysis-driven metabolism to carry out their antibacterial functions. Here, we show that IRE1α, a sensor of ER stress, and its downstream transcription factor XBP1 support glycolysis in macrophages during infection with Brucella abortus or challenge with Salmonella LPS. Interestingly, these stimuli activate IRE1α by independent mechanisms. While the IRE1α-XBP1 signaling axis promotes the glycolytic switch, activation of this pathway is not sufficient to increase glycolysis in macrophages. This study furthers our understanding of the pathways that drive macrophage immunometabolism and highlights a new role for IRE1α and XBP1 in innate immunity.
Collapse
Affiliation(s)
- Bevin C. English
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Hannah P. Savage
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Scott P. Mahan
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Vladimir E. Diaz-Ochoa
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Briana M. Young
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Basel H. Abuaita
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Gautam Sule
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mary X. O’Riordan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| | - Renée M. Tsolis
- Department of Medical Microbiology and Immunology, University of California—Davis, Davis, California, USA
| |
Collapse
|
12
|
Servais C, Vassen V, Verhaeghe A, Küster N, Carlier E, Phégnon L, Mayard A, Auberger N, Vincent S, De Bolle X. Lipopolysaccharide biosynthesis and traffic in the envelope of the pathogen Brucella abortus. Nat Commun 2023; 14:911. [PMID: 36806059 PMCID: PMC9938171 DOI: 10.1038/s41467-023-36442-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 01/31/2023] [Indexed: 02/19/2023] Open
Abstract
Lipopolysaccharide is essential for most Gram-negative bacteria as it is a main component of the outer membrane. In the pathogen Brucella abortus, smooth lipopolysaccharide containing the O-antigen is required for virulence. Being part of the Rhizobiales, Brucella spp. display unipolar growth and lipopolysaccharide was shown to be incorporated at the active growth sites, i.e. the new pole and the division site. By localizing proteins involved in the lipopolysaccharide transport across the cell envelope, from the inner to the outer membrane, we show that the lipopolysaccharide incorporation sites are determined by the inner membrane complex of the lipopolysaccharide transport system. Moreover, we identify the main O-antigen ligase of Brucella spp. involved in smooth lipopolysaccharide synthesis. Altogether, our data highlight a layer of spatiotemporal organization of the lipopolysaccharide biosynthesis pathway and identify an original class of bifunctional O-antigen ligases.
Collapse
Affiliation(s)
- Caroline Servais
- Research Unit in Biology of Microorganisms (URBM), Narilis, University of Namur (UNamur), 61 rue de Bruxelles, 5000, Namur, Belgium
| | - Victoria Vassen
- Research Unit in Biology of Microorganisms (URBM), Narilis, University of Namur (UNamur), 61 rue de Bruxelles, 5000, Namur, Belgium
| | - Audrey Verhaeghe
- Research Unit in Biology of Microorganisms (URBM), Narilis, University of Namur (UNamur), 61 rue de Bruxelles, 5000, Namur, Belgium
| | - Nina Küster
- Research Unit in Biology of Microorganisms (URBM), Narilis, University of Namur (UNamur), 61 rue de Bruxelles, 5000, Namur, Belgium
| | - Elodie Carlier
- Research Unit in Biology of Microorganisms (URBM), Narilis, University of Namur (UNamur), 61 rue de Bruxelles, 5000, Namur, Belgium
| | - Léa Phégnon
- Research Unit in Biology of Microorganisms (URBM), Narilis, University of Namur (UNamur), 61 rue de Bruxelles, 5000, Namur, Belgium
| | - Aurélie Mayard
- Research Unit in Biology of Microorganisms (URBM), Narilis, University of Namur (UNamur), 61 rue de Bruxelles, 5000, Namur, Belgium
| | - Nicolas Auberger
- Université de Poitiers, IC2MP, UMR CNRS 7285, Equipe "OrgaSynth", Groupe Glycochimie, 4 rue Michel Brunet, 86073, Poitiers, France
| | - Stéphane Vincent
- Bio-organic Chemistry Unit (CBO), Narilis, University of Namur (UNamur), 61 rue de Bruxelles, 5000, Namur, Belgium
| | - Xavier De Bolle
- Research Unit in Biology of Microorganisms (URBM), Narilis, University of Namur (UNamur), 61 rue de Bruxelles, 5000, Namur, Belgium.
| |
Collapse
|
13
|
Pascual DW, Goodwin ZI, Bhagyaraj E, Hoffman C, Yang X. Activation of mucosal immunity as a novel therapeutic strategy for combating brucellosis. Front Microbiol 2022; 13:1018165. [PMID: 36620020 PMCID: PMC9814167 DOI: 10.3389/fmicb.2022.1018165] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Brucellosis is a disease of livestock that is commonly asymptomatic until an abortion occurs. Disease in humans results from contact of infected livestock or consumption of contaminated milk or meat. Brucella zoonosis is primarily caused by one of three species that infect livestock, Bacillus abortus in cattle, B. melitensis in goats and sheep, and B. suis in pigs. To aid in disease prophylaxis, livestock vaccines are available, but are only 70% effective; hence, improved vaccines are needed to mitigate disease, particularly in countries where disease remains pervasive. The absence of knowing which proteins confer complete protection limits development of subunit vaccines. Instead, efforts are focused on developing new and improved live, attenuated Brucella vaccines, since these mimic attributes of wild-type Brucella, and stimulate host immune, particularly T helper 1-type responses, required for protection. In considering their development, the new mutants must address Brucella's defense mechanisms normally active to circumvent host immune detection. Vaccination approaches should also consider mode and route of delivery since disease transmission among livestock and humans is believed to occur via the naso-oropharyngeal tissues. By arming the host's mucosal immune defenses with resident memory T cells (TRMs) and by expanding the sources of IFN-γ, brucellae dissemination from the site of infection to systemic tissues can be prevented. In this review, points of discussion focus on understanding the various immune mechanisms involved in disease progression and which immune players are important in fighting disease.
Collapse
|
14
|
The regulon of Brucella abortus two-component system BvrR/BvrS reveals the coordination of metabolic pathways required for intracellular life. PLoS One 2022; 17:e0274397. [PMID: 36129877 PMCID: PMC9491525 DOI: 10.1371/journal.pone.0274397] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/26/2022] [Indexed: 11/19/2022] Open
Abstract
Brucella abortus is a facultative intracellular pathogen causing a severe zoonotic disease worldwide. The two-component regulatory system (TCS) BvrR/BvrS of B. abortus is conserved in members of the Alphaproteobacteria class. It is related to the expression of genes required for host interaction and intracellular survival. Here we report that bvrR and bvrS are part of an operon composed of 16 genes encoding functions related to nitrogen metabolism, DNA repair and recombination, cell cycle arrest, and stress response. Synteny of this genomic region within close Alphaproteobacteria members suggests a conserved role in coordinating the expression of carbon and nitrogen metabolic pathways. In addition, we performed a ChIP-Seq analysis after exposure of bacteria to conditions that mimic the intracellular environment. Genes encoding enzymes at metabolic crossroads of the pentose phosphate shunt, gluconeogenesis, cell envelope homeostasis, nucleotide synthesis, cell division, and virulence are BvrR/BvrS direct targets. A 14 bp DNA BvrR binding motif was found and investigated in selected gene targets such as virB1, bvrR, pckA, omp25, and tamA. Understanding gene expression regulation is essential to elucidate how Brucella orchestrates a physiological response leading to a furtive pathogenic strategy.
Collapse
|
15
|
Mena-Bueno S, Poveda-Urkixo I, Irazoki O, Palacios L, Cava F, Zabalza-Baranguá A, Grilló MJ. Brucella melitensis Wzm/Wzt System: Changes in the Bacterial Envelope Lead to Improved Rev1Δwzm Vaccine Properties. Front Microbiol 2022; 13:908495. [PMID: 35875565 PMCID: PMC9306315 DOI: 10.3389/fmicb.2022.908495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
The lipopolysaccharide (LPS) O-polysaccharide (O-PS) is the main virulence factor in Brucella. After synthesis in the cytoplasmic membrane, O-PS is exported to the periplasm by the Wzm/Wzt system, where it is assembled into a LPS. This translocation also engages a bactoprenol carrier required for further biosynthesis pathways, such as cell wall biogenesis. Targeting O-PS export by blockage holds great potential for vaccine development, but little is known about the biological implications of each Wzm/Wzt moiety. To improve this knowledge and to elucidate its potential application as a vaccine, we constructed and studied wzm/wzt single- and double-deletion mutants, using the attenuated strain Brucella melitensis Rev1 as the parental strain. This allowed us to describe the composition of Brucella peptidoglycan for the first time. We observed that these mutants lack external O-PS yet trigger changes in genetic transcription and in phenotypic properties associated with the outer membrane and cell wall. The three mutants are highly attenuated; unexpectedly, Rev1Δwzm also excels as an immunogenic and effective vaccine against B. melitensis and Brucella ovis in mice, revealing that low persistence is not at odds with efficacy. Rev1Δwzm is attenuated in BeWo trophoblasts, does not infect mouse placentas, and is safe in pregnant ewes. Overall, these attributes and the minimal serological interference induced in sheep make Rev1Δwzm a highly promising vaccine candidate.
Collapse
Affiliation(s)
- Sara Mena-Bueno
- Animal Health Department, Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Pamplona, Spain
- Agronomy, Biotecnology and Food Department, Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Irati Poveda-Urkixo
- Animal Health Department, Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Pamplona, Spain
| | - Oihane Irazoki
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Leyre Palacios
- Animal Health Department, Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Pamplona, Spain
| | - Felipe Cava
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Ana Zabalza-Baranguá
- Animal Health Department, Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Pamplona, Spain
| | - María Jesús Grilló
- Animal Health Department, Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Pamplona, Spain
- *Correspondence: María Jesús Grilló,
| |
Collapse
|
16
|
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
|
17
|
Moreno E, Blasco JM, Letesson JJ, Gorvel JP, Moriyón I. Pathogenicity and Its Implications in Taxonomy: The Brucella and Ochrobactrum Case. Pathogens 2022; 11:377. [PMID: 35335701 PMCID: PMC8954888 DOI: 10.3390/pathogens11030377] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/09/2022] [Accepted: 03/16/2022] [Indexed: 11/21/2022] Open
Abstract
The intracellular pathogens of the genus Brucella are phylogenetically close to Ochrobactrum, a diverse group of free-living bacteria with a few species occasionally infecting medically compromised patients. A group of taxonomists recently included all Ochrobactrum organisms in the genus Brucella based on global genome analyses and alleged equivalences with genera such as Mycobacterium. Here, we demonstrate that such equivalencies are incorrect because they overlook the complexities of pathogenicity. By summarizing Brucella and Ochrobactrum divergences in lifestyle, structure, physiology, population, closed versus open pangenomes, genomic traits, and pathogenicity, we show that when they are adequately understood, they are highly relevant in taxonomy and not unidimensional quantitative characters. Thus, the Ochrobactrum and Brucella differences are not limited to their assignments to different "risk-groups", a biologically (and hence, taxonomically) oversimplified description that, moreover, does not support ignoring the nomen periculosum rule, as proposed. Since the epidemiology, prophylaxis, diagnosis, and treatment are thoroughly unrelated, merging free-living Ochrobactrum organisms with highly pathogenic Brucella organisms brings evident risks for veterinarians, medical doctors, and public health authorities who confront brucellosis, a significant zoonosis worldwide. Therefore, from taxonomical and practical standpoints, the Brucella and Ochrobactrum genera must be maintained apart. Consequently, we urge researchers, culture collections, and databases to keep their canonical nomenclature.
