1
|
Tang M, Lei Y, Chen K, Ding M, Ou Q, Tang J, Zhang Y, Tang T, Wang C. Reducing the degree of crosslinking of peptidoglycan in Listeria monocytogenes promoted the secretion of membrane vesicles. Biotechnol Bioeng 2024; 121:3629-3641. [PMID: 39014884 DOI: 10.1002/bit.28807] [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: 03/11/2024] [Revised: 06/18/2024] [Accepted: 06/23/2024] [Indexed: 07/18/2024]
Abstract
Listeria monocytogenes (LM) is a Gram-positive (G+) bacterium that secretes nanoscale membrane vesicles (MVs). LM MVs comprise various bacterial components and may have potential as an antigen or drug-delivery vehicle; however, the low yield of the LM MVs limits related research. G+-bacterial MVs germinate from the bacterial plasma membrane and must pass through a thick crosslinked peptidoglycan layer for release. Herein, we aimed to increase the release of MVs by reducing the degree of crosslinking of peptidoglycan. We knocked out two genes related to the longitudinal crosslinking of peptidoglycan, dal and dat, and supplemented the knocked-out dal gene through plasmid expression to obtain a stably inherited recombinant strain LMΔdd::pCW633. The structure, particle size, and main protein components of MVs secreted by this recombinant strain were consistent with those secreted from the wild strain, but the yield of MVs was considerably increased (p < 0.05). Furthermore, Listeria ivanovii (LI) was found to secrete MVs that differed in the composition of the main proteins compared with those of LM MVs. The abovementioned method was also feasible for promoting the secretion of MVs from the attenuated LM strain and LI wild-type and attenuated strains. Our study provides a new method to increase the secretion of MVs derived from Listeria that could be extended to other G+ bacteria.
Collapse
Affiliation(s)
- Mingyuan Tang
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yao Lei
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Kehan Chen
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Mingming Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Qian Ou
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jing Tang
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yunwen Zhang
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tian Tang
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chuan Wang
- Department of Public Health Laboratory Sciences, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Tang J, Song H, Li S, Lam SM, Ping J, Yang M, Li N, Chang T, Yu Z, Liu W, Lu Y, Zhu M, Tang Z, Liu Z, Guo YR, Shui G, Veillette A, Zeng Z, Wu N. TMEM16F Expressed in Kupffer Cells Regulates Liver Inflammation and Metabolism to Protect Against Listeria Monocytogenes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402693. [PMID: 39136057 PMCID: PMC11497084 DOI: 10.1002/advs.202402693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/20/2024] [Indexed: 10/25/2024]
Abstract
Infection by bacteria leads to tissue damage and inflammation, which need to be tightly controlled by host mechanisms to avoid deleterious consequences. It is previously reported that TMEM16F, a calcium-activated lipid scramblase expressed in various immune cell types including T cells and neutrophils, is critical for the control of infection by bacterium Listeria monocytogenes (Lm) in vivo. This function correlated with the capacity of TMEM16F to repair the plasma membrane (PM) damage induced in T cells in vitro, by the Lm toxin listeriolysin O (LLO). However, whether the protective effect of TMEM16F on Lm infection in vivo is mediated by an impact in T cells, or in other cell types, is not determined. Herein, the immune cell types and mechanisms implicated in the protective effect of TMEM16F against Lm in vivo are elucidated. Cellular protective effects of TMEM16F correlated with its capacity of lipid scrambling and augment PM fluidity. Using cell type-specific TMEM16F-deficient mice, the indication is obtained that TMEM16F expressed in liver Kupffer cells (KCs), but not in T cells or B cells, is key for protection against Listeria in vivo. In the absence of TMEM16F, Listeria induced PM rupture and fragmentation of KCs in vivo. KC death associated with greater liver damage, inflammatory changes, and dysregulated liver metabolism. Overall, the results uncovered that TMEM16F expressed in Kupffer cells is crucial to protect the host against Listeria infection. This influence is associated with the capacity of Kupffer cell-expressed TMEM16F to prevent excessive inflammation and abnormal liver metabolism.
Collapse
Affiliation(s)
- Jianlong Tang
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Hua Song
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
| | - Shimin Li
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100101China
| | - Jieming Ping
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Mengyun Yang
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Na Li
- Department of biochemistry and molecular biologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Teding Chang
- Department of Traumatic SurgeryTongji Trauma CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ze Yu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyNo. 1095 Jiefang AvenueWuhan430030China
| | - Weixiang Liu
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Yan Lu
- Department of Clinical ImmunologyThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Min Zhu
- Department of Thoracic SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zhaohui Tang
- Department of Traumatic SurgeryTongji Trauma CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zheng Liu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyNo. 1095 Jiefang AvenueWuhan430030China
| | - Yusong R. Guo
- Department of biochemistry and molecular biologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Cell Architecture Research CenterTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100101China
| | - André Veillette
- Laboratory of Molecular OncologyInstitut de recherches cliniques de Montréal (IRCM)MontréalQuébecH2W1R7Canada
- Department of MedicineUniversity of MontréalMontréalQuébecH3T 1J4Canada
- Department of MedicineMcGill UniversityMontréalQuébecH3G 1Y6Canada
| | - Zhutian Zeng
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
- Department of OncologyThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefei230001China
| | - Ning Wu
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
- Cell Architecture Research CenterTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| |
Collapse
|
3
|
Liu C, Qian R, Shi W, Kou L, Wang J, Ma X, Ren H, Gao S, Ren J. EⅡB Mutation Reduces the Pathogenicity of Listeria monocytogenes by Negatively Regulating Biofilm Formation Ability, Infective Capacity, and Virulence Gene Expression. Vet Sci 2024; 11:301. [PMID: 39057985 PMCID: PMC11281496 DOI: 10.3390/vetsci11070301] [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: 05/14/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
To explore the role of the membrane permease ⅡB (EⅡB) gene of Listeria pathogenicity island 4 (LIPI-4) in the virulence of Listeria monocytogenes, both an EⅡB deletion strain (∆EⅡB) and a complemented strain were constructed. In vitro experiments demonstrated that EⅡB deletion affected the biofilm formation ability of the wild-type strain (Lm928). Moreover, this deletion decreased the intracellular proliferation abilities of L. monocytogenes. Mice infected with ∆EⅡB survived longer and experienced less weight loss on days 1, 2, and 3 post-infection. The bacterial load in the liver tissue of ∆EⅡB-infected mice was significantly reduced, and a considerable decrease in the blood levels of inflammatory cytokines IL-β, IL-6, IL-10, and TNF-α were observed. Following EⅡB deletion, 65% (13/20) of genes were downregulated, 25% (5/20) were upregulated, and 10% (2/20) showed no change. These findings suggest that EⅡB deletion may reduce both the in vivo and in vitro virulence levels as well as the biofilm formation ability of Lm928 by downregulating the transcription levels of genes associated with virulence and biofilm formation. These findings provide a foundation for further examining the pathogenic mechanisms of LIPI-4 and EⅡB in L. monocytogenes.
Collapse
Affiliation(s)
| | | | | | | | - Jing Wang
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (C.L.); (R.Q.); (W.S.); (L.K.); (H.R.); (S.G.); (J.R.)
| | - Xun Ma
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (C.L.); (R.Q.); (W.S.); (L.K.); (H.R.); (S.G.); (J.R.)
| | | | | | | |
Collapse
|
4
|
Tieu MV, Pham DT, Cho S. Bacteria-based cancer therapy: Looking forward. Biochim Biophys Acta Rev Cancer 2024; 1879:189112. [PMID: 38761983 DOI: 10.1016/j.bbcan.2024.189112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/25/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
The field of bacteria-based cancer therapy, which focuses on the key role played by the prevalence of bacteria, specifically in tumors, in controlling potential targets for cancer therapy, has grown enormously over the past few decades. In this review, we discuss, for the first time, the global cancer situation and the timeline for using bacteria in cancer therapy. We also explore how interdisciplinary collaboration has contributed to the evolution of bacteria-based cancer therapies. Additionally, we address the challenges that need to be overcome for bacteria-based cancer therapy to be accepted in clinical trials and the latest advancements in the field. The groundbreaking technologies developed through bacteria-based cancer therapy have opened up new therapeutic strategies for a wide range of therapeutics in cancer.
Collapse
Affiliation(s)
- My-Van Tieu
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Republic of Korea
| | - Duc-Trung Pham
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Republic of Korea
| | - Sungbo Cho
- Department of Electronic Engineering, Gachon University, Seongnam-si 13120, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea.
| |
Collapse
|
5
|
De Greve H, Fioravanti A. Single domain antibodies from camelids in the treatment of microbial infections. Front Immunol 2024; 15:1334829. [PMID: 38827746 PMCID: PMC11140111 DOI: 10.3389/fimmu.2024.1334829] [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: 11/07/2023] [Accepted: 04/29/2024] [Indexed: 06/04/2024] Open
Abstract
Infectious diseases continue to pose significant global health challenges. In addition to the enduring burdens of ailments like malaria and HIV, the emergence of nosocomial outbreaks driven by antibiotic-resistant pathogens underscores the ongoing threats. Furthermore, recent infectious disease crises, exemplified by the Ebola and SARS-CoV-2 outbreaks, have intensified the pursuit of more effective and efficient diagnostic and therapeutic solutions. Among the promising options, antibodies have garnered significant attention due to their favorable structural characteristics and versatile applications. Notably, nanobodies (Nbs), the smallest functional single-domain antibodies of heavy-chain only antibodies produced by camelids, exhibit remarkable capabilities in stable antigen binding. They offer unique advantages such as ease of expression and modification and enhanced stability, as well as improved hydrophilicity compared to conventional antibody fragments (antigen-binding fragments (Fab) or single-chain variable fragments (scFv)) that can aggregate due to their low solubility. Nanobodies directly target antigen epitopes or can be engineered into multivalent Nbs and Nb-fusion proteins, expanding their therapeutic potential. This review is dedicated to charting the progress in Nb research, particularly those derived from camelids, and highlighting their diverse applications in treating infectious diseases, spanning both human and animal contexts.