Collapse
Affiliation(s)
- Edgardo Moreno
- Escuela de Medicina Veterinaria, Universidad Nacional, Heredia 40101, Costa Rica
| | - José María Blasco
- Centro de Investigación y Tecnología Agroalimentaria, Instituto Agroalimentario de Aragón, Universidad de Zaragoza, 50059 Zaragoza, Spain;
| | - Jean Jacques Letesson
- Unité de Recherche en Biologie des Microorganismes, Faculty of Science, University of Namur, 5000 Namur, Belgium;
| | - Jean Pierre Gorvel
- Centre d’Immunologie de Marseille-Luminy, Aix-Marseille Université, CNRS, INSERM, CIML, 13009 Marseille, France
| | - Ignacio Moriyón
- Instituto de Salud Tropical y Departamento de Microbiología y Parasitología, Universidad de Navarra, 31008 Pamplona, Spain;
| |
Collapse
|
18
|
Muñoz PM, Conde-Álvarez R, Andrés-Barranco S, de Miguel MJ, Zúñiga-Ripa A, Aragón-Aranda B, Salvador-Bescós M, Martínez-Gómez E, Iriarte M, Barberán M, Vizcaíno N, Moriyón I, Blasco JM. A Brucella melitensis H38ΔwbkF rough mutant protects against Brucella ovis in rams. Vet Res 2022; 53:16. [PMID: 35236406 PMCID: PMC8889640 DOI: 10.1186/s13567-022-01034-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/20/2022] [Indexed: 01/05/2023] Open
Abstract
Brucella melitensis and Brucella ovis are gram-negative pathogens of sheep that cause severe economic losses and, although B. ovis is non-zoonotic, B. melitensis is the main cause of human brucellosis. B. melitensis carries a smooth (S) lipopolysaccharide (LPS) with an N-formyl-perosamine O-polysaccharide (O-PS) that is absent in the rough LPS of B. ovis. Their control and eradication require vaccination, but B. melitensis Rev 1, the only vaccine available, triggers anti-O-PS antibodies that interfere in the S-brucellae serodiagnosis. Since eradication and serological surveillance of the zoonotic species are priorities, Rev 1 is banned once B. melitensis is eradicated or where it never existed, hampering B. ovis control and eradication. To develop a B. ovis specific vaccine, we investigated three Brucella live vaccine candidates lacking N-formyl-perosamine O-PS: Bov::CAΔwadB (CO2-independent B. ovis with truncated LPS core oligosaccharide); Rev1::wbdRΔwbkC (carrying N-acetylated O-PS); and H38ΔwbkF (B. melitensis rough mutant with intact LPS core). After confirming their attenuation and protection against B. ovis in mice, were tested in rams for efficacy. H38ΔwbkF yielded similar protection to Rev 1 against B. ovis but Bov::CAΔwadB and Rev1::wbdRΔwbkC conferred no or poor protection, respectively. All H38ΔwbkF vaccinated rams developed a protracted antibody response in ELISA and immunoprecipitation B. ovis diagnostic tests. In contrast, all remained negative in Rose Bengal and complement fixation tests used routinely for B. melitensis diagnosis, though some became positive in S-LPS ELISA owing to LPS core epitope reactivity. Thus, H38ΔwbkF is an interesting candidate for the immunoprophylaxis of B. ovis in B. melitensis-free areas.
Collapse
Affiliation(s)
- Pilar M Muñoz
- Departamento de Ciencia Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, Spain.
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, Spain.
| | - Raquel Conde-Álvarez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Sara Andrés-Barranco
- Departamento de Ciencia Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, Spain
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, Spain
| | - María-Jesús de Miguel
- Departamento de Ciencia Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, Spain
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, Spain
| | - Amaia Zúñiga-Ripa
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Beatriz Aragón-Aranda
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Miriam Salvador-Bescós
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Estrella Martínez-Gómez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
- Otology and Neurotology Group CTS495, Department of Genomic Medicine, GENYO Centre for Genomics and Oncological Research, Pfizer-University of Granada-Junta de Andalucía, Granada, Spain
| | - Maite Iriarte
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | | | - Nieves Vizcaíno
- Departamento de Microbiología y Genética, Universidad de Salamanca, Salamanca, Spain
| | - Ignacio Moriyón
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - José M Blasco
- Departamento de Ciencia Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, Spain
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, Spain
| |
Collapse
|
19
|
Francisco S, Billod JM, Merino J, Punzón C, Gallego A, Arranz A, Martin-Santamaria S, Fresno M. Induction of TLR4/TLR2 Interaction and Heterodimer Formation by Low Endotoxic Atypical LPS. Front Immunol 2022; 12:748303. [PMID: 35140704 PMCID: PMC8818788 DOI: 10.3389/fimmu.2021.748303] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/27/2021] [Indexed: 12/01/2022] Open
Abstract
The Toll-like receptor 4 (TLR4)/myeloid differentiation protein-2 (MD-2) complex is considered the major receptor of the innate immune system to recognize lipopolysaccharides (LPSs). However, some atypical LPSs with different lipid A and core saccharide moiety structures and compositions than the well-studied enterobacterial LPSs can induce a TLR2-dependent response in innate immune cells. Ochrobactrum intermedium, an opportunistic pathogen, presents an atypical LPS. In this study, we found that O. intermedium LPS exhibits a weak inflammatory activity compared to Escherichia coli LPS and, more importantly, is a specific TLR4/TLR2 agonist, able to signal through both receptors. Molecular docking analysis of O. intermedium LPS predicts a favorable formation of a TLR2/TLR4/MD-2 heterodimer complex, which was experimentally confirmed by fluorescence resonance energy transfer (FRET) in cells. Interestingly, the core saccharide plays an important role in this interaction. This study reveals for the first time TLR4/TLR2 heterodimerization that is induced by atypical LPS and may help to escape from recognition by the innate immune system.
Collapse
Affiliation(s)
- Sara Francisco
- Diomune S. L., Parque Científico de Madrid, Madrid, Spain
- Department of Molecular Biology, Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Cientificas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| | - Jean-Marc Billod
- Department of Structural Biology, Centro de Investigaciones Biologicas “Margarita Salas”, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid, Spain
| | - Javier Merino
- Diomune S. L., Parque Científico de Madrid, Madrid, Spain
- Department of Molecular Biology, Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Cientificas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Punzón
- Diomune S. L., Parque Científico de Madrid, Madrid, Spain
| | - Alicia Gallego
- Department of Molecular Biology, Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Cientificas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| | - Alicia Arranz
- Department of Molecular Biology, Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Cientificas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| | - Sonsoles Martin-Santamaria
- Department of Structural Biology, Centro de Investigaciones Biologicas “Margarita Salas”, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid, Spain
| | - Manuel Fresno
- Diomune S. L., Parque Científico de Madrid, Madrid, Spain
- Department of Molecular Biology, Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Cientificas (CSIC)-Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
20
|
Undercover Agents of Infection: The Stealth Strategies of T4SS-Equipped Bacterial Pathogens. Toxins (Basel) 2021; 13:toxins13100713. [PMID: 34679006 PMCID: PMC8539587 DOI: 10.3390/toxins13100713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 11/16/2022] Open
Abstract
Intracellular bacterial pathogens establish their replicative niches within membrane-encompassed compartments, called vacuoles. A subset of these bacteria uses a nanochannel called the type 4 secretion system (T4SS) to inject effector proteins that subvert the host cell machinery and drive the biogenesis of these compartments. These bacteria have also developed sophisticated ways of altering the innate immune sensing and response of their host cells, which allow them to cause long-lasting infections and chronic diseases. This review covers the mechanisms employed by intravacuolar pathogens to escape innate immune sensing and how Type 4-secreted bacterial effectors manipulate host cell mechanisms to allow the persistence of bacteria.
Collapse
|
21
|
Sun HL, Du XF, Tang YX, Li GQ, Yang SY, Wang LH, Li XW, Ma CJ, Jiang RM. Impact of immune checkpoint molecules on FoxP3 + Treg cells and related cytokines in patients with acute and chronic brucellosis. BMC Infect Dis 2021; 21:1025. [PMID: 34592958 PMCID: PMC8482665 DOI: 10.1186/s12879-021-06730-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
Background The immunoregulatory functions of regulatory T cells (Tregs) in the development and progression of some chronic infectious diseases are mediated by immune checkpoint molecules and immunosuppressive cytokines. However, little is known about the immunosuppressive functions of Tregs in human brucellosis, which is a major burden in low-income countries. In this study, expressions of immune checkpoint molecules and Treg-related cytokines in patients with acute and chronic Brucella infection were evaluated to explore their impact at different stages of infection. Methods Forty patients with acute brucellosis and 19 patients with chronic brucellosis admitted to the Third People’s Hospital of Linfen in Shanxi Province between August 2016 and November 2017 were enrolled. Serum and peripheral blood mononuclear cells were isolated from patients before antibiotic treatment and from 30 healthy subjects. The frequency of Tregs (CD4+ CD25+ FoxP3+ T cells) and expression of CTLA-4, GITR, and PD-1 on Treg cells were detected by flow cytometry. Levels of Treg-related cytokines, including IL-35, TGF-β1, and IL-10, were measured by customised multiplex cytokine assays using the Luminex platform. Results The frequency of Tregs was higher in chronic patients than in healthy controls (P = 0.026) and acute patients (P = 0.042); The frequency of CTLA-4+ Tregs in chronic patients was significantly higher than that in healthy controls (P = 0.011). The frequencies of GITR+ and PD-1+ Tregs were significantly higher in acute and chronic patients than in healthy controls (P < 0.05), with no significant difference between the acute and chronic groups (all P > 0.05). Serum TGF-β1 levels were higher in chronic patients (P = 0.029) and serum IL-10 levels were higher in acute patients (P = 0.033) than in healthy controls. We detected weak correlations between serum TGF-β1 levels and the frequencies of Tregs (R = 0.309, P = 0.031) and CTLA-4+ Tregs (R = 0.302, P = 0.035). Conclusions Treg cell immunity is involved in the chronicity of Brucella infection and indicates the implication of Tregs in the prognosis of brucellosis. CTLA-4 and TGF-β1 may contribute to Tregs-mediated immunosuppression in the chronic infection stage of a Brucella infection.
Collapse
Affiliation(s)
- Hua-Li Sun
- Department of Infectious Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiu-Fang Du
- Department of Infectious Diseases, The Third People's Hospital of Linfen City, Linfen, Shanxi, China
| | - Yun-Xia Tang
- The Laboratory of Infectious Diseases Centre, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Guo-Qiang Li
- Department of Laboratory Medicine, The Third People's Hospital of Linfen City, Linfen, Shanxi, China
| | - Si-Yuan Yang
- The Laboratory of Infectious Diseases Centre, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ling-Hang Wang
- The Laboratory of Infectious Diseases Centre, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xing-Wang Li
- Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Cheng-Jie Ma
- The Laboratory of Infectious Diseases Centre, Beijing Ditan Hospital, Capital Medical University, Beijing, China.
| | - Rong-Meng Jiang
- Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
22
|
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
|
23
|
Stranahan LW, Arenas-Gamboa AM. When the Going Gets Rough: The Significance of Brucella Lipopolysaccharide Phenotype in Host-Pathogen Interactions. Front Microbiol 2021; 12:713157. [PMID: 34335551 PMCID: PMC8319746 DOI: 10.3389/fmicb.2021.713157] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/22/2021] [Indexed: 01/18/2023] Open
Abstract
Brucella is a facultatively intracellular bacterial pathogen and the cause of worldwide zoonotic infections, infamous for its ability to evade the immune system and persist chronically within host cells. Despite the frequent association with attenuation in other Gram-negative bacteria, a rough lipopolysaccharide phenotype is retained by Brucella canis and Brucella ovis, which remain fully virulent in their natural canine and ovine hosts, respectively. While these natural rough strains lack the O-polysaccharide they, like their smooth counterparts, are able to evade and manipulate the host immune system by exhibiting low endotoxic activity, resisting destruction by complement and antimicrobial peptides, entering and trafficking within host cells along a similar pathway, and interfering with MHC-II antigen presentation. B. canis and B. ovis appear to have compensated for their roughness by alterations to their outer membrane, especially in regards to outer membrane proteins. B. canis, in particular, also shows evidence of being less proinflammatory in vivo, suggesting that the rough phenotype may be associated with an enhanced level of stealth that could allow these pathogens to persist for longer periods of time undetected. Nevertheless, much additional work is required to understand the correlates of immune protection against the natural rough Brucella spp., a critical step toward development of much-needed vaccines. This review will highlight the significance of rough lipopolysaccharide in the context of both natural disease and host–pathogen interactions with an emphasis on natural rough Brucella spp. and the implications for vaccine development.
Collapse
Affiliation(s)
- Lauren W Stranahan
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Angela M Arenas-Gamboa
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
24
|
Xia P, Wu Y, Lian S, Yan L, Meng X, Duan Q, Zhu G. Research progress on Toll-like receptor signal transduction and its roles in antimicrobial immune responses. Appl Microbiol Biotechnol 2021; 105:5341-5355. [PMID: 34180006 PMCID: PMC8236385 DOI: 10.1007/s00253-021-11406-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/07/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022]
Abstract
When microorganisms invade a host, the innate immune system first recognizes the pathogen-associated molecular patterns of these microorganisms through pattern recognition receptors (PRRs). Toll-like receptors (TLRs) are known transmembrane PRRs existing in both invertebrates and vertebrates. Upon ligand recognition, TLRs initiate a cascade of signaling events; promote the pro-inflammatory cytokine, type I interferon, and chemokine expression; and play an essential role in the modulation of the host's innate and adaptive immunity. Therefore, it is of great significance to improve our understanding of antimicrobial immune responses by studying the role of TLRs and their signal molecules in the host's defense against invading microbes. This paper aims to summarize the specificity of TLRs in recognition of conserved microbial components, such as lipoprotein, lipopolysaccharide, flagella, endosomal nucleic acids, and other bioactive metabolites derived from microbes. This set of interactions helps to elucidate the immunomodulatory effect of TLRs and the signal transduction changes involved in the infectious process and provide a novel therapeutic strategy to combat microbial infections.
Collapse
Affiliation(s)
- Pengpeng Xia
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Yunping Wu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Siqi Lian
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Li Yan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Xia Meng
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Qiangde Duan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Guoqiang Zhu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| |
Collapse
|
25
|
Yan X, Hu S, Yang Y, Xu D, Liu W, Li G, Cai W, Bu Z. Proteomics Investigation of the Time Course Responses of RAW264.7 Macrophages to Infections With the Wild-Type and Twin-Arginine Translocation Mutant Strains of Brucella melitensis. Front Cell Infect Microbiol 2021; 11:679571. [PMID: 34195100 PMCID: PMC8238042 DOI: 10.3389/fcimb.2021.679571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
Brucella, a notorious intracellular pathogen, causes chronic infections in many mammals, including humans. The twin-arginine translocation (Tat) pathway transports folded proteins across the cytoplasmic membrane; protein substrates translocated by Brucella include ABC transporters, oxidoreductases, and cell envelope biosynthesis proteins. Previously, we showed that a Tat mutant of Brucella melitensis M28 exhibits reduced survival within murine macrophages. In this study, we compared the host responses elicited by wild-type M28 and its Tat-mutant strains ex vivo. We utilized label-free quantitative proteomics to assess proteomic changes in RAW264.7 macrophages after infection with M28 and its Tat mutants. A total of 6085 macrophage proteins were identified with high confidence, and 79, 50, and 99 proteins were differentially produced upon infection with the Tat mutant at 4, 24, and 48 hpi, respectively, relative to the wild-type infection. Gene ontology and KEGG enrichment analysis indicated that immune response-related proteins were enriched among the upregulated proteins. Compared to the wild-type M28 infection, the most upregulated proteins upon Tat-mutant infection included the cytosolic nucleic acid signaling pathway-related proteins IFIH1, DHX58, IFI202, IFI204, and ISG15 and the NF-κB signaling pathway-related proteins PTGS2, CD40, and TRAF1, suggesting that the host increases the production of these proteins in response to Tat mutant infection. Upregulation of some proteins was further verified by a parallel reaction monitoring (PRM) assay. ELISA and qRT-PCR assays indicated that Tat mutant infection significantly induced proinflammatory cytokine (TNF-α and IL-6) and nitric oxide (NO) production. Finally, we showed that the Tat mutant displays higher sensitivity to nitrosative stress than the wild type and that treatment with the NO synthase inhibitor L-NMMA significantly increases the intracellular survival of the Tat mutant, indicating that NO production contributes to restricting Tat mutant survival within macrophages. Collectively, this work improves our understanding of host immune responses to Tat mutants and provides insights into the mechanisms underlying the attenuated virulence of Tat mutants.
Collapse
Affiliation(s)
- Xin Yan
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Sen Hu
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yan Yang
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Da Xu
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Wenxing Liu
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ganwu Li
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.,Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Wentong Cai
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| | - Zhigao Bu
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, China
| |
Collapse
|
26
|
Zhao X, Shen H, Liang S, Zhu D, Wang M, Jia R, Chen S, Liu M, Yang Q, Wu Y, Zhang S, Huang J, Ou X, Mao S, Gao Q, Zhang L, Liu Y, Yu Y, Pan L, Cheng A. The lipopolysaccharide outer core transferase genes pcgD and hptE contribute differently to the virulence of Pasteurella multocida in ducks. Vet Res 2021; 52:37. [PMID: 33663572 PMCID: PMC7931556 DOI: 10.1186/s13567-021-00910-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
Fowl cholera caused by Pasteurella multocida exerts a massive economic burden on the poultry industry. Lipopolysaccharide (LPS) is essential for the growth of P. multocida genotype L1 strains in chickens and specific truncations to the full length LPS structure can attenuate bacterial virulence. Here we further dissected the roles of the outer core transferase genes pcgD and hptE in bacterial resistance to duck serum, outer membrane permeability and virulence in ducks. Two P. multocida mutants, ΔpcgD and ΔhptE, were constructed, and silver staining confirmed that they all produced truncated LPS profiles. Inactivation of pcgD or hptE did not affect bacterial susceptibility to duck serum and outer membrane permeability but resulted in attenuated virulence in ducks to some extent. After high-dose inoculation, ΔpcgD showed remarkably reduced colonization levels in the blood and spleen but not in the lung and liver and caused decreased injuries in the spleen and liver compared with the wild-type strain. In contrast, the ΔhptE loads declined only in the blood, and ΔhptE infection caused decreased splenic lesions but also induced severe hepatic lesions. Furthermore, compared with the wild-type strain, ΔpcgD was significantly attenuated upon oral or intramuscular challenge, whereas ΔhptE exhibited reduced virulence only upon oral infection. Therefore, the pcgD deletion caused greater virulence attenuation in ducks, indicating the critical role of pcgD in P. multocida infection establishment and survival.
Collapse
Affiliation(s)
- Xinxin Zhao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Hui Shen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Sheng Liang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Dekang Zhu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Mingshu Wang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Renyong Jia
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Shun Chen
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Mafeng Liu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Qiao Yang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Ying Wu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Shaqiu Zhang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Juan Huang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Xumin Ou
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Sai Mao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Qun Gao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Ling Zhang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yunya Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yanling Yu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Leichang Pan
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Anchun Cheng
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, Sichuan, China.
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
27
|
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
|
28
|
The Twin-Arginine Translocation System Is Important for Stress Resistance and Virulence of Brucella melitensis. Infect Immun 2020; 88:IAI.00389-20. [PMID: 32778612 DOI: 10.1128/iai.00389-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/03/2020] [Indexed: 12/25/2022] Open
Abstract
Brucella, the causative agent of brucellosis, is a stealthy intracellular pathogen that is highly pathogenic to a range of mammals, including humans. The twin-arginine translocation (Tat) pathway transports folded proteins across the cytoplasmic membrane and has been implicated in virulence in many bacterial pathogens. However, the roles of the Tat system and related substrates in Brucella remain unclear. We report here that disruption of Tat increases the sensitivity of Brucella melitensis M28 to the membrane stressor sodium dodecyl sulfate (SDS), indicating cell envelope defects, as well as to EDTA. In addition, mutating Tat renders M28 bacteria more sensitive to oxidative stress caused by H2O2 Further, loss of Tat significantly attenuates B. melitensis infection in murine macrophages ex vivo Using a mouse model for persistent infection, we demonstrate that Tat is required for full virulence of B. melitensis M28. Genome-wide in silico prediction combined with an in vivo amidase reporter assay indicates that at least 23 proteins are authentic Tat substrates, and they are functionally categorized into solute-binding proteins, oxidoreductases, cell envelope biosynthesis enzymes, and others. A comprehensive deletion study revealed that 6 substrates contribute significantly to Brucella virulence, including an l,d-transpeptidase, an ABC transporter solute-binding protein, and a methionine sulfoxide reductase. Collectively, our work establishes that the Tat pathway plays a critical role in Brucella virulence.