Collapse
Affiliation(s)
- Henri De Greve
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Antonella Fioravanti
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
- VIB-VUB Center for Structural Biology, Vrije Universiteit Brussel, Brussels, Belgium
- Fondazione ParSeC – Parco delle Scienze e della Cultura, Prato, Italy
| |
Collapse
|
6
|
Zakaria AI, Sabala RF. Potential public health hazards related to consumption of poultry contaminated with antibiotic resistant Listeria monocytogenes in Egypt. BMC Microbiol 2024; 24:41. [PMID: 38287241 PMCID: PMC10823629 DOI: 10.1186/s12866-024-03183-x] [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: 05/18/2023] [Accepted: 01/05/2024] [Indexed: 01/31/2024] Open
Abstract
Listeria monocytogenes is an important foodborne pathogen that incorporated into many serious infections in human especially immunocompromised individuals, pregnant women, the elderly, and newborns. The consumption of food contaminated with such bacteria is considered a source of potential risk for consumers. Therefore, a total of 250 poultry purchased in highly popular poultry stores besides 50 swabs from workers hands in the same stores, in Mansoura City had been tested for the L. monocytogenes prevalence, virulence genes, and antibiotic resistance profile illustrating the health hazards from such poultry. The L. monocytogenes were recovered from 9.6% of poultry samples while not detected from workers hand swabs. The antimicrobial susceptibility of 24 L. monocytogenes strains against 24 antibiotics of seven different classes revealed high susceptibility rates to erythromycin (79.17%), streptomycin (66.67%), gentamycin (66.67%), vancomycin (58.33%), chloramphenicol (58.33%) and cefotaxime (41.67%). The majority (79.2%) of L. monocytogenes were classified as multidrug resistant strains with high resistance to tetracyclines and β-lactams antibiotics while 16.7% of the strains were categorized as extensively resistant ones. The iap virulence-specific determination gene had been detected in all recovered L. monocytogenes isolates while 83.33 and 70.83% of the isolates harbored hylA and actA genes. In addition, the study confirmed the capability of most L. monocytogenes isolates for biofilm formation by moderate to strong production and the quantitative risk assessment illustrated the risk of developing listeriosis as the risk value exceeded 100. The current results illustrate that poultry meat can be a source of pathogenic antibiotic resistant strains that may cause infection with limited or no treatment in immunosuppressed consumers via the food chain.
Collapse
Affiliation(s)
- Amira Ibrahim Zakaria
- Department of Food Hygiene, Safety and Technology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Rana Fahmi Sabala
- Department of Food Hygiene, Safety and Technology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
7
|
Elfmann C, Zhu B, Stülke J, Halbedel S. ListiWiki: A database for the foodborne pathogen Listeria monocytogenes. Int J Med Microbiol 2023; 313:151591. [PMID: 38043216 DOI: 10.1016/j.ijmm.2023.151591] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023] Open
Abstract
Listeria monocytogenes is a Gram positive foodborne pathogen that regularly causes outbreaks of systemic infectious diseases. The bacterium maintains a facultative intracellular lifestyle; it thrives under a variety of environmental conditions and is able to infect human host cells. L. monocytogenes is genetically tractable and therefore has become an attractive model system to study the mechanisms employed by facultative intracellular bacteria to invade eukaryotic cells and to replicate in their cytoplasm. Besides its importance for basic research, L. monocytogenes also serves as a paradigmatic pathogen in genomic epidemiology, where the relative stability of its genome facilitates successful outbreak detection and elucidation of transmission chains in genomic pathogen surveillance systems. In both terms, it is necessary to keep the annotation of the L. monocytogenes genome up to date. Therefore, we have created the database ListiWiki (http://listiwiki.uni-goettingen.de/) which stores comprehensive information on the widely used L. monocytogenes reference strain EDG-e. ListiWiki is designed to collect information on genes, proteins and RNAs and their relevant functional characteristics, but also further information such as mutant phenotypes, available biological material, and publications. In its present form, ListiWiki combines the most recent annotation of the EDG-e genome with published data on gene essentiality, gene expression and subcellular protein localization. ListiWiki also predicts protein-protein interactions networks based on protein homology to Bacillus subtilis proteins, for which detailed interaction maps have been compiled in the sibling database SubtiWiki. Furthermore, crystallographic information of proteins is made accessible through integration of Protein Structure Database codes and AlphaFold structure predictions. ListiWiki is an easy-to-use web interface that has been developed with a focus on an intuitive access to all information. Use of ListiWiki is free of charge and its content can be edited by all members of the scientific community after registration. In our labs, ListiWiki has already become an important and easy to use tool to quickly access genome annotation details that we can keep updated with advancing knowledge. It also might be useful to promote the comprehensive understanding of the physiology and virulence of an important human pathogen.
Collapse
Affiliation(s)
- Christoph Elfmann
- Department of General Microbiology, Göttingen Center for Molecular Biosciences, Georg-August University Göttingen, Göttingen, Germany
| | - Bingyao Zhu
- Department of General Microbiology, Göttingen Center for Molecular Biosciences, Georg-August University Göttingen, Göttingen, Germany
| | - Jörg Stülke
- Department of General Microbiology, Göttingen Center for Molecular Biosciences, Georg-August University Göttingen, Göttingen, Germany.
| | - Sven Halbedel
- FG11 Division of Enteropathogenic bacteria and Legionella, Robert Koch Institute, Burgstrasse 37, 38855 Wernigerode, Germany; Institute for Medical Microbiology and Hospital Hygiene, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
8
|
Yamazaki T, Nagatoishi S, Yamawaki T, Nozawa T, Matsunaga R, Nakakido M, Caaveiro JMM, Nakagawa I, Tsumoto K. Anti-InlA single-domain antibodies that inhibit the cell invasion of Listeria monocytogenes. J Biol Chem 2023; 299:105254. [PMID: 37716701 PMCID: PMC10582769 DOI: 10.1016/j.jbc.2023.105254] [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: 04/09/2023] [Revised: 08/24/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023] Open
Abstract
Listeriosis, caused by infection with Listeria monocytogenes, is a severe disease with a high mortality rate. The L. monocytogenes virulence factor, internalin family protein InlA, which binds to the host receptor E-cadherin, is necessary to invade host cells. Here, we isolated two single-domain antibodies (VHHs) that bind to InlA with picomolar affinities from an alpaca immune library using the phage display method. These InlA-specific VHHs inhibited the binding of InlA to the extracellular domains of E-cadherin in vitro as shown by biophysical interaction analysis. Furthermore, we determined that the VHHs inhibited the invasion of L. monocytogenes into host cells in culture. High-resolution X-ray structure analyses of the complexes of VHHs with InlA revealed that the VHHs bind to the same binding site as E-cadherin against InlA. We conclude that these VHHs have the potential for use as drugs to treat listeriosis.
Collapse
Affiliation(s)
- Taichi Yamazaki
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Satoru Nagatoishi
- Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, Tokyo, Japan.
| | - Tsukushi Yamawaki
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Takashi Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryo Matsunaga
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Makoto Nakakido
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Jose M M Caaveiro
- Laboratory of Global Healthcare, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan; Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, Tokyo, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
9
|
Matsuda Y, Yamauchi H, Hara H. Activation of inflammasomes and mechanisms for intracellular recognition of Listeria monocytogenes. Microbiol Immunol 2023; 67:429-437. [PMID: 37461376 DOI: 10.1111/1348-0421.13091] [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: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 10/06/2023]
Abstract
The high mortality rate associated with Listeria monocytogenes can be attributed to its ability to invade the body systemically and to activate inflammasomes. Both of these processes are facilitated by expressing a major virulence factor known as listeriolysin O, a 56 kDa pore-forming protein encoded by the hly gene. Listeriolysin O plays a crucial role in the pathogenesis of the bacterium by facilitating the escape of the pathogen from the phagosome into the cytosol. This process is essential for the successful establishment of infection. In addition, listeriolysin O is known as an immunomodulator that activates host signal transduction. In addition to listeriolysin O, Listeria expresses a variety of bacterial ligands, such as lipoteichoic acid, nucleotide, and flagellin, that are recognized by host intracellular pattern-recognition receptors including Nod-like receptors, AIM2-like receptors, and RIG-I-like receptors. This review introduces intracellular recognition of Listeria monocytogenes since recent studies have revealed that the activation of inflammasome exacerbates Gram-positive bacteria infection.