Collapse
|
29
|
Abstract
Brucellosis is a bacterial disease of domestic animals and humans. The pathogenic ability of Brucella organisms relies on their stealthy strategy and their capacity to replicate within host cells and to induce long-lasting infections. Brucella organisms barely induce neutrophil activation and survive within these leukocytes by resisting microbicidal mechanisms. Very few Brucella-infected neutrophils are found in the target organs, except for the bone marrow, early in infection. Still, Brucella induces a mild reactive oxygen species formation and, through its lipopolysaccharide, promotes the premature death of neutrophils, which release chemokines and express "eat me" signals. This effect drives the phagocytosis of infected neutrophils by mononuclear cells that become thoroughly susceptible to Brucella replication and vehicles for bacterial dispersion. The premature death of the infected neutrophils proceeds without NETosis, necrosis/oncosis, or classical apoptosis morphology. In the absence of neutrophils, the Th1 response exacerbates and promotes bacterial removal, indicating that Brucella-infected neutrophils dampen adaptive immunity. This modulatory effect opens a window for bacterial dispersion in host tissues before adaptive immunity becomes fully activated. However, the hyperactivation of immunity is not without a price, since neutropenic Brucella-infected animals develop cachexia in the early phases of the disease. The delay in the immunological response seems a sine qua non requirement for the development of long-lasting brucellosis. This property may be shared with other pathogenic alphaproteobacteria closely related to Brucella We propose a model in which Brucella-infected polymorphonuclear neutrophils (PMNs) function as "Trojan horse" vehicles for bacterial dispersal and as modulators of the Th1 adaptive immunity in infection.
Collapse
|
30
|
Trotta A, Milillo MA, Serafino A, Castillo LA, Birnberg Weiss F, Delpino MV, Giambartolomei GH, Fernández GC, Barrionuevo P. Brucella abortus-infected platelets modulate the activation of neutrophils. Immunol Cell Biol 2020; 98:743-756. [PMID: 32623755 DOI: 10.1111/imcb.12373] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 01/18/2023]
Abstract
Brucellosis is a contagious disease caused by bacteria of the genus Brucella. Platelets (PLTs) have been widely involved in the modulation of the immune response. We have previously reported the modulation of Brucella abortus-mediated infection of monocytes. As a result, PLTs cooperate with monocytes and increase their inflammatory capacity, promoting the resolution of the infection. Extending these results, in this study we demonstrate that patients with brucellosis present slightly elevated levels of complexes between PLTs and both monocytes and neutrophils. We then assessed whether PLTs were capable of modulating functional aspects of neutrophils. The presence of PLTs throughout neutrophil infection increased the production of interleukin-8, CD11b surface expression and reactive oxygen species formation, whereas it decreased the expression of CD62L, indicating an activated status of these cells. We next analyzed whether this modulation was mediated by released factors. To discriminate between these options, neutrophils were treated with supernatants collected from B. abortus-infected PLTs. Our results show that CD11b expression was induced by soluble factors of PLTs but direct contact between cell populations was needed to enhance the respiratory burst. Additionally, B. abortus-infected PLTs recruit polymorphonuclear (PMN) cells to the site of infection. Finally, the presence of PLTs did not modify the initial invasion of PMN cells by B. abortus but improved the control of the infection at extended times. Altogether, our results demonstrate that PLTs interact with neutrophils and promote a proinflammatory phenotype which could also contribute to the resolution of the infection.
Collapse
Affiliation(s)
- Aldana Trotta
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - M Ayelén Milillo
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Agustina Serafino
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Luis A Castillo
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Federico Birnberg Weiss
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - M Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Buenos Aires, Argentina
| | | | - Gabriela C Fernández
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Paula Barrionuevo
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| |
Collapse
|
31
|
Ouahrani-Bettache S, Jiménez De Bagüés MP, De La Garza J, Freddi L, Bueso JP, Lyonnais S, Al Dahouk S, De Biase D, Köhler S, Occhialini A. Lethality of Brucella microti in a murine model of infection depends on the wbkE gene involved in O-polysaccharide synthesis. Virulence 2020; 10:868-878. [PMID: 31635539 PMCID: PMC6844557 DOI: 10.1080/21505594.2019.1682762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Brucella microti was isolated a decade ago from wildlife and soil in Europe. Compared to the classical Brucella species, it exhibits atypical virulence properties such as increased growth in human and murine macrophages and lethality in experimentally infected mice. A spontaneous rough (R) mutant strain, derived from the smooth reference strain CCM4915T, showed increased macrophage colonization and was non-lethal in murine infections. Whole-genome sequencing and construction of an isogenic mutant of B. microti and Brucella suis 1330 revealed that the R-phenotype was due to a deletion in a single gene, namely wbkE (BMI_I539), encoding a putative glycosyltransferase involved in lipopolysaccharide (LPS) O-polysaccharide biosynthesis. Complementation of the R-strains with the wbkE gene restored the smooth phenotype and the ability of B. microti to kill infected mice. LPS with an intact O-polysaccharide is therefore essential for lethal B. microti infections in the murine model, demonstrating its importance in pathogenesis.
Collapse
Affiliation(s)
| | - María P Jiménez De Bagüés
- Unidad de Tecnología en Producción y Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria, Instituto Agroalimentario de Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | | | - Luca Freddi
- IRIM, CNRS, University Montpellier, INSERM, Montpellier, France
| | - Juan P Bueso
- Laboratorio Agroalimentario, Gobierno de Aragón, Zaragoza, Spain
| | | | - Sascha Al Dahouk
- Department of Biological Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Daniela De Biase
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Laboratory affiliated to the Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Latina, Italy
| | - Stephan Köhler
- IRIM, CNRS, University Montpellier, INSERM, Montpellier, France
| | | |
Collapse
|
32
|
Horne JE, Brockwell DJ, Radford SE. Role of the lipid bilayer in outer membrane protein folding in Gram-negative bacteria. J Biol Chem 2020; 295:10340-10367. [PMID: 32499369 PMCID: PMC7383365 DOI: 10.1074/jbc.rev120.011473] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/03/2020] [Indexed: 01/09/2023] Open
Abstract
β-Barrel outer membrane proteins (OMPs) represent the major proteinaceous component of the outer membrane (OM) of Gram-negative bacteria. These proteins perform key roles in cell structure and morphology, nutrient acquisition, colonization and invasion, and protection against external toxic threats such as antibiotics. To become functional, OMPs must fold and insert into a crowded and asymmetric OM that lacks much freely accessible lipid. This feat is accomplished in the absence of an external energy source and is thought to be driven by the high thermodynamic stability of folded OMPs in the OM. With such a stable fold, the challenge that bacteria face in assembling OMPs into the OM is how to overcome the initial energy barrier of membrane insertion. In this review, we highlight the roles of the lipid environment and the OM in modulating the OMP-folding landscape and discuss the factors that guide folding in vitro and in vivo We particularly focus on the composition, architecture, and physical properties of the OM and how an understanding of the folding properties of OMPs in vitro can help explain the challenges they encounter during folding in vivo Current models of OMP biogenesis in the cellular environment are still in flux, but the stakes for improving the accuracy of these models are high. OMP folding is an essential process in all Gram-negative bacteria, and considering the looming crisis of widespread microbial drug resistance it is an attractive target. To bring down this vital OMP-supported barrier to antibiotics, we must first understand how bacterial cells build it.
Collapse
Affiliation(s)
- Jim E Horne
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - David J Brockwell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
33
|
Aragón-Aranda B, de Miguel MJ, Lázaro-Antón L, Salvador-Bescós M, Zúñiga-Ripa A, Moriyón I, Iriarte M, Muñoz PM, Conde-Álvarez R. Development of attenuated live vaccine candidates against swine brucellosis in a non-zoonotic B. suis biovar 2 background. Vet Res 2020; 51:92. [PMID: 32703299 PMCID: PMC7376850 DOI: 10.1186/s13567-020-00815-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/05/2020] [Indexed: 02/07/2023] Open
Abstract
Brucella is a genus of gram-negative bacteria that cause brucellosis. B. abortus and B. melitensis infect domestic ruminants while B. suis (biovars 1-3) infect swine, and all these bacteria but B. suis biovar 2 are zoonotic. Live attenuated B. abortus S19 and B. melitensis Rev1 are effective vaccines in domestic ruminants, though both can infect humans. However, there is no swine brucellosis vaccine. Here, we investigated the potential use as vaccines of B. suis biovar 2 rough (R) lipopolysaccharide (LPS) mutants totally lacking O-chain (Bs2ΔwbkF) or only producing internal O-chain precursors (Bs2Δwzm) and mutants with a smooth (S) LPS defective in the core lateral branch (Bs2ΔwadB and Bs2ΔwadD). We also investigated mutants in the pyruvate phosphate dikinase (Bs2ΔppdK) and phosphoenolpyruvate carboxykinase (Bs2ΔpckA) genes encoding enzymes bridging phosphoenolpyruvate and the tricarboxylic acid cycle. When tested in the OIE mouse model at the recommended R or S vaccine doses (108 and 105 CFU, respectively), CFU/spleen of all LPS mutants were reduced with respect to the wild type and decreased faster for the R than for the S mutants. At those doses, protection against B. suis was similar for Bs2ΔwbkF, Bs2Δwzm, Bs2ΔwadB and the Rev1 control (105 CFU). As described before for B. abortus, B. suis biovar 2 carried a disabled pckA so that a double mutant Bs2ΔppdKΔpckA had the same metabolic phenotype as Bs2ΔppdK and ppdK mutation was enough to generate attenuation. At 105 CFU, Bs2ΔppdK also conferred the same protection as Rev1. As compared to other B. suis vaccine candidates described before, the mutants described here simultaneously carry irreversible deletions easy to identify as vaccine markers, lack antibiotic-resistance markers and were obtained in a non-zoonotic background. Since R vaccines should not elicit antibodies to the S-LPS and wzm mutants carry immunogenic O-chain precursors and did not improve Bs2ΔwbkF, the latter seems a better R vaccine candidate than Bs2Δwzm. However, taking into account that all R vaccines interfere in ELISA and other widely used assays, whether Bs2ΔwbkF is advantageous over Bs2ΔwadB or Bs2ΔppdK requires experiments in the natural host.
Collapse
Affiliation(s)
- Beatriz Aragón-Aranda
- Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA) and Dpto. de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008, Pamplona, Spain
| | - María Jesús de Miguel
- Unidad de Producción y Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Avda. Montañana 930, 50059, Zaragoza, Spain.,Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, Spain
| | - Leticia Lázaro-Antón
- Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA) and Dpto. de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008, Pamplona, Spain
| | - Miriam Salvador-Bescós
- Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA) and Dpto. de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008, Pamplona, Spain
| | - Amaia Zúñiga-Ripa
- Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA) and Dpto. de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008, Pamplona, Spain
| | - Ignacio Moriyón
- Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA) and Dpto. de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008, Pamplona, Spain
| | - Maite Iriarte
- Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA) and Dpto. de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008, Pamplona, Spain
| | - Pilar M Muñoz
- Unidad de Producción y Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Avda. Montañana 930, 50059, Zaragoza, Spain. .,Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, Spain.
| | - Raquel Conde-Álvarez
- Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA) and Dpto. de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008, Pamplona, Spain.
| |
Collapse
|
34
|
Degos C, Hysenaj L, Gonzalez‐Espinoza G, Arce‐Gorvel V, Gagnaire A, Papadopoulos A, Pasquevich KA, Méresse S, Cassataro J, Mémet S, Gorvel J. Omp25‐dependent engagement of SLAMF1 byBrucella abortusin dendritic cells limits acute inflammation and favours bacterial persistence in vivo. Cell Microbiol 2020; 22:e13164. [DOI: 10.1111/cmi.13164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/10/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Clara Degos
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Lisiena Hysenaj
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | | | - Vilma Arce‐Gorvel
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Aurélie Gagnaire
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Alexia Papadopoulos
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Karina Alejandra Pasquevich
- Instituto de Investigaciones BiotecnológicasUniversidad Nacional de San Martín (UNSAM)‐CONICET Buenos Aires Argentina
| | - Stéphane Méresse
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Juliana Cassataro
- Instituto de Investigaciones BiotecnológicasUniversidad Nacional de San Martín (UNSAM)‐CONICET Buenos Aires Argentina
| | - Sylvie Mémet
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Jean‐Pierre Gorvel
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| |
Collapse
|
35
|
Avila-Calderón ED, Flores-Romo L, Sharon W, Donis-Maturano L, Becerril-García MA, Arreola MGA, Reynoso BA, Güemes FS, Contreras-Rodríguez A. Dendritic cells and Brucella spp. interaction: the sentinel host and the stealthy pathogen. Folia Microbiol (Praha) 2020; 65:1-16. [PMID: 30783994 PMCID: PMC7224029 DOI: 10.1007/s12223-019-00691-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/06/2019] [Indexed: 01/18/2023]
Abstract
As dendritic cells (DCs) are among the first cells to encounter antigens, these cells trigger both innate and T cell responses, and are the most potent antigen-presenting cells. Brucella spp., which is an intracellular facultative and stealthy pathogen, is able to evade the bactericidal activities of professional phagocytes. Several studies have demonstrated that Brucella can survive and replicate intracellularly, thereby provoking impaired maturation of DCs. Therefore, the interaction between DCs and Brucella becomes an interesting model to study the immune response. In this review, we first will describe the most common techniques for DCs differentiation in vitro as well as general features of brucellosis. Then, the interaction of DCs and Brucella, including pathogen recognition, molecular mechanisms of bacterial pathogenesis, and intracellular trafficking of Brucella to subvert innate response, will be reviewed. Finally, we will debate diversity in immunological DC response and the controversial role of DC activation against Brucella infection.
Collapse
Affiliation(s)
- Eric Daniel Avila-Calderón
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, CINVESTAV-IPN, Av. IPN No 2508, Zacatenco, C.P 07330, Mexico city, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico
| | - Leopoldo Flores-Romo
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, CINVESTAV-IPN, Av. IPN No 2508, Zacatenco, C.P 07330, Mexico city, Mexico
| | - Witonsky Sharon
- Center for Molecular Medicine and Infectious Diseases/Center for One Health, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061-0442, USA
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061-0442, USA
| | - Luis Donis-Maturano
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana 3918, Zona Playitas, 22860, Ensenada, Baja California, Mexico
| | - Miguel Angel Becerril-García
- Departamento de Microbiología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Avenida Francisco I Madero y Dr. Aguirre Pequeño S/N Mitras Centro, 64460, Monterrey, Nuevo León, Mexico
| | - Ma Guadalupe Aguilera Arreola
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico
| | - Beatriz Arellano Reynoso
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico city, Mexico
| | - Francisco Suarez Güemes
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico city, Mexico
| | - Araceli Contreras-Rodríguez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico.
| |
Collapse
|
36
|
Tang T, Chen G, Guo A, Xu Y, Zhao L, Wang M, Lu C, Jiang Y, Zhang C. Comparative proteomic and genomic analyses of Brucella abortus biofilm and planktonic cells. Mol Med Rep 2019; 21:731-743. [PMID: 31974592 PMCID: PMC6947884 DOI: 10.3892/mmr.2019.10888] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 07/26/2019] [Indexed: 12/24/2022] Open
Abstract
The present study aimed to explore the differences in protein and gene expression of Brucella abortus cultured under biofilm and planktonic conditions. The proteins unique to biofilms and planktonic B. abortus were separated by two-dimensional (2-D) electrophoresis and then identified by matrix-assisted laser desorption/ionization-tandem time of flight-mass spectrometry (MALDI-TOF/TOF-MS). High-throughput sequencing and bioinformatic analyses were performed to identify differentially expressed genes between B. abortus cultured under biofilm and planktonic conditions. The proteins and genes identified by proteomic and genomic analyses were further evaluated via western blot and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analyses. 2-D electrophoresis identified 20 differentially expressed protein spots between biofilms and planktonic cells, which corresponded to 18 individual proteins (12 downregulated and 6 upregulated) after MALDI-TOF/TOF-MS analysis, including elongation factor Tu and enolase. RT-qPCR analysis revealed that all of the 18 genes were downregulated in biofilms compared with planktonic cells. Western blot analysis identified 9 downregulated and 3 upregulated proteins. High-throughput sequencing and bioinformatic analyses identified 14 function and pathway-associated genes (e.g., BAbS19_I14970). RT-qPCR analysis of the 14 genes showed that they were upregulated in biofilm compared with in planktonic state. In conclusion, these differentially expressed genes may play important roles in bacterial defense, colonization, invasion, and virulence.
Collapse
Affiliation(s)
- Taishan Tang
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Guoqiang Chen
- Division of Animal and Plant Quarantine Supervision, Suzhou Entry Exit Inspection and Quarantine Bureau, Suzhou, Jiangsu 215021, P.R. China
| | - Aizhen Guo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Ye Xu
- Animal, Plant and Food Inspection Center, Jiangsu Entry Exit Inspection and Quarantine Bureau, Nanjing, Jiangsu 210001, P.R. China
| | - Linli Zhao
- The Inspection and Quarantine Technology Center, Inner Mongolia Entry Exit Inspection and Quarantine Bureau, Hohhot, Inner Mongolia 010020, P.R. China
| | - Mengrui Wang
- Animal, Plant and Food Inspection Center, Jiangsu Entry Exit Inspection and Quarantine Bureau, Nanjing, Jiangsu 210001, P.R. China
| | - Chengping Lu
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Yuan Jiang
- Animal, Plant and Food Inspection Center, Jiangsu Entry Exit Inspection and Quarantine Bureau, Nanjing, Jiangsu 210001, P.R. China
| | - Changyin Zhang
- Animal, Plant and Food Inspection Center, Jiangsu Entry Exit Inspection and Quarantine Bureau, Nanjing, Jiangsu 210001, P.R. China
| |
Collapse
|
37
|
Nayeri Fasaei B, Naserli S, Zahraei Salehi T, Saeedinia A, Behroozikhah A, Ashrafi Tamai I. New Brucella abortus S19 Mutant to Improve Distinction Between Infected and Vaccinated Animals. IRANIAN JOURNAL OF BIOTECHNOLOGY 2019; 17:e2159. [PMID: 32195284 PMCID: PMC7080968 DOI: 10.29252/ijb.2159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Background Using Brucella abortus Strain 19 (S19) to control bovine brucellosis is restricted due to induce antibodies to the O-side chain of the smooth lipopolysaccharide (LPS) which may be difficult to differentiate vaccinated and infected animals. Furthermore, it is virulent for humans and can induce abortion to cattle. Objectives The aim of this study was to employ gene knockout B. abortus S19 for the first time to eliminate diagnostic defects and obtain the attenuated mutant strain. Material and Methods The wbkA gene, which is one of the LPS O-chain coding genes, was knocked out in vaccinal Brucella abortus S19. The proliferative response and immunoglobulin M production were analyzed in wbkA deletion strain-infected BALB/c mice. Results The loss of wbkA gene function resulted in induction of the splenocyte proliferative response in mice infected by the mutant S19 strain compare to those induced by parental S19 and RB51 strains. Moreover, wbkA mutant did not induce any IgM antibody response using the enzyme-linked immunosorbent assay. Conclusions As a result, the new mutant S19 strain had deficiency in its LPS O-chain structure, besides cannot induce IgM response then, reduce mistakes to discriminate between vaccinated and infected animal, and also can be considered as a new vaccine candidate.
Collapse
Affiliation(s)
- Bahar Nayeri Fasaei
- Department of Microbiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Soulmaz Naserli
- Department of Microbiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Taghi Zahraei Salehi
- Department of Microbiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Alireza Saeedinia
- Department of Bioscience and Biotechnology, Malek-Ashtar University of Technology, Tehran, Iran
| | - Alimohammad Behroozikhah
- Department of Brucellosis, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Iradj Ashrafi Tamai
- Department of Microbiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
38
|
López-Santiago R, Sánchez-Argáez AB, De Alba-Núñez LG, Baltierra-Uribe SL, Moreno-Lafont MC. Immune Response to Mucosal Brucella Infection. Front Immunol 2019; 10:1759. [PMID: 31481953 PMCID: PMC6710357 DOI: 10.3389/fimmu.2019.01759] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/11/2019] [Indexed: 01/18/2023] Open
Abstract
Brucellosis is one of the most prevalent bacterial zoonosis of worldwide distribution. The disease is caused by Brucella spp., facultative intracellular pathogens. Brucellosis in animals results in abortion of fetuses, while in humans, it frequently manifests flu-like symptoms and a typical undulant fever, being osteoarthritis a common complication of the chronic infection. The two most common ways to acquire the infection in humans are through the ingestion of contaminated dairy products or by inhalation of contaminated aerosols. Brucella spp. enter the body mainly through the gastrointestinal and respiratory mucosa; however, most studies of immune response to Brucella spp. are performed analyzing models of systemic immunity. It is necessary to better understand the mucosal immune response induced by Brucella infection since this is the main entry site for the bacterium. In this review, some virulence factors and the mechanisms needed for pathogen invasion and persistence are discussed. Furthermore, some aspects of local immune responses induced during Brucella infection will be reviewed. With this knowledge, better vaccines can be designed focused on inducing protective mucosal immune response.