Collapse
Affiliation(s)
- Yasuyuki Matsuda
- Department of Infectious Diseases, Division of Microbiology and Immunochemistry, Asahikawa Medical University, Asahikawa, Japan
| | - Hajime Yamauchi
- Department of Infectious Diseases, Division of Microbiology and Immunochemistry, Asahikawa Medical University, Asahikawa, Japan
| | - Hideki Hara
- Department of Infectious Diseases, Division of Microbiology and Immunochemistry, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
10
|
Gan Y, Zhao G, Wang Z, Zhang X, Wu MX, Lu M. Bacterial Membrane Vesicles: Physiological Roles, Infection Immunology, and Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301357. [PMID: 37357142 PMCID: PMC10477901 DOI: 10.1002/advs.202301357] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/19/2023] [Indexed: 06/27/2023]
Abstract
Bacterial or fungal membrane vesicles, traditionally considered as microbial metabolic wastes, are secreted mainly from the outer membrane or cell membrane of microorganisms. However, recent studies have shown that these vesicles play essential roles in direct or indirect communications among microorganisms and between microorganisms and hosts. This review aims to provide an updated understanding of the physiological functions and emerging applications of bacterial membrane vesicles, with a focus on their biogenesis, mechanisms of adsorption and invasion into host cells, immune stimulatory effects, and roles in the much-concerned problem of bacterial resistance. Additionally, the potential applications of these vesicles as biomarkers, vaccine candidates, and drug delivery platforms are discussed.
Collapse
Affiliation(s)
- Yixiao Gan
- Department of Transfusion MedicineHuashan HospitalFudan UniversityShanghai200040P. R. China
| | - Gang Zhao
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200240P. R. China
| | - Zhicheng Wang
- Department of Transfusion MedicineHuashan HospitalFudan UniversityShanghai200040P. R. China
| | - Xingcai Zhang
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02138USA
| | - Mei X. Wu
- Wellman Center for PhotomedicineMassachusetts General HospitalDepartment of DermatologyHarvard Medical School, 50 Blossom StreetBostonMA02114USA
| | - Min Lu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200240P. R. China
| |
Collapse
|
11
|
Zare M, Farhadi A, Zare F, Dehbidi GR, Zarghampoor F, Ahmadi MKB, Behbahani AB. Genetically engineered E. coli invade epithelial cells and transfer their genetic cargo into the cells: an approach to a gene delivery system. Biotechnol Lett 2023:10.1007/s10529-023-03387-7. [PMID: 37166604 DOI: 10.1007/s10529-023-03387-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/12/2023]
Abstract
PURPOSE Despite advances in gene therapy, the lack of safe and efficient gene delivery systems limited the clinical effectiveness of gene therapy. Due to the inherent potential of bacteria, they can be considered as a good option for the gene transfer system. This study aimed to create a genetically engineered bacterium capable of entering epithelial cells and transferring its genetic cargo to the cell's cytoplasm, eventually expressing the gene of interest in the cell. METHODS The invasin (inv) gene from Yersinia pseudotuberculosis and the listeriolysin (hlyA) gene from Listeria monocytogenes were isolated by PCR assay and inserted into a pACYCDuet-1 vector. The recombinant plasmid was then transformed into E. coli strain BL21. Subsequently, pEGFP-C1 plasmids containing a CMV promoter were transformed into the engineered bacteria. Finally, the engineered bacteria containing the reporter genes were incubated with the HeLa and LNCaP cell lines. Fluorescence microscopy, flow cytometry, and TEM were used to monitor bacterial entry into the cells and gene expression. We used native E. coli strain BL21 as a control. RESULTS A fluorescence microscope showed that, in contrast to the control group, the manipulated E. coli were able to penetrate the cells and transport the plasmid pEGFP-C1 to the target cells. Flow cytometry also showed fluorescence intensity of 54.7% in HeLa cells and 71% in LNCaP cells, respectively. In addition, electron micrographs revealed the presence of bacteria in the cell endosomes and in the cytoplasm of the cells. CONCLUSION This study shows that genetically engineered E. coli can enter cells, transport cargo into cells, and induce gene expression in the target cell. In addition, flow cytometry shows that the gene transfer efficiency was sufficient for protein expression.
Collapse
Affiliation(s)
- Maryam Zare
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Division of Medical Biotechnology, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Farhadi
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Division of Medical Biotechnology, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farahnaz Zare
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Rafiei Dehbidi
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Zarghampoor
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Karimi Baba Ahmadi
- Department of Advanced Medical Technology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Abbas Behzad Behbahani
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
12
|
Bhattacharjee R, Nandi A, Sinha A, Kumar H, Mitra D, Mojumdar A, Patel P, Jha E, Mishra S, Rout PK, Panda PK, Suar M, Verma SK. Phage-tail-like bacteriocins as a biomedical platform to counter anti-microbial resistant pathogens. Biomed Pharmacother 2022; 155:113720. [PMID: 36162371 DOI: 10.1016/j.biopha.2022.113720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022] Open
Abstract
Phage Tail Like bacteriocins (PTLBs) has been an area of interest in the last couple of years owing to their varied application against multi-drug resistant (MDR), anti-microbial resistant (AMR) pathogens and their evolutionary link with the dsDNA virus and bacteriophages. PTLBs are defective phages derived from Myoviridae and Siphoviridae phages, PTLBs are distinguished into R-type (Rigid type) characterized by a non-flexible contractile nanotube resembling Myoviridae phage contractile tails, and F-type (Flexible type) with a flexible non-contractile rod-like structure similar to Siphoviridae phages. In this review, we have discussed the structural association, mechanism, and characterization of PTLBs. Moreover, we have elucidated the symbiotic biological function and application of PTLBs against MDR and XDR pathogens and highlighted the evolutionary role of PTLBs. The difficulties that must be overcome to implement PTLBs clinically are also discussed. It is imperative that these issues be addressed by academics in future studies before being implemented in clinical settings. This article is novel in its way as it will not only provide us with a gateway that acts as a novel strategy for scholars to mitigate and control the uprising issue of AMR pathogens but also promote the development of clinical studies for PTLBs.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Aditya Nandi
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Adrija Sinha
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Hrithik Kumar
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala 695551, India
| | - Disha Mitra
- University of Calcutta, 92, APC Road, Kolkata 700009, India
| | - Abhik Mojumdar
- Center for Research Equipment, Korea Basic Science Institute (KBSI), Ochang Center, Cheongju, Chungcheongbuk 28119, Republic of Korea; Department of Bio-Analytical Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Paritosh Patel
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Ealisha Jha
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Suman Mishra
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Prabhat Kumar Rout
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden.
| | - Mrutyunjay Suar
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India.
| | - Suresh K Verma
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden.
| |
Collapse
|
13
|
Mitchell MK, Ellermann M. Long Chain Fatty Acids and Virulence Repression in Intestinal Bacterial Pathogens. Front Cell Infect Microbiol 2022; 12:928503. [PMID: 35782143 PMCID: PMC9247172 DOI: 10.3389/fcimb.2022.928503] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
When bacterial pathogens enter the gut, they encounter a complex milieu of signaling molecules and metabolites produced by host and microbial cells or derived from external sources such as the diet. This metabolomic landscape varies throughout the gut, thus establishing a biogeographical gradient of signals that may be sensed by pathogens and resident bacteria alike. Enteric bacterial pathogens have evolved elaborate mechanisms to appropriately regulate their virulence programs, which involves sensing and responding to many of these gut metabolites to facilitate successful gut colonization. Long chain fatty acids (LCFAs) represent major constituents of the gut metabolome that can impact bacterial functions. LCFAs serve as important nutrient sources for all cellular organisms and can function as signaling molecules that regulate bacterial metabolism, physiology, and behaviors. Moreover, in several enteric pathogens, including Salmonella enterica, Listeria monocytogenes, Vibrio cholerae, and enterohemorrhagic Escherichia coli, LCFA sensing results in the transcriptional repression of virulence through two general mechanisms. First, some LCFAs function as allosteric inhibitors that decrease the DNA binding affinities of transcriptional activators of virulence genes. Second, some LCFAs also modulate the activation of histidine kinase receptors, which alters downstream intracellular signaling networks to repress virulence. This mini-review will summarize recent studies that have investigated the molecular mechanisms by which different LCFA derivatives modulate the virulence of enteric pathogens, while also highlighting important gaps in the field regarding the roles of LCFAs as determinants of infection and disease.
Collapse
|
14
|
Assessment of Listeria monocytogenes Surface Proteins Identified from Proteomics Analysis for Use as Diagnostic Biomarkers. Appl Environ Microbiol 2022; 88:e0003522. [PMID: 35477262 DOI: 10.1128/aem.00035-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Gram-positive bacterium Listeria monocytogenes is an important pathogen that causes a foodborne illness with a high percentage of fatalities. Surface proteins, specifically expressed from a wide range of L. monocytogenes serotypes under selective enrichment culture conditions, can serve as targets for the detection and isolation of this pathogen using antibody-based methods. Among a number of surface proteins identified by mass spectrometry in a previous proteomic study, six candidates (annotated as LMOf2365_0148, LMOf2365_0312, LMOf2365_0546, LMOf2365_1883, LMOf2365_2111, and LMOf2365_2742) were selected here for investigating their expression in the bacterial cells cultured in vitro by raising rabbit polyclonal antibodies (PAbs) against the recombinant form of each candidate. These protein candidates contained regions conserved among various L. monocytogenes isolates but variable in other Listeria species. LMOf2365_0148, an uncharacterized protein with a LPXTG motif accountable for covalent linkage to the cell wall peptidoglycan, exhibited a strong reaction signal from anti-LMOf2365_0148 PAb binding to the cell surface, as detected by immunofluorescence microscopy. Further study, through the generation of a panel of mouse monoclonal antibodies (MAbs) to the recombinant LMOf2365_0148, showed that one of the MAbs, M3686, reacted to bacterial isolates belonging to all three lineages of L. monocytogenes under Health Canada's standard enrichment culture conditions (MFHPB-07 and MFHPB-30). These results demonstrated the potential of using LMOf2365_0148 as a surface biomarker, in conjunction with specific MAbs developed here, for the isolation and detection of L. monocytogenes from foods and food processing environments. IMPORTANCE Strains of Listeria monocytogenes are differentiated serologically into at least 13 serotypes and grouped phylogenetically into 4 distinct lineages (I, II, III, and IV). No single monoclonal antibody (MAb) reported to date is capable of binding to the surface of L. monocytogenes strains representing all the serotypes. This study assessed the expression of six surface proteins selected from a previous proteomic study and demonstrated that surface protein LMOf2365_0148 has the greatest potential as a surface biomarker. A panel of 24 MAbs to LMOf2365_0148 were assessed extensively, revealing that one of the MAbs, M3686, reacted to a wide range of L. monocytogenes isolates (lineage I, II, and III isolates) grown under standard enrichment culture conditions and thus led to the conclusion that LMOf2365_0148 is a useful novel surface biomarker for identifying, detecting, and isolating the pathogen from food and environmental samples.