Collapse
Affiliation(s)
- Rubén López-Santiago
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Ana Beatriz Sánchez-Argáez
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Liliana Gabriela De Alba-Núñez
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Martha Cecilia Moreno-Lafont
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
39
|
Demars A, Lison A, Machelart A, Van Vyve M, Potemberg G, Vanderwinden JM, De Bolle X, Letesson JJ, Muraille E. Route of Infection Strongly Impacts the Host-Pathogen Relationship. Front Immunol 2019; 10:1589. [PMID: 31354728 PMCID: PMC6637429 DOI: 10.3389/fimmu.2019.01589] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/25/2019] [Indexed: 12/24/2022] Open
Abstract
Live attenuated vaccines play a key role in the control of many human and animal pathogens. Their rational development is usually helped by identification of the reservoir of infection, the lymphoid subpopulations associated with protective immunity as well as the virulence genes involved in pathogen persistence. Here, we compared the course of Brucella melitensis infection in C57BL/6 mice infected via intraperitoneal (i.p.), intranasal (i.n.) and intradermal (i.d.) route and demonstrated that the route of infection strongly impacts all of these parameters. Following i.p. and i.n. infection, most infected cells observed in the spleen or lung were F4/80+ myeloid cells. In striking contrast, infected Ly6G+ neutrophils and CD140a+ fibroblasts were also observed in the skin after i.d. infection. The virB operon encoding for the type IV secretion system is considered essential to deflecting vacuolar trafficking in phagocytic cells and allows Brucella to multiply and persist. Unexpectedly, the ΔvirB Brucella strain, which does not persist in the lung after i.n. infection, persists longer in skin tissues than the wild strain after i.d. infection. While the CD4+ T cell-mediated Th1 response is indispensable to controlling the Brucella challenge in the i.p. model, it is dispensable for the control of Brucella in the i.d. and i.n. models. Similarly, B cells are indispensable in the i.p. and i.d. models but dispensable in the i.n. model. γδ+ T cells appear able to compensate for the absence of αβ+ T cells in the i.d. model but not in the other models. Taken together, our results demonstrate the crucial importance of the route of infection for the host pathogen relationship.
Collapse
Affiliation(s)
- Aurore Demars
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Aurore Lison
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Arnaud Machelart
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Margaux Van Vyve
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Georges Potemberg
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | | | - Xavier De Bolle
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Jean-Jacques Letesson
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
| | - Eric Muraille
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, NARILIS, Université de Namur, Namur, Belgium
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Bruxelles, Belgium
| |
Collapse
|
40
|
Vassen V, Valotteau C, Feuillie C, Formosa-Dague C, Dufrêne YF, De Bolle X. Localized incorporation of outer membrane components in the pathogen Brucella abortus. EMBO J 2019; 38:e100323. [PMID: 30635335 PMCID: PMC6396147 DOI: 10.15252/embj.2018100323] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 12/21/2022] Open
Abstract
The zoonotic pathogen Brucella abortus is part of the Rhizobiales, which are alpha-proteobacteria displaying unipolar growth. Here, we show that this bacterium exhibits heterogeneity in its outer membrane composition, with clusters of rough lipopolysaccharide co-localizing with the essential outer membrane porin Omp2b, which is proposed to allow facilitated diffusion of solutes through the porin. We also show that the major outer membrane protein Omp25 and peptidoglycan are incorporated at the new pole and the division site, the expected growth sites. Interestingly, lipopolysaccharide is also inserted at the same growth sites. The absence of long-range diffusion of main components of the outer membrane could explain the apparent immobility of the Omp2b clusters, as well as unipolar and mid-cell localizations of newly incorporated outer membrane proteins and lipopolysaccharide. Unipolar growth and limited mobility of surface structures also suggest that new surface variants could arise in a few generations without the need of diluting pre-existing surface antigens.
Collapse
Affiliation(s)
- Victoria Vassen
- Research Unit in Biology of Microorganisms (URBM), Narilis University of Namur (UNamur), Namur, Belgium
| | - Claire Valotteau
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain (UCL), Louvain-la-Neuve, Belgium
| | - Cécile Feuillie
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain (UCL), Louvain-la-Neuve, Belgium
| | - Cécile Formosa-Dague
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain (UCL), Louvain-la-Neuve, Belgium
| | - Yves F Dufrêne
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain (UCL), Louvain-la-Neuve, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium
| | - Xavier De Bolle
- Research Unit in Biology of Microorganisms (URBM), Narilis University of Namur (UNamur), Namur, Belgium
| |
Collapse
|
41
|
Bialer MG, Ruiz-Ranwez V, Sycz G, Estein SM, Russo DM, Altabe S, Sieira R, Zorreguieta A. MapB, the Brucella suis TamB homologue, is involved in cell envelope biogenesis, cell division and virulence. Sci Rep 2019; 9:2158. [PMID: 30770847 PMCID: PMC6377625 DOI: 10.1038/s41598-018-37668-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/05/2018] [Indexed: 01/18/2023] Open
Abstract
Brucella species are Gram-negative, facultative intracellular pathogens responsible for a worldwide zoonosis. The envelope of Brucella exhibits unique characteristics that make these bacteria furtive pathogens and resistant to several host defence compounds. We have identified a Brucella suis gene (mapB) that appeared to be crucial for cell envelope integrity. Indeed, the typical resistance of Brucella to both lysozyme and the cationic lipopeptide polymyxin B was markedly reduced in a ∆mapB mutant. MapB turned out to represent a TamB orthologue. This last protein, together with TamA, a protein belonging to the Omp85 family, form a complex that has been proposed to participate in the translocation of autotransporter proteins across the outer membrane (OM). Accordingly, we observed that MapB is required for proper assembly of an autotransporter adhesin in the OM, as most of the autotransporter accumulated in the mutant cell periplasm. Both assessment of the relative amounts of other specific outer membrane proteins (OMPs) and a proteome approach indicated that the absence of MapB did not lead to an extensive alteration in OMP abundance, but to a reduction in the relative amounts of a protein subset, including proteins from the Omp25/31 family. Electron microscopy revealed that ∆mapB cells exhibit multiple anomalies in cell morphology, indicating that the absence of the TamB homologue in B. suis severely affects cell division. Finally, ∆mapB cells were impaired in macrophage infection and showed an attenuated virulence phenotype in the mouse model. Collectively, our results indicate that the role of B. suis TamB homologue is not restricted to participating in the translocation of autotransporters across the OM but that it is essential for OM stability and protein composition and that it is involved in cell envelope biogenesis, a process that is inherently coordinated with cell division.
Collapse
Affiliation(s)
- Magalí Graciela Bialer
- Fundación Instituto Leloir, IIBBA-CONICET. Patricias Argentinas 435, (C1405BWE), Buenos Aires, Argentina
| | - Verónica Ruiz-Ranwez
- Fundación Instituto Leloir, IIBBA-CONICET. Patricias Argentinas 435, (C1405BWE), Buenos Aires, Argentina
| | - Gabriela Sycz
- Fundación Instituto Leloir, IIBBA-CONICET. Patricias Argentinas 435, (C1405BWE), Buenos Aires, Argentina
| | - Silvia Marcela Estein
- Laboratorio de Inmunología, Facultad de Ciencias Veterinarias, Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires (U.N.C.P.B.A), Tandil, Argentina
| | - Daniela Marta Russo
- Fundación Instituto Leloir, IIBBA-CONICET. Patricias Argentinas 435, (C1405BWE), Buenos Aires, Argentina
| | - Silvia Altabe
- Instituto de Biología Molecular y Celular de Rosario (IBR) and Departamento de Microbiología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Esmeralda y Ocampo, Rosario, Argentina
| | - Rodrigo Sieira
- Fundación Instituto Leloir, IIBBA-CONICET. Patricias Argentinas 435, (C1405BWE), Buenos Aires, Argentina
| | - Angeles Zorreguieta
- Fundación Instituto Leloir, IIBBA-CONICET. Patricias Argentinas 435, (C1405BWE), Buenos Aires, Argentina.
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
42
|
Herrou J, Willett JW, Fiebig A, Varesio LM, Czyż DM, Cheng JX, Ultee E, Briegel A, Bigelow L, Babnigg G, Kim Y, Crosson S. Periplasmic protein EipA determines envelope stress resistance and virulence in Brucella abortus. Mol Microbiol 2019; 111:637-661. [PMID: 30536925 DOI: 10.1111/mmi.14178] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2018] [Indexed: 12/17/2022]
Abstract
Molecular components of the Brucella abortus cell envelope play a major role in its ability to infect, colonize and survive inside mammalian host cells. In this study, we have defined a role for a conserved gene of unknown function in B. abortus envelope stress resistance and infection. Expression of this gene, which we name eipA, is directly activated by the essential cell cycle regulator, CtrA. eipA encodes a soluble periplasmic protein that adopts an unusual eight-stranded β-barrel fold. Deletion of eipA attenuates replication and survival in macrophage and mouse infection models, and results in sensitivity to treatments that compromise the cell envelope integrity. Transposon disruption of genes required for LPS O-polysaccharide biosynthesis is synthetically lethal with eipA deletion. This genetic connection between O-polysaccharide and eipA is corroborated by our discovery that eipA is essential in Brucella ovis, a naturally rough species that harbors mutations in several genes required for O-polysaccharide production. Conditional depletion of eipA expression in B. ovis results in a cell chaining phenotype, providing evidence that eipA directly or indirectly influences cell division in Brucella. We conclude that EipA is a molecular determinant of Brucella virulence that functions to maintain cell envelope integrity and influences cell division.
Collapse
Affiliation(s)
- Julien Herrou
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Jonathan W Willett
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Aretha Fiebig
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Lydia M Varesio
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Daniel M Czyż
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Jason X Cheng
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Eveline Ultee
- Department of Biology, Universiteit Leiden, Leiden, Netherlands
| | - Ariane Briegel
- Department of Biology, Universiteit Leiden, Leiden, Netherlands
| | - Lance Bigelow
- Biosciences Division, Argonne National Laboratory, Argonne, IL, USA
| | - Gyorgy Babnigg
- Biosciences Division, Argonne National Laboratory, Argonne, IL, USA
| | - Youngchang Kim
- Biosciences Division, Argonne National Laboratory, Argonne, IL, USA
| | - Sean Crosson
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
43
|
Lanne ABM, Goode A, Prattley C, Kumari D, Drasbek MR, Williams P, Conde-Álvarez R, Moriyón I, Bonev BB. Molecular recognition of lipopolysaccharide by the lantibiotic nisin. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1861:83-92. [PMID: 30296414 DOI: 10.1016/j.bbamem.2018.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 01/01/2023]
Abstract
Nisin is a lanthionine antimicrobial effective against diverse Gram-positive bacteria and is used as a food preservative worldwide. Its action is mediated by pyrophosphate recognition of the bacterial cell wall receptors lipid II and undecaprenyl pyrophosphate. Nisin/receptor complexes disrupt cytoplasmic membranes, inhibit cell wall synthesis and dysregulate bacterial cell division. Gram-negative bacteria are much more tolerant to antimicrobials including nisin. In contrast to Gram-positives, Gram-negative bacteria possess an outer membrane, the major constituent of which is lipopolysaccharide (LPS). This contains surface exposed phosphate and pyrophosphate groups and hence can be targeted by nisin. Here we describe the impact of LPS on membrane stability in response to nisin and the molecular interactions occurring between nisin and membrane-embedded LPS from different Gram-negative bacteria. Dye release from liposomes shows enhanced susceptibility to nisin in the presence of LPS, particularly rough LPS chemotypes that lack an O-antigen whereas LPS from microorganisms sharing similar ecological niches with antimicrobial producers provides only modest enhancement. Increased susceptibility was observed with LPS from pathogenic Klebsiella pneumoniae compared to LPS from enteropathogenic Salmonella enterica and gut commensal Escherichia coli. LPS from Brucella melitensis, an intra-cellular pathogen which is adapted to invade professional and non-professional phagocytes, appears to be refractory to nisin. Molecular complex formation between nisin and LPS was studied by solid state MAS NMR and revealed complex formation between nisin and LPS from most organisms investigated except B. melitensis. LPS/nisin complex formation was confirmed in outer membrane extracts from E. coli.
Collapse
Affiliation(s)
- Alice B M Lanne
- School of Life Sciences, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - Alice Goode
- School of Life Sciences, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - Charlotte Prattley
- School of Life Sciences, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - Divya Kumari
- School of Life Sciences, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - Mette Ryun Drasbek
- DuPont Nutrition Biosciences ApS, Edwin Rahrs Vej 38, DK-8220 Brabrand, Denmark
| | - Paul Williams
- School of Life Sciences, CBS, University of Nottingham, Nottingham NG7 2RD, UK
| | - Raquel Conde-Álvarez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008 Pamplona, Spain
| | - Ignacio Moriyón
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008 Pamplona, Spain
| | - Boyan B Bonev
- School of Life Sciences, QMC, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
44
|
Salvador-Bescós M, Gil-Ramírez Y, Zúñiga-Ripa A, Martínez-Gómez E, de Miguel MJ, Muñoz PM, Cloeckaert A, Zygmunt MS, Moriyón I, Iriarte M, Conde-Álvarez R. WadD, a New Brucella Lipopolysaccharide Core Glycosyltransferase Identified by Genomic Search and Phenotypic Characterization. Front Microbiol 2018; 9:2293. [PMID: 30319590 PMCID: PMC6171495 DOI: 10.3389/fmicb.2018.02293] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/07/2018] [Indexed: 01/20/2023] Open
Abstract
Brucellosis, an infectious disease caused by Brucella, is one of the most extended bacterial zoonosis in the world and an important cause of economic losses and human suffering. The lipopolysaccharide (LPS) of Brucella plays a major role in virulence as it impairs normal recognition by the innate immune system and delays the immune response. The LPS core is a branched structure involved in resistance to complement and polycationic peptides, and mutants in glycosyltransferases required for the synthesis of the lateral branch not linked to the O-polysaccharide (O-PS) are attenuated and have been proposed as vaccine candidates. For this reason, the complete understanding of the genes involved in the synthesis of this LPS section is of particular interest. The chemical structure of the Brucella LPS core suggests that, in addition to the already identified WadB and WadC glycosyltransferases, others could be implicated in the synthesis of this lateral branch. To clarify this point, we identified and constructed mutants in 11 ORFs encoding putative glycosyltransferases in B. abortus. Four of these ORFs, regulated by the virulence regulator MucR (involved in LPS synthesis) or the BvrR/BvrS system (implicated in the synthesis of surface components), were not required for the synthesis of a complete LPS neither for virulence or interaction with polycationic peptides and/or complement. Among the other seven ORFs, six seemed not to be required for the synthesis of the core LPS since the corresponding mutants kept the O-PS and reacted as the wild type with polyclonal sera. Interestingly, mutant in ORF BAB1_0953 (renamed wadD) lost reactivity against antibodies that recognize the core section while kept the O-PS. This suggests that WadD is a new glycosyltransferase adding one or more sugars to the core lateral branch. WadD mutants were more sensitive than the parental strain to components of the innate immune system and played a role in chronic stages of infection. These results corroborate and extend previous work indicating that the Brucella LPS core is a branched structure that constitutes a steric impairment preventing the elements of the innate immune system to fight against Brucella.
Collapse
Affiliation(s)
- Miriam Salvador-Bescós
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Yolanda Gil-Ramírez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Amaia Zúñiga-Ripa
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Estrella Martínez-Gómez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - María J de Miguel
- Unidad de Tecnología en Producción y Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria, Instituto Agroalimentario de Aragón - IA2 (CITA - Universidad de Zaragoza), Zaragoza, Spain
| | - Pilar M Muñoz
- Unidad de Tecnología en Producción y Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria, Instituto Agroalimentario de Aragón - IA2 (CITA - Universidad de Zaragoza), Zaragoza, Spain
| | - Axel Cloeckaert
- Institut National de la Recherche Agronomique, Université François Rabelais de Tours, UMR 1282, Nouzilly, France
| | - Michel S Zygmunt
- Institut National de la Recherche Agronomique, Université François Rabelais de Tours, UMR 1282, Nouzilly, France
| | - Ignacio Moriyón
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Maite Iriarte
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Raquel Conde-Álvarez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
45
|
Zhao Y, Hanniffy S, Arce-Gorvel V, Conde-Alvarez R, Oh S, Moriyón I, Mémet S, Gorvel JP. Immunomodulatory properties of Brucella melitensis lipopolysaccharide determinants on mouse dendritic cells in vitro and in vivo. Virulence 2018; 9:465-479. [PMID: 28968180 PMCID: PMC5955181 DOI: 10.1080/21505594.2017.1386831] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The lipopolysaccharide (LPS) is a major virulence factor of Brucella, a facultative intracellular pathogenic Gram-negative bacterium. Brucella LPS exhibits a low toxicity and its atypical structure was postulated to delay the host immune response, favouring the establishment of chronic disease. Here we carried out an in-depth in vitro and in vivo characterisation of the immunomodulatory effects of Brucella LPS on different dendritic cell (DC) subpopulations. By using LPSs from bacteria that share some of Brucella LPS structural features, we demonstrated that the core component of B. melitensis wild-type (Bm-wt) LPS accounts for the low activation potential of Brucella LPS in mouse GM-CSF-derived (GM-) DCs. Contrary to the accepted dogma considering Brucella LPS a poor TLR4 agonist and DC activator, Bm-wt LPS selectively induced expression of surface activation markers and cytokine secretion from Flt3-Ligand-derived (FL-) DCs in a TLR4-dependent manner. It also primed in vitro T cell proliferation by FL-DCs. In contrast, modified LPS with a defective core purified from Brucella carrying a mutated wadC gene (Bm-wadC), efficiently potentiated mouse and human DC activation and T cell proliferation in vitro. In vivo, Bm-wt LPS promoted scant activation of splenic DC subsets and limited recruitment of monocyte- DC like cells in the spleen, conversely to Bm-wadC LPS. Bm-wadC live bacteria drove high cytokine secretion levels in sera of infected mice. Altogether, these results illustrate the immunomodulatory properties of Brucella LPS and the enhanced DC activation ability of the wadC mutation with potential for vaccine development targeting Brucella core LPS structure.
Collapse
Affiliation(s)
- Yun Zhao
- a Centre d'Immunologie de Marseille-Luminy, CIML, Aix Marseille Univ, CNRS, INSERM , Marseille , France
| | - Sean Hanniffy
- a Centre d'Immunologie de Marseille-Luminy, CIML, Aix Marseille Univ, CNRS, INSERM , Marseille , France
| | - Vilma Arce-Gorvel
- a Centre d'Immunologie de Marseille-Luminy, CIML, Aix Marseille Univ, CNRS, INSERM , Marseille , France
| | - Raquel Conde-Alvarez
- b Departamento de Microbiología y Parasitología , Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra , c/Irunlarrea 1, Pamplona , Spain
| | - SangKon Oh
- c Baylor Institute for Immunology Research , 3434 Live Oak St., Dallas , TX , U.S.A
| | - Ignacio Moriyón
- b Departamento de Microbiología y Parasitología , Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra , c/Irunlarrea 1, Pamplona , Spain
| | - Sylvie Mémet
- a Centre d'Immunologie de Marseille-Luminy, CIML, Aix Marseille Univ, CNRS, INSERM , Marseille , France
| | - Jean-Pierre Gorvel
- a Centre d'Immunologie de Marseille-Luminy, CIML, Aix Marseille Univ, CNRS, INSERM , Marseille , France
| |
Collapse
|
46
|
Pérez-Etayo L, de Miguel MJ, Conde-Álvarez R, Muñoz PM, Khames M, Iriarte M, Moriyón I, Zúñiga-Ripa A. The CO 2-dependence of Brucella ovis and Brucella abortus biovars is caused by defective carbonic anhydrases. Vet Res 2018; 49:85. [PMID: 30185220 PMCID: PMC6126018 DOI: 10.1186/s13567-018-0583-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/03/2018] [Indexed: 12/14/2022] Open
Abstract
Brucella bacteria cause brucellosis, a major zoonosis whose control requires efficient diagnosis and vaccines. Identification of classical Brucella spp. has traditionally relied on phenotypic characterization, including surface antigens and 5–10% CO2 necessity for growth (CO2-dependence), a trait of Brucella ovis and most Brucella abortus biovars 1–4 strains. Although molecular tests are replacing phenotypic methods, CO2-dependence remains of interest as it conditions isolation and propagation and reflects Brucella metabolism, an area of active research. Here, we investigated the connection of CO2-dependence and carbonic anhydrases (CA), the enzymes catalyzing the hydration of CO2 to the bicarbonate used by anaplerotic and biosynthetic carboxylases. Based on the previous demonstration that B. suis carries two functional CAs (CAI and CAII), we analyzed the CA sequences of CO2-dependent and -independent brucellae and spontaneous mutants. The comparisons strongly suggested that CAII is not functional in CO2-dependent B. abortus and B. ovis, and that a modified CAII sequence explains the CO2-independent phenotype of spontaneous mutants. Then, by mutagenesis and heterologous plasmid complementation and chromosomal insertion we proved that CAI alone is enough to support CO2-independent growth of B. suis in rich media but not of B. abortus in rich media or B. suis in minimal media. Finally, we also found that insertion of a heterologous active CAII into B. ovis reverted the CO2-dependence but did not alter its virulence in the mouse model. These results allow a better understanding of central aspects of Brucella metabolism and, in the case of B. ovis, provide tools for large-scale production of diagnostic antigens and vaccines.