Collapse
|
15
|
Mo T, Wu F, Dou X, Wang D, Xia H, Li X. A Retrospective Study of Rare Listeria Meningoencephalitis in Immunocompetent Children in China. Front Neurol 2022; 13:827145. [PMID: 35309567 PMCID: PMC8924113 DOI: 10.3389/fneur.2022.827145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Listeria meningoencephalitis (LMM) is very rare in healthy children. We aimed to assess the clinical features, differential diagnosis, treatment options, and outcomes of LMM in immunocompetent children through a retrospective study. Methods The clinical symptoms, laboratory findings, imaging features, antibiotic use, and metagenomic next-generation sequencing (mNGS) results of the cerebrospinal fluid (CSF) were obtained from immunocompetent children who were diagnosed with LMM and admitted to the Xi'an Children's Hospital from May 2018 to July 2020. Results The data from 8 immunocompetent children were retrospectively analyzed in this study. The cohort included data from 5 males and 3 females who were aged from 1 year and 7 months to 16 years and 6 months. A total of 4 patients had chilled food before onset. The complications included hyponatremia (3/8), hydrocephalus (2/8), and hemophagocytic syndrome (1/8). In total, 8 patients were diagnosed with Listeria monocytogenes by positive CSF culture or mNGS results. The positive rate of CSF culture was 62.5% (5/8). A total of 5 patients conducted CSF mNGS, and the results of the mNGS were positive in 4 patients (80%, 4/5) and suspected in 1 patient. A total of 7 patients changed their therapeutic regimen to combined antibacterial therapies that included linezolid and meropenem (5/8), or ampicillin and meropenem (2/8). A total of 5 patients had favorable outcomes (Glasgow Outcome Scale, GOSE = 5) while two patients had unfavorable outcomes (GOSE = 1) and were complicated with hyponatremia and hydrocephalus. Conclusions Listeria meningoencephalitis (LMM) can occur in children with normal immune function and is commonly mistaken for other central nervous system infections. L. monocytogenes can be quickly and accurately detected by mNGS. Hyponatremia and hydrocephalus may indicate unfavorable outcomes.
Collapse
Affiliation(s)
- Tingting Mo
- Department of Pediatric Neurology, Xi'an Children's Hospital, Xi'an, China
| | - Fang Wu
- Department of Pediatric Neurology, Xi'an Children's Hospital, Xi'an, China
| | - Xiangjun Dou
- Department of Pediatric Neurology, Xi'an Children's Hospital, Xi'an, China
| | - Dong Wang
- Department of Pediatric Neurology, Xi'an Children's Hospital, Xi'an, China
| | - Han Xia
- Department of Scientific Affaires, Hugobiotech Co., Ltd., Beijing, China
| | - Xia Li
- Department of Pediatric Neurology, Xi'an Children's Hospital, Xi'an, China
| |
Collapse
|
16
|
Liang S, Hu X, Wang R, Fang M, Yu Y, Xiao X. The combination of thymol and cinnamaldehyde reduces the survival and virulence of Listeria monocytogenes on autoclaved chicken breast. J Appl Microbiol 2022; 132:3937-3950. [PMID: 35178822 DOI: 10.1111/jam.15496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/24/2022] [Accepted: 02/14/2022] [Indexed: 11/28/2022]
Abstract
AIMS To reveal the antibacterial mechanism of the combination of thymol and cinnamaldehyde to Listeria monocytogenes ATCC 19115 on autoclaved chicken breast. METHODS AND RESULTS In this study, Listeria monocytogenes ATCC 19115 on autoclaved chicken breast was exposed to the stress of 125 μg/mL thymol and 125 μg/mL cinnamaldehyde, and transcriptome analysis was used to reveal the crucial antibacterial mechanism. According to the results, 1303 significantly differentially expressed genes (DEGs) were identified. Treated by thymol and cinnamaldehyde in combination, pyrimidine and branched-chain amino acids biosynthesis of L. monocytogenes were thwarted which impairs its nucleic acid biosynthesis and intracellular metabolism. The up-regulated DEGs involved in membrane composition and function contributed to membrane repair. Besides, pyruvate catabolism and TCA cycle were restrained which brought about the disturbance of amino acid metabolism. ABC transporters were also perturbed, for instance, the uptake of cysteine, D-methionine and betaine was activated, while the uptake of vitamin, iron and carnitine was repressed. Thus, L. monocytogenes tended to activate PTS, glycolysis, glycerol catabolism, and pentose phosphate pathways to obtain energy to adapt to the hostile condition. Noticeably, DEGs involved in virulence factors were totally down-regulated, including genes devoted to encoding flagella, chemotaxis, biofilm formation, internalin as well as virulence gene clusters. CONCLUSIONS The combination of thymol and cinnamaldehyde is effective to reduce the survival and potential virulence of L. monocytogenes on autoclaved chicken breast. SIGNIFICANCE AND IMPACT OF STUDY This work contributes to providing theoretical information for the application and optimization of thymol and cinnamaldehyde in ready-to-eat meat products to inhibit L. monocytogenes.
Collapse
Affiliation(s)
- Siwei Liang
- Research Center of Food Safety and Detection, School of Food Sciences and Engineering, South China University of Technology, Guangdong Province, Guangzhou City, 510640, China
| | - Xinyi Hu
- Research Center of Food Safety and Detection, School of Food Sciences and Engineering, South China University of Technology, Guangdong Province, Guangzhou City, 510640, China
| | - Ruifei Wang
- Research Center of Food Safety and Detection, School of Food Sciences and Engineering, South China University of Technology, Guangdong Province, Guangzhou City, 510640, China
| | - Meimei Fang
- Research Center of Food Safety and Detection, School of Food Sciences and Engineering, South China University of Technology, Guangdong Province, Guangzhou City, 510640, China
| | - Yigang Yu
- Research Center of Food Safety and Detection, School of Food Sciences and Engineering, South China University of Technology, Guangdong Province, Guangzhou City, 510640, China
| | - Xinglong Xiao
- Research Center of Food Safety and Detection, School of Food Sciences and Engineering, South China University of Technology, Guangdong Province, Guangzhou City, 510640, China
| |
Collapse
|
17
|
Boichis E, Ran Sapir S, Herskovits AA. Bone Marrow-Derived Macrophage (BMDM ) Infection by Listeria monocytogenes. Methods Mol Biol 2022; 2427:83-93. [PMID: 35619027 DOI: 10.1007/978-1-0716-1971-1_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Listeria monocytogenes is a gram-positive bacterium adapted to life as both an environmental saprophyte and a pathogenic parasite of mammalian hosts, with a transcriptomic program tailored for each niche. Study of the L. monocytogenes pathogenic lifestyle requires conditions that mimic the mammalian niche. Of the myriad experimental models used to achieve such conditions, the bone marrow-derived macrophage (BMDM) is a relatively simple and reliable primary immune cell model for L. monocytogenes infections. Here we describe the extraction, preparation, and storage of BMDMs and their use in L. monocytogenes infection experiments.
Collapse
Affiliation(s)
- Etai Boichis
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shai Ran Sapir
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Anat A Herskovits
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
18
|
Shi D, Anwar TM, Pan H, Chai W, Xu S, Yue M. Genomic Determinants of Pathogenicity and Antimicrobial Resistance for 60 Global Listeria monocytogenes Isolates Responsible for Invasive Infections. Front Cell Infect Microbiol 2021; 11:718840. [PMID: 34778102 PMCID: PMC8579135 DOI: 10.3389/fcimb.2021.718840] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/15/2021] [Indexed: 12/18/2022] Open
Abstract
Listeria monocytogenes remains a significant public health threat, causing invasive listeriosis manifested as septicemia, meningitis, and abortion, with up to 30% of cases having a fatal outcome. Tracking the spread of invasive listeriosis requires an updated knowledge for virulence factors (VFs) and antimicrobial resistance features, which is an essential step toward its clinical diagnosis and treatment. Taking advantage of high-throughput genomic sequencing, we proposed that the differential genes based on the pathogenomic composition could be used to evaluate clinical observations and therapeutic options for listeriosis. Here, we performed the comparative genomic analysis of 60 strains from five continents with a diverse range of sources, representing serotypes 1/2a, 1/2b, 1/2c, and 4b, comprising lineage I and lineage II and including 13 newly contributed Chinese isolates from clinical cases. These strains were associated with globally distributed clonal groups linked with confirmed foodborne listeriosis outbreak and sporadic cases. We found that L. monocytogenes strains from clonal complex (CC) CC8, CC7, CC9, and CC415 carried most of the adherence and invasive genes. Conversely, CC1, CC2, CC4, and CC6 have the least number of adherence and invasive genes. Additionally, Listeria pathogenicity island-1 (LIPI-1), LIPI-2, intracellular survival, surface anchoring, and bile salt resistance genes were detected in all isolates. Importantly, LIPI-3 genes were harbored in CC3, CC224, and ST619 of the Chinese isolates and in CC1, CC4, and CC6 of other worldwide isolates. Notably, Chinese isolates belonging to CC14 carried antibiotic resistance genes (ARGs) against β-lactams (blaTEM-101, blaTEM-105) and macrolide (ermC-15), whereas CC7 and CC8 isolates harbored ARGs against aminoglycoside (aadA10_2, aadA6_1), which may pose a threat to therapeutic efficacy. Phylogenomic analysis showed that CC8, CC7, and CC5 of Chinese isolates, CC8 (Swiss and Italian isolates), and CC5 and CC7 (Canadian isolates) are closely clustered together and belonged to the same CC. Additionally, CC381 and CC29 of Chinese isolates shared the same genomic pattern as CC26 of Swiss isolate and CC37 of Canadian isolate, respectively, indicating strong phylogenomic relation between these isolates. Collectively, this study highlights considerable clonal diversity with well-recognized virulence and antimicrobial-resistant determinants among Chinese and worldwide isolates that stress to design improved strategies for clinical therapies.
Collapse
Affiliation(s)
- Dawei Shi
- Division II of In Vitro Diagnostics for Infectious Diseases, Institute for In Vitro Diagnostics Control, National Institutes for Food and Drug Control, Beijing, China
| | - Tanveer Muhammad Anwar
- Institute of Preventive Veterinary Sciences & Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Hang Pan
- Institute of Preventive Veterinary Sciences & Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Wenqin Chai
- Institute of Preventive Veterinary Sciences & Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Sihong Xu
- Division II of In Vitro Diagnostics for Infectious Diseases, Institute for In Vitro Diagnostics Control, National Institutes for Food and Drug Control, Beijing, China
| | - Min Yue
- Institute of Preventive Veterinary Sciences & Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Hainan Institute of Zhejiang University, Sanya, China
| |
Collapse
|
19
|
Planas A. Peptidoglycan Deacetylases in Bacterial Cell Wall Remodeling and Pathogenesis. Curr Med Chem 2021; 29:1293-1312. [PMID: 34525907 DOI: 10.2174/0929867328666210915113723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 11/22/2022]
Abstract
The bacterial cell wall peptidoglycan (PG) is a dynamic structure that is constantly synthesized, re-modeled and degraded during bacterial division and growth. Post-synthetic modifications modulate the action of endogenous autolysis during PG lysis and remodeling for growth and sporulation, but also they are a mechanism used by pathogenic bacteria to evade the host innate immune system. Modifica-tions of the glycan backbone are limited to the C-2 amine and the C-6 hydroxyl moieties of either Glc-NAc or MurNAc residues. This paper reviews the functional roles and properties of peptidoglycan de-N-acetylases (distinct PG GlcNAc and MurNAc deacetylases) and recent progress through genetic stud-ies and biochemical characterization to elucidate their mechanism of action, 3D structures, substrate specificities and biological functions. Since they are virulence factors in pathogenic bacteria, peptidogly-can deacetylases are potential targets for the design of novel antimicrobial agents.
Collapse
Affiliation(s)
- Antoni Planas
- Laboratory of Biochemistry, Institut Químic de Sarrià. University Ramon Llull, 08017 Barcelona. Spain
| |
Collapse
|
20
|
Shahid AD, Lu Y, Iqbal MA, Lin L, Huang S, Jiang X, Chen S. Listeria monocytogenes crosses blood brain barrier through Rho GTPases induced migration of macrophages and inflammatory interleukin expression. Microb Pathog 2021; 159:105143. [PMID: 34400281 DOI: 10.1016/j.micpath.2021.105143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 10/20/2022]
Abstract
Listeria monocytogenes crossing the blood-brain barrier in the form of "Trojan Horse" is of great significance for the establishment of bacterial encephalitis and meningitis. Induction of cell migration and crossing the blood-brain barrier is very important to understand the Listeria pathogenesis. The Rho GTPases family is considered a key factor in regulating cell migration. This study was designed to investigate the expression of Rho GTPases and their effect on the behavior of cell migration and the stimulation of immune factors. Selective Rho GTPases were investigated by real-time PCR and Western blot. Among these, the expression of RhoA was significantly increased following the infection of Listeria monocytogenes in macrophages. Further, we found that RhoA improves the migration of macrophages and expression of IL-1β, IL-6, and TNF-α. The expression of IL-1β, IL-6 and TNF-α possibly facilitates the migration and adhesion of macrophages to cross the blood-brain barrier. This study provides preliminary ground to investigate the detailed mechanism of Listeria monocytogenes crossing the blood-brain barrier.
Collapse
Affiliation(s)
| | - Ye Lu
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China; Department of Clinical Laboratory, Yixing People's Hospital, Affiliated Jiangsu University, Wuxi, 214200, China
| | | | - Lin Lin
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Shuang Huang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xugan Jiang
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Shengxia Chen
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
21
|
Susmitha A, Bajaj H, Madhavan Nampoothiri K. The divergent roles of sortase in the biology of Gram-positive bacteria. ACTA ACUST UNITED AC 2021; 7:100055. [PMID: 34195501 PMCID: PMC8225981 DOI: 10.1016/j.tcsw.2021.100055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 12/16/2022]
Abstract
The bacterial cell wall contains numerous surface-exposed proteins, which are covalently anchored and assembled by a sortase family of transpeptidase enzymes. The sortase are cysteine transpeptidases that catalyzes the covalent attachment of surface protein to the cell wall peptidoglycan. Among the reported six classes of sortases, each distinct class of sortase plays a unique biological role in anchoring a variety of surface proteins to the peptidoglycan of both pathogenic and non-pathogenic Gram-positive bacteria. Sortases not only exhibit virulence and pathogenesis properties to host cells, but also possess a significant role in gut retention and immunomodulation in probiotic microbes. The two main distinct functions are to attach proteins directly to the cell wall or assemble pili on the microbial surface. This review provides a compendium of the distribution of different classes of sortases present in both pathogenic and non-pathogenic Gram-positive bacteria and also the noteworthy role played by them in bacterial cell wall assembly which enables each microbe to effectively interact with its environment.
Collapse
Affiliation(s)
- Aliyath Susmitha
- Microbial Processes and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Harsha Bajaj
- Microbial Processes and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India
| | - Kesavan Madhavan Nampoothiri
- Microbial Processes and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
22
|
Lopes-Luz L, Mendonça M, Bernardes Fogaça M, Kipnis A, Bhunia AK, Bührer-Sékula S. Listeria monocytogenes: review of pathogenesis and virulence determinants-targeted immunological assays. Crit Rev Microbiol 2021; 47:647-666. [PMID: 33896354 DOI: 10.1080/1040841x.2021.1911930] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Listeria monocytogenes is one of the most invasive foodborne pathogens and is responsible for numerous outbreaks worldwide. Most of the methods to detect this bacterium in food require selective enrichment using traditional bacterial culture techniques that can be time-consuming and labour-intensive. Moreover, molecular methods are expensive and need specific technical knowledge. In contrast, immunological approaches are faster, simpler, and user-friendly alternatives and have been developed for the detection of L. monocytogenes in food, environmental, and clinical samples. These techniques are dependent on the constitutive expression of L. monocytogenes antigens and the specificity of the antibodies used. Here, updated knowledge on pathogenesis and the key immunogenic virulence determinants of L. monocytogenes that are used for the generation of monoclonal and polyclonal antibodies for the serological assay development are summarised. In addition, immunological approaches based on enzyme-linked immunosorbent assay, immunofluorescence, lateral flow immunochromatographic assays, and immunosensors with relevant improvements are highlighted. Though the sensitivity and specificity of the assays were improved significantly, methods still face many challenges that require further validation before use.
Collapse
Affiliation(s)
- Leonardo Lopes-Luz
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brasil
| | - Marcelo Mendonça
- Curso de Medicina Veterinária, Universidade Federal do Agreste de Pernambuco, Garanhuns, Brasil
| | | | - André Kipnis
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brasil
| | - Arun K Bhunia
- Department of Food Science, Purdue University, West Lafayette, IN, USA.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA.,Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Samira Bührer-Sékula
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brasil
| |
Collapse
|
23
|
Gao A, Tang H, Zhang Q, Liu R, Wang L, Liu Y, Qi Z, Shen Y. Mst1/2-ALK promotes NLRP3 inflammasome activation and cell apoptosis during Listeria monocytogenes infection. J Microbiol 2021; 59:681-692. [PMID: 33877580 DOI: 10.1007/s12275-021-0638-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/15/2021] [Accepted: 03/05/2021] [Indexed: 10/21/2022]
Abstract
Listeria monocytogenes (L. monocytogenes) is a Gram-positive intracellular foodborne pathogen that causes severe diseases, such as meningitis and sepsis. The NLR family pyrin domain-containing 3 (NLRP3) inflammasome has been reported to participate in host defense against pathogen infection. However, the exact molecular mechanisms underlying NLRP3 inflammasome activation remain to be fully elucidated. In the present study, the roles of mammalian Ste20-like kinases 1/2 (Mst1/2) and Anaplastic Lymphoma Kinase (ALK) in the activation of the NLRP3 inflammasome induced by L. monocytogenes infection were investigated. The expression levels of Mst1/2, phospho (p)-ALK, p-JNK, Nek7, and NLRP3 downstream molecules including activated cas-pase-1 (p20) and mature interleukin (IL)-1β (p17), were up-regulated in L. monocytogenes-infected macrophages. The ALK inhibitor significantly decreased the expression of p-JNK, Nek7, and NLRP3 downstream molecules in macrophages infected with L. monocytogenes. Furthermore, the Mst1/2 inhibitor markedly inhibited the L. monocytogenes-induced activation of ALK, subsequently downregulating the expression of p-JNK, Nek7, and NLRP3 downstream molecules. Therefore, our study demonstrated that Mst1/2-ALK mediated the activation of the NLRP3 inflammasome by promoting the interaction between Nek7 and NLRP3 via JNK during L. monocytogenes infection, which subsequently increased the maturation and release of proinflammatory cytokine to resist pathogen infection. Moreover, Listeriolysin O played a key role in the process. In addition, we also found that the L. monocytogenes-induced apoptosis of J774A.1 cells was reduced by the Mst1/2 or ALK inhibitor. The present study reported, for the first time, that the Mst1/2-ALK-JNK-NLRP3 signaling pathway plays a vital proinflammatory role during L. monocytogenes infection.
Collapse
Affiliation(s)
- Aijiao Gao
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, P. R. China
| | - Huixin Tang
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, P. R. China
| | - Qian Zhang
- Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin, 300071, P. R. China
| | - Ruiqing Liu
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, P. R. China
| | - Lin Wang
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, P. R. China
| | - Yashan Liu
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, P. R. China
| | - Zhi Qi
- Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin, 300071, P. R. China.
| | - Yanna Shen
- School of Medical Laboratory, Tianjin Medical University, Tianjin, 300203, P. R. China.
| |
Collapse
|
24
|
PDCD4-mediated downregulation of Listeria monocytogenes burden in macrophages. Cent Eur J Immunol 2021; 46:38-46. [PMID: 33897282 PMCID: PMC8056355 DOI: 10.5114/ceji.2021.105244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/03/2020] [Indexed: 11/17/2022] Open
Abstract
Introduction Macrophages are effector cells of the innate immune system and defend against invading pathogens. Previous reports have shown that infection with Listeria monocytogenes upregulates miR-21a expression in macrophages. Aim of the study We aimed to verify whether programmed cell death 4 (PDCD4) is involved in the high bacterial burden observed in macrophages during late-stage L. monocytogenes infections. Material and methods We examined the expression of miR-21a and its known target PDCD4 in macrophages after L. monocytogenes infection. The macrophages’ uptake ability of L. monocytogenes was measured using FluoSpheres Carboxylate-modified microspheres. We depleted PDCD4 by transfecting macrophages with siPDCD4. Results In macrophages, PDCD4 protein was downregulated 5 h, but not 2 h, after L. monocytogenes infection. Our results validated the hypothesis that PDCD4-depleted macrophages present a higher L. monocytogenes burden. Moreover, we found that the activation of c-Jun and STAT3 accompanied PDCD4 downregulation. Conclusions Our results showed that PDCD4 mediated the suppression of L. monocytogenes infection in macrophages via c-Jun/STAT3 signalling activation.
Collapse
|
25
|
Collu D, Marras L, Sanna A, Carrucciu G, Pinna A, Carraro V, Sanna G, Coroneo V. Evaluation of growth potential and growth dynamics of Listeria monocytogenes on ready-to-eat fresh fruit. Ital J Food Saf 2021; 10:9337. [PMID: 33907689 PMCID: PMC8056446 DOI: 10.4081/ijfs.2021.9337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/22/2021] [Indexed: 11/23/2022] Open
Abstract
The consumption of fresh or RTE fruits is increasing every year and Listeria monocytogenes has been identified on raw or minimally processed fruits. A food product can become contaminated with L. monocytogenes anywhere along the pathway of food production during planting, harvesting, packaging, distribution and serving. The aim of this work was to assess the microbiological risks associated with consumption of ready- to- eat fruit such as melon, pineapple, coconut and fruit salad. The presence of Escherichia coli, Salmonella spp. and L. monocytogenes was also evaluated. Microbiological challenge tests were carried out for the evaluation of the L. monocytogenes growth potential in RTE fruit stored at 4 and 8°C. E. coli counts resulted under the detection limit of 10 CFU g-1, Salmonella and L. monocytogenes were not detected (absence in 25g). The growth potential values in coconut and melon (δ>0.5) showed the growth capacity of Listeria at the temperatures considered. A low initial load, also derived from good hygiene practices, and correct storage temperatures are essential to reduce bacterial growth in RTE fruit. The challenge test showed how each type of RTE fruit has a different commercial life based on its specific growth potential and that food should be stored at temperatures not higher than 4°C for a short period.
Collapse
Affiliation(s)
- David Collu
- Department of Medical Sciences and Public Health, University of Cagliari
| | - Luisa Marras
- Department of Medical Sciences and Public Health, University of Cagliari
| | - Adriana Sanna
- Department of Medical Sciences and Public Health, University of Cagliari
| | - Gerolamo Carrucciu
- Department of Medical Sciences and Public Health, University of Cagliari
| | - Antonella Pinna
- Department of Medical Sciences and Public Health, University of Cagliari
| | - Valentina Carraro
- Department of Medical Sciences and Public Health, University of Cagliari
| | - Giuseppina Sanna
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Valentina Coroneo
- Department of Medical Sciences and Public Health, University of Cagliari
| |
Collapse
|
26
|
Cardiotropic Isolates of Listeria monocytogenes with Enhanced Vertical Transmission Dependent upon the Bacterial Surface Protein InlB. Infect Immun 2021; 89:IAI.00321-20. [PMID: 33139387 DOI: 10.1128/iai.00321-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022] Open
Abstract
Listeria monocytogenes is a facultative Gram-positive intracellular bacterium that is capable of causing serious invasive infections in pregnant women, resulting in abortion, still-birth, and disseminated fetal infection. Previously, a clinical L. monocytogenes isolate, 07PF0776, was identified as having an enhanced ability to target cardiac tissue. This tissue tropism appeared to correlate with amino acid variations found within internalin B (InlB), a bacterial surface protein associated with host cell invasion. Given that the mammalian receptor bound by InlB, Met, is abundantly expressed by placental tissue, we assessed isolate 07PF0776 for its ability to be transmitted from mother to fetus. Pregnant Swiss Webster mice were infected on gestational day E13 via tail vein injection with the standard isolate 10403S, a noncardiotropic strain, or 07PF0776, the cardiac isolate. Pregnant mice infected with 07PF0776 exhibited significantly enhanced transmission of L. monocytogenes to placentas and fetuses compared to 10403S. Both bacterial burdens and the frequency of placental and fetal infection were increased in mice infected with the cardiac isolate. Strain 07PF0776 also exhibited an enhanced ability to invade Jar human trophoblast tissue culture cells in comparison to 10403S, and was found to have increased levels of InlB associated with the bacterial cell surface. Overexpression of surface InlB via genetic manipulation was sufficient to confer enhanced invasion of the placenta and fetus to both 10403S and 07PF0776. These data support a central role for surface InlB in promoting vertical transmission of L. monocytogenes.
Collapse
|
27
|
Henderson LO, Gaballa A, Orsi RH, Boor KJ, Wiedmann M, Guariglia-Oropeza V. Transcriptional profiling of the L. monocytogenes PrfA regulon identifies six novel putative PrfA-regulated genes. FEMS Microbiol Lett 2020; 367:5998225. [PMID: 33220686 DOI: 10.1093/femsle/fnaa193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/19/2020] [Indexed: 12/28/2022] Open
Abstract
The transcriptional activator Positive Regulatory Factor A (PrfA) regulates expression of genes essential for virulence in Listeria monocytogenes. To define the PrfA regulon, the 10403S wildtype (WT) strain, a constitutively active prfA* mutant, and an isogenic ∆prfA mutant were grown under PrfA-inducing conditions in a medium containing glucose-1-phosphate and pre-treated with 0.2% activated charcoal. RNA-seq-generated transcript levels were compared as follows: (i) prfA* and WT; (ii) WT and ∆prfA and (iii) prfA* and ∆prfA. Significantly higher transcript levels in the induced WT or constitutively active PrfA* were identified for 18 genes and 2 ncRNAs in at least one of the three comparisons. These genes included: (i) 10/12 of the genes previously identified as directly PrfA-regulated; (ii) 2 genes previously identified as PrfA-regulated, albeit likely indirectly; and (iii) 6 genes newly identified as PrfA-regulated, including one (LMRG_0 2046) with a σA-dependent promoter and PrfA box located within an upstream open reading frame. LMRG_0 2046, which encodes a putative cyanate permease, is reported to be downregulated by a σB-dependent anti-sense RNA. This newly identified overlap between the σB and PrfA regulons highlights the complexity of regulatory networks important for fine-tuning bacterial gene expression in response to the rapidly changing environmental conditions associated with infection.
Collapse
Affiliation(s)
- L O Henderson
- Department of Food Science, Cornell University, 352 Stocking Hall Ithaca, NY 14853, USA
| | - A Gaballa
- Department of Food Science, Cornell University, 352 Stocking Hall Ithaca, NY 14853, USA
| | - R H Orsi
- Department of Food Science, Cornell University, 352 Stocking Hall Ithaca, NY 14853, USA
| | - K J Boor
- Department of Food Science, Cornell University, 352 Stocking Hall Ithaca, NY 14853, USA
| | - M Wiedmann
- Department of Food Science, Cornell University, 352 Stocking Hall Ithaca, NY 14853, USA
| | - V Guariglia-Oropeza
- Department of Food Science, Cornell University, 352 Stocking Hall Ithaca, NY 14853, USA
| |
Collapse
|
28
|
Harrand AS, Strawn LK, Illas-Ortiz PM, Wiedmann M, Weller D. Listeria monocytogenes Prevalence Varies More within Fields Than between Fields or over Time on Conventionally Farmed New York Produce Fields. J Food Prot 2020; 83:1958-1966. [PMID: 32609818 PMCID: PMC7995327 DOI: 10.4315/jfp-20-120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022]
Abstract
ABSTRACT Past studies have shown that the on-farm distribution of Listeria monocytogenes is affected by environmental factors (e.g., weather). However, most studies were conducted at large scales (e.g., across farms), whereas few studies examined drivers of L. monocytogenes prevalence at smaller scales (e.g., within a single field). This study was performed to address this knowledge gap by (i) tracking L. monocytogenes distribution in two fields on one farm over a growing season and (ii) identifying factors associated with L. monocytogenes isolation from drag swab, soil, and agricultural water samples. Overall, L. monocytogenes was detected in 78% (21 of 27), 19% (7 of 36), and 8% (37 of 486) of water, drag swab, and soil samples, respectively. All isolates were characterized by pulsed-field gel electrophoresis. Of the 43 types identified, 14 were isolated on multiple sampling visits and/or from multiple sample types, indicating persistence in or repeated introduction into the farm environment during the study. Our findings also suggest that L. monocytogenes prevalence, even at the small spatial scale studied here, (i) was not uniform and (ii) varied more within fields than between fields or over time. This is illustrated by plot (in-field variation), field (between-field variation), and sampling visit (time), accounting for 18, 2, and 3% of variance in odds of isolating L. monocytogenes, respectively. Moreover, according to random forest analysis, water-related factors were among the top-ranked factors associated with L. monocytogenes isolation from all sample types. For example, the likelihood of isolating L. monocytogenes from drag and soil samples increased monotonically as rainfall increased. Overall, findings from this single-farm study suggests that mitigation strategies for L. monocytogenes in produce fields should focus on water-associated risk factors (e.g., rain and distance to water) and be tailored to specific high-risk in-field areas. HIGHLIGHTS
Collapse
Affiliation(s)
- A. S. Harrand
- Department of Food Science, Cornell University, 354 Stocking Hall, Ithaca, NY 14853, USA
| | - Laura. K. Strawn
- Department of Food Science, Cornell University, 354 Stocking Hall, Ithaca, NY 14853, USA,Department of Food Science and Technology, Eastern Shore Agriculture Research and Extension Center, Virginia Polytechnic Institute and State University, 33446 Research Drive, Painter, VA 23420, USA
| | | | - Martin Wiedmann
- Department of Food Science, Cornell University, 354 Stocking Hall, Ithaca, NY 14853, USA
| | - Daniel Weller
- Department of Food Science, Cornell University, 354 Stocking Hall, Ithaca, NY 14853, USA,Present affiliation: Department of Biostatistics and Computational Biology, University of Rochester, 265 Crittenden Boulevard, Rochester, NY 14642, USA,Corresponding author: Daniel Weller, /
| |
Collapse
|
29
|
Kannan S, Balakrishnan J, Govindasamy A. Listeria monocytogens - Amended understanding of its pathogenesis with a complete picture of its membrane vesicles, quorum sensing, biofilm and invasion. Microb Pathog 2020; 149:104575. [PMID: 33091581 DOI: 10.1016/j.micpath.2020.104575] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 12/29/2022]
Abstract
Listeria monocytogenes is a ubiquitous, intracellular foodborne pathogen that causes listeriosis in animals and humans. Pathogenic Listeria monocytogenes easily adapted to the conditions of human gastrointestinal tract and tolerate the counter changes such as acidity, bile, osmolarity, and antimicrobial peptides. They secrete specialized biologically active extra organ called membrane vesicles which comprises proteins, lipids, and lipopolysaccharides. Listerial vesicles possess functional versatility and play a significant role in pathogenesis by cell-free intercellular communication and toxin packaging. L. monocytogenes can attach promptly and decisively to inert substratum including intestinal mucosa, and forms biofilms and causes detrimental effects. Further, they invade the host cells through quorum sensing (QS) controlled virulence determinants and biofilms. The precise degree to which the bacterium retains the intracellular ambiance of host cells remains unknown. The machinery associated with intracellular survival, and the role of membrane vesicles, quorum sensing, and the Agr system in Listeria monocytogenes largely remains unclear. The current review focused to understand the role of membrane vesicles mediated pathogenesis biofilms, and delivers auxiliary impetus to understanding the potentials of virulence mediated invasion in Listeria monocytogenes.
Collapse
Affiliation(s)
- Suganya Kannan
- Central Research Laboratory, Vinayaka Mission's Medical College and Hospital, Vinayaka Mission's Research Foundation (Deemed to be University), Karaikal, India.
| | - Jeyakumar Balakrishnan
- Central Research Laboratory, Vinayaka Mission's Medical College and Hospital, Vinayaka Mission's Research Foundation (Deemed to be University), Karaikal, India
| | - Ambujam Govindasamy
- Department of General Surgery, Vinayaka Mission's Medical College and Hospital, Vinayaka Mission Research Foundation (Deemed to be University), Karaikal, India
| |
Collapse
|
30
|
Nowak J, Visnovsky SB, Cruz CD, Fletcher GC, van Vliet AHM, Hedderley D, Butler R, Flint S, Palmer J, Pitman AR. Inactivation of the gene encoding the cationic antimicrobial peptide resistance factor MprF increases biofilm formation but reduces invasiveness of Listeria monocytogenes. J Appl Microbiol 2020; 130:464-477. [PMID: 32687650 DOI: 10.1111/jam.14790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 06/18/2020] [Accepted: 07/14/2020] [Indexed: 11/27/2022]
Abstract
AIMS To understand the genetics involved in surface attachment and biofilm formation of Listeria monocytogenes. METHODS AND RESULTS An in vitro screen of a Himar1 transposon library of L. monocytogenes strain 15G01 identified three transposants that produced significantly different biofilm levels when compared to the wild-type strain; two mutants exhibited enhanced biofilm formation and one produced less biofilm biomass than the wild-type. The mutant 15G01 mprF::Himar1, which had a transposon insertion in the mprF gene, was selected for further analysis. The mutant produced a more densely populated biofilm on solid surfaces such as stainless steel and polystyrene, as determined using scanning electron and light microscopy. The 15G01 mprF::Himar1 mutant remained viable in biofilms, but showed an increase in sensitivity to the cationic antimicrobial gallidermin. The mutant also displayed reduced invasiveness in CaCo-2 intestinal cells, suggesting virulence properties are compromised by the inactivation of mprF. CONCLUSIONS Biofilm formation and gallidermin resistance of L. monocytogenes is influenced by mprF, but this trait is associated with a compromise in invasiveness. SIGNIFICANCE AND IMPACT OF THE STUDY The presence of pathogenic microorganisms in the food processing environment can cause a significant problem, especially when these microorganisms are established as biofilms. This study shows that the inactivation of the mprF gene results in enhanced biofilm formation and abiotic surface attachment of L. monocytogenes.
Collapse
Affiliation(s)
- J Nowak
- The New Zealand Institute for Plant & Food Research Limited, Auckland, New Zealand.,Institute of Food, Nutrition and Human Health, Massey University, Palmerston North, New Zealand
| | - S B Visnovsky
- The New Zealand Institute for Plant & Food Research Limited, Lincoln, New Zealand
| | - C D Cruz
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - G C Fletcher
- The New Zealand Institute for Plant & Food Research Limited, Auckland, New Zealand
| | - A H M van Vliet
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - D Hedderley
- The New Zealand Institute for Plant & Food Research Limited, Palmerston North, New Zealand
| | - R Butler
- The New Zealand Institute for Plant & Food Research Limited, Lincoln, New Zealand
| | - S Flint
- Institute of Food, Nutrition and Human Health, Massey University, Palmerston North, New Zealand
| | - J Palmer
- Institute of Food, Nutrition and Human Health, Massey University, Palmerston North, New Zealand
| | - A R Pitman
- The Foundation for Arable Research, Christchurch, New Zealand
| |
Collapse
|
31
|
Kaptchouang Tchatchouang CD, Fri J, De Santi M, Brandi G, Schiavano GF, Amagliani G, Ateba CN. Listeriosis Outbreak in South Africa: A Comparative Analysis with Previously Reported Cases Worldwide. Microorganisms 2020; 8:E135. [PMID: 31963542 PMCID: PMC7023107 DOI: 10.3390/microorganisms8010135] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/26/2019] [Accepted: 12/27/2019] [Indexed: 01/01/2023] Open
Abstract
Listeria species are Gram-positive, rod-shaped, facultative anaerobic bacteria, which do not produce endospores. The genus, Listeria, currently comprises 17 characterised species of which only two (L. monocytogenes and L. ivanovii) are known to be pathogenic to humans. Food products and related processing environments are commonly contaminated with pathogenic species. Outbreaks and sporadic cases of human infections resulted in considerable economic loss. South Africa witnessed the world's largest listeriosis outbreak, characterised by a progressive increase in cases of the disease from January 2017 to July 2018. Of the 1060 laboratory-confirmed cases of listeriosis reported by the National Institute of Communicable Diseases (NICD), 216 deaths were recorded. Epidemiological investigations indicated that ready-to-eat processed meat products from a food production facility contaminated with L. monocytogenes was responsible for the outbreak. Multilocus sequence typing (MLST) revealed that a large proportion (91%) of the isolates from patients were sequence type 6 (ST6). Recent studies revealed a recurrent occurrence of small outbreaks of listeriosis with more severe side-effects in humans. This review provides a comparative analysis of a recently reported and most severe outbreak of listeriosis in South Africa, with those previously encountered in other countries worldwide. The review focuses on the transmission of the pathogen, clinical symptoms of the disease and its pathogenicity. The review also focuses on the major outbreaks of listeriosis reported in different parts of the world, sources of contamination, morbidity, and mortality rates as well as cost implications. Based on data generated during the outbreak of the disease in South Africa, listeriosis was added to the South African list of mandatory notifiable medical conditions. Surveillance systems were strengthened in the South African food chain in order to assist in preventing and facilitating early detection of both sporadic cases and outbreaks of infections caused by these pathogens in humans.
Collapse
Affiliation(s)
| | - Justine Fri
- Department of Microbiology, North-West University, Mafikeng Campus, Private Bag X2046, Mmabatho 2735, South Africa; (C.-D.K.T.); (J.F.)
| | - Mauro De Santi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, via S. Chiara 27, 61029 Urbino (PU), Italy; (M.D.S.); (G.B.); (G.A.)
- Department of Humanities, University of Urbino Carlo Bo, via Bramante 17, 61029 Urbino (PU), Italy;
| | - Giorgio Brandi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, via S. Chiara 27, 61029 Urbino (PU), Italy; (M.D.S.); (G.B.); (G.A.)
- Department of Humanities, University of Urbino Carlo Bo, via Bramante 17, 61029 Urbino (PU), Italy;
| | | | - Giulia Amagliani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, via S. Chiara 27, 61029 Urbino (PU), Italy; (M.D.S.); (G.B.); (G.A.)
| | - Collins Njie Ateba
- Department of Microbiology, North-West University, Mafikeng Campus, Private Bag X2046, Mmabatho 2735, South Africa; (C.-D.K.T.); (J.F.)
- Food Security and Safety Niche Area, Faculty of Natural and Agricultural Sciences, North-West University, Mafikeng Campus, Mmabatho, Mafikeng 2735, South Africa
| |
Collapse
|
32
|
Argov T, Sapir SR, Pasechnek A, Azulay G, Stadnyuk O, Rabinovich L, Sigal N, Borovok I, Herskovits AA. Coordination of cohabiting phage elements supports bacteria-phage cooperation. Nat Commun 2019; 10:5288. [PMID: 31754112 PMCID: PMC6872733 DOI: 10.1038/s41467-019-13296-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 10/30/2019] [Indexed: 01/08/2023] Open
Abstract
Bacterial pathogens often carry multiple prophages and other phage-derived elements within their genome, some of which can produce viral particles in response to stress. Listeria monocytogenes 10403S harbors two phage elements in its chromosome, both of which can trigger bacterial lysis under stress: an active prophage (ϕ10403S) that promotes the virulence of its host and can produce infective virions, and a locus encoding phage tail-like bacteriocins. Here, we show that the two phage elements are co-regulated, with the bacteriocin locus controlling the induction of the prophage and thus its activity as a virulence-associated molecular switch. More specifically, a metalloprotease encoded in the bacteriocin locus is upregulated in response to stress and acts as an anti-repressor for CI-like repressors encoded in each phage element. Our results provide molecular insight into the phenomenon of polylysogeny and its intricate adaptation to complex environments.
Collapse
Affiliation(s)
- Tal Argov
- The School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Shai Ran Sapir
- The School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Anna Pasechnek
- The School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Gil Azulay
- The School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Olga Stadnyuk
- The School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Lev Rabinovich
- The School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Nadejda Sigal
- The School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Ilya Borovok
- The School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel
| | - Anat A Herskovits
- The School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel.
| |
Collapse
|
33
|
Phan TK, Bindra GK, Williams SA, Poon IK, Hulett MD. Combating Human Pathogens and Cancer by Targeting Phosphoinositides and Their Metabolism. Trends Pharmacol Sci 2019; 40:866-882. [DOI: 10.1016/j.tips.2019.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022]
|
34
|
Coban A, Pennone V, Sudagidan M, Molva C, Jordan K, Aydin A. Prevalence, virulence characterization, and genetic relatedness of Listeria monocytogenes isolated from chicken retail points and poultry slaughterhouses in Turkey. Braz J Microbiol 2019; 50:1063-1073. [PMID: 31478167 PMCID: PMC6863211 DOI: 10.1007/s42770-019-00133-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/02/2019] [Indexed: 12/17/2022] Open
Abstract
Listeria monocytogenes is one of the most important foodborne pathogens and is a causal agent of listeriosis in humans and animals. The aim of this study was to determine the prevalence, serogroups, antibiotic susceptibility, virulence factor genes, and genetic relatedness of L. monocytogenes strains isolated from 500 poultry samples in Turkey. The isolation sources of 103 L. monocytogenes strains were retail markets (n = 100) and slaughterhouses (n = 3). L. monocytogenes strains were identified as serogroups 1/2a-3a (75.7%, lineage I), 1/2c-3c (14.56%, lineage I), 1/2b-3b-7 (5.82%, lineage II), 4a-4c (2.91%, lineage III), and 4b-4d-4e (0.97%, lineage III). Most of the L. monocytogenes strains (93.2%) were susceptible to the antibiotics tested. PCR analysis indicated that the majority of the strains (95% to 100%) contained most of the virulence genes (hylA, plcA, plcB, prfA, mpl, actA, dltA, fri, flaA inlA, inlC, and inlJ). Pulsed-field gel electrophoresis (PFGE) demonstrated that there were 18 pulsotypes grouped at a similarity of > 90% among the strains. These results indicate that it is necessary to prevent the presence of L. monocytogenes in the poultry-processing environments to help prevent outbreaks of listeriosis and protect public health.
Collapse
Affiliation(s)
- Aysen Coban
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
- Faculty of Veterinary Medicine, Department of Food Hygiene and Technology, İstanbul University-Cerrahpaşa, Avcilar, Istanbul, Turkey
| | - Vincenzo Pennone
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - Mert Sudagidan
- KIT-ARGEM R&D Center, Konya Food and Agriculture University, Meram, Konya, Turkey
| | - Celenk Molva
- Department of Food Engineering, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Kieran Jordan
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - Ali Aydin
- Faculty of Veterinary Medicine, Department of Food Hygiene and Technology, İstanbul University-Cerrahpaşa, Avcilar, Istanbul, Turkey.
| |
Collapse
|
35
|
Osonga F, Akgul A, Miller RM, Eshun GB, Yazgan I, Akgul A, Sadik OA. Antimicrobial Activity of a New Class of Phosphorylated and Modified Flavonoids. ACS OMEGA 2019; 4:12865-12871. [PMID: 31460413 PMCID: PMC6681995 DOI: 10.1021/acsomega.9b00077] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/03/2019] [Indexed: 06/01/2023]
Abstract
The surge of resistant food pathogens is a major threat worldwide. Previous research conducted on phytochemicals has shown their antibacterial activity against pathogenic bacteria. The design of antimicrobial agents to curb pathogenic disease remains a challenge demanding critical attention. Flavonoids such as apigenin and quercetin were evaluated against Gram-positive and Gram-negative bacteria. The results indicated that the antibacterial activity of each flavonoid occurred at a different minimum inhibitory concentration. However, the antimicrobial activity results of the modified flavonoids were also reported, and it was observed that the Gram-positive bacteria were more susceptible in comparison to the Gram-negative bacteria. The cell wall structure of the Gram-positive and Gram-negative bacteria could be the main reason for the bacteria susceptibility. Modified flavonoids could be used as a suitable alternative antimicrobial agent for the treatment of infectious diseases. Our results indicated 100% inhibition of Listeria monocytogenes, Pseudomonas aeruginosa, and Aeromonas hydrophila with modified flavonoids.
Collapse
Affiliation(s)
- Francis
J. Osonga
- Department
of Chemistry, Center for Research
in Advanced Sensing Technologies & Environmental Sustainability
(CREATES), State University of New York
at Binghamton, PO Box 6000, Binghamton, New York 13902, United States
| | - Ali Akgul
- Department of Basic
Sciences, College of Veterinary Medicine and Department of Sustainable
Bioproducts, College of Forest Resources, Mississippi State University, Starkville, Mississippi 39762, United States
| | - Roland M. Miller
- Department
of Chemistry, Center for Research
in Advanced Sensing Technologies & Environmental Sustainability
(CREATES), State University of New York
at Binghamton, PO Box 6000, Binghamton, New York 13902, United States
| | - Gaddi B. Eshun
- Department
of Chemistry, Center for Research
in Advanced Sensing Technologies & Environmental Sustainability
(CREATES), State University of New York
at Binghamton, PO Box 6000, Binghamton, New York 13902, United States
| | - Idris Yazgan
- Department
of Chemistry, Center for Research
in Advanced Sensing Technologies & Environmental Sustainability
(CREATES), State University of New York
at Binghamton, PO Box 6000, Binghamton, New York 13902, United States
| | - Ayfer Akgul
- Department of Basic
Sciences, College of Veterinary Medicine and Department of Sustainable
Bioproducts, College of Forest Resources