Collapse
Affiliation(s)
- Lara Pérez-Etayo
- Instituto de Salud Tropical y Departamento de Microbiología y Parasitología-IDISNA, Universidad de Navarra, 31008, Pamplona, Spain
| | - María Jesús de Miguel
- Unidad de Producción y Sanidad Animal del Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Instituto Agroalimentario de Aragón (IA2), Zaragoza, Spain
| | - Raquel Conde-Álvarez
- Instituto de Salud Tropical y Departamento de Microbiología y Parasitología-IDISNA, Universidad de Navarra, 31008, Pamplona, Spain
| | - Pilar M Muñoz
- Unidad de Producción y Sanidad Animal del Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Instituto Agroalimentario de Aragón (IA2), Zaragoza, Spain
| | - Mammar Khames
- Department of Biology, University of Medea, 26000, Medea, Algeria.,National Veterinary High School, Algiers, Algeria
| | - Maite Iriarte
- Instituto de Salud Tropical y Departamento de Microbiología y Parasitología-IDISNA, Universidad de Navarra, 31008, Pamplona, Spain
| | - Ignacio Moriyón
- Instituto de Salud Tropical y Departamento de Microbiología y Parasitología-IDISNA, Universidad de Navarra, 31008, Pamplona, Spain
| | - Amaia Zúñiga-Ripa
- Instituto de Salud Tropical y Departamento de Microbiología y Parasitología-IDISNA, Universidad de Navarra, 31008, Pamplona, Spain.
| |
Collapse
|
47
|
Depletion of Complement Enhances the Clearance of Brucella abortus in Mice. Infect Immun 2018; 86:IAI.00567-18. [PMID: 30082480 DOI: 10.1128/iai.00567-18] [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] [Received: 07/20/2018] [Accepted: 07/30/2018] [Indexed: 11/20/2022] Open
Abstract
Brucellosis is a bacterial disease of animals and humans. Brucella abortus barely activates the innate immune system at the onset of infection, and this bacterium is resistant to the microbicidal action of complement. Since complement stands as the first line of defense during bacterial invasions, we explored the role of complement in B. abortus infections. Brucella abortus-infected mice depleted of complement with cobra venom factor (CVF) showed the same survival rate as mice in the control group. The complement-depleted mice readily eliminated B. abortus from the spleen and did so more efficiently than the infected controls after 7 days of infection. The levels of the proinflammatory cytokines tumor necrosis factor alpha and interleukin-6 (IL-6) remained within background levels in complement-depleted B. abortus-infected mice. In contrast, the levels of the immune activator cytokine gamma interferon and the regulatory cytokine IL-10 were significantly increased. No significant histopathological changes in the liver and spleen were observed between the complement-depleted B. abortus-infected mice and the corresponding controls. The action exerted by Brucella on the immune system in the absence of complement may correspond to a broader phenomenon that involves several components of innate immunity.
Collapse
|
48
|
Alterations of Metabolic and Lipid Profiles in Polymyxin-Resistant Pseudomonas aeruginosa. Antimicrob Agents Chemother 2018; 62:AAC.02656-17. [PMID: 29632014 DOI: 10.1128/aac.02656-17] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 04/05/2018] [Indexed: 12/27/2022] Open
Abstract
Multidrug-resistant Pseudomonas aeruginosa presents a global medical challenge, and polymyxins are a key last-resort therapeutic option. Unfortunately, polymyxin resistance in P. aeruginosa has been increasingly reported. The present study was designed to define metabolic differences between paired polymyxin-susceptible and -resistant P. aeruginosa strains using untargeted metabolomics and lipidomics analyses. The metabolomes of wild-type P. aeruginosa strain K ([PAK] polymyxin B MIC, 1 mg/liter) and its paired pmrB mutant strains, PAKpmrB6 and PAKpmrB12 (polymyxin B MICs of 16 mg/liter and 64 mg/liter, respectively) were characterized using liquid chromatography-mass spectrometry, and metabolic differences were identified through multivariate and univariate statistics. PAKpmrB6 and PAKpmrB12, which displayed lipid A modifications with 4-amino-4-deoxy-l-arabinose, showed significant perturbations in amino acid and carbohydrate metabolism, particularly the intermediate metabolites from 4-amino-4-deoxy-l-arabinose synthesis and the methionine salvage cycle pathways. The genomics result showed a premature termination (Y275stop) in speE (encoding spermidine synthase) in PAKpmrB6, and metabolomics data revealed a decreased intracellular level of spermidine in PAKpmrB6 compared to that in PAKpmrB12 Our results indicate that spermidine may play an important role in high-level polymyxin resistance in P. aeruginosa Interestingly, both pmrB mutants had decreased levels of phospholipids, fatty acids, and acyl-coenzyme A compared to those in the wild-type PAK. Moreover, the more resistant PAKpmrB12 mutant exhibited much lower levels of phospholipids than the PAKpmrB6 mutant, suggesting that the decreased phospholipid level was associated with polymyxin resistance. In summary, this study provides novel mechanistic information on polymyxin resistance in P. aeruginosa and highlights its impacts on bacterial metabolism.
Collapse
|
49
|
Martínez-Gómez E, Ståhle J, Gil-Ramírez Y, Zúñiga-Ripa A, Zaccheus M, Moriyón I, Iriarte M, Widmalm G, Conde-Álvarez R. Genomic Insertion of a Heterologous Acetyltransferase Generates a New Lipopolysaccharide Antigenic Structure in Brucella abortus and Brucella melitensis. Front Microbiol 2018; 9:1092. [PMID: 29887851 PMCID: PMC5981137 DOI: 10.3389/fmicb.2018.01092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/07/2018] [Indexed: 12/31/2022] Open
Abstract
Brucellosis is a bacterial zoonosis of worldwide distribution caused by bacteria of the genus Brucella. In Brucella abortus and Brucella melitensis, the major species infecting domestic ruminants, the smooth lipopolysaccharide (S-LPS) is a virulence factor. This S-LPS carries a N-formyl-perosamine homopolymer O-polysaccharide that is the major antigen in serodiagnostic tests and is required for virulence. We report that the Brucella O-PS can be structurally and antigenically modified using wbdR, the acetyl-transferase gene involved in N-acetyl-perosamine synthesis in Escherichia coli O157:H7. Brucella constructs carrying plasmidic wbdR expressed a modified O-polysaccharide but were unstable, a problem circumvented by inserting wbdR into a neutral site of chromosome II. As compared to wild-type bacteria, both kinds of wbdR constructs expressed shorter O-polysaccharides and NMR analyses showed that they contained both N-formyl and N-acetyl-perosamine. Moreover, deletion of the Brucella formyltransferase gene wbkC in wbdR constructs generated bacteria producing only N-acetyl-perosamine homopolymers, proving that wbdR can replace for wbkC. Absorption experiments with immune sera revealed that the wbdR constructs triggered antibodies to new immunogenic epitope(s) and the use of monoclonal antibodies proved that B. abortus and B. melitensis wbdR constructs respectively lacked the A or M epitopes, and the absence of the C epitope in both backgrounds. The wbdR constructs showed resistance to polycations similar to that of the wild-type strains but displayed increased sensitivity to normal serum similar to that of a per R mutant. In mice, the wbdR constructs produced chronic infections and triggered antibody responses that can be differentiated from those evoked by the wild-type strain in S-LPS ELISAs. These results open the possibilities of developing brucellosis vaccines that are both antigenically tagged and lack the diagnostic epitopes of virulent field strains, thereby solving the diagnostic interference created by current vaccines against Brucella.
Collapse
Affiliation(s)
- Estrella Martínez-Gómez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Jonas Ståhle
- Department of Organic Chemistry, Arrhenius Laboratory, Stockholm University, Stockholm, Sweden
| | - Yolanda Gil-Ramírez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Amaia Zúñiga-Ripa
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Mona Zaccheus
- Department of Organic Chemistry, Arrhenius Laboratory, Stockholm University, Stockholm, Sweden
| | - Ignacio Moriyón
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Maite Iriarte
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Göran Widmalm
- Department of Organic Chemistry, Arrhenius Laboratory, Stockholm University, Stockholm, Sweden
| | - Raquel Conde-Álvarez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
50
|
Guidolin LS, Arce-Gorvel V, Ciocchini AE, Comerci DJ, Gorvel JP. Cyclic β-glucans at the bacteria-host cells interphase: One sugar ring to rule them all. Cell Microbiol 2018; 20:e12850. [PMID: 29624823 DOI: 10.1111/cmi.12850] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/20/2018] [Accepted: 03/23/2018] [Indexed: 01/08/2023]
Abstract
Cyclic β-1,2-D-glucans (CβG) are natural bionanopolymers present in the periplasmic space of many Proteobacteria. These molecules are sugar rings made of 17 to 25 D-glucose units linked exclusively by β-1,2-glycosidic bonds. CβG are important for environmental sensing and osmoadaptation in bacteria, but most importantly, they play key roles in complex host-cell interactions such as symbiosis, pathogenesis, and immunomodulation. In the last years, the identification and characterisation of the enzymes involved in the synthesis of CβG allowed to know in detail the steps necessary for the formation of these sugar rings. Due to its peculiar structure, CβG can complex large hydrophobic molecules, a feature possibly related to its function in the interaction with the host. The capabilities of the CβG to function as molecular boxes and to solubilise hydrophobic compounds are attractive for application in the development of drugs, in food industry, nanotechnology, and chemistry. More importantly, its excellent immunomodulatory properties led to the proposal of CβG as a new class of adjuvants for vaccine development.
Collapse
Affiliation(s)
- Leticia S Guidolin
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, IIB-INTECH CONICET, Buenos Aires, Argentina
| | | | - Andrés E Ciocchini
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, IIB-INTECH CONICET, Buenos Aires, Argentina
| | - Diego J Comerci
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, IIB-INTECH CONICET, Buenos Aires, Argentina.,Comisión Nacional de Energía Atómica, CNEA, Grupo Pecuario, Centro Atómico Ezeiza, Buenos Aires, Argentina
| | | |
Collapse
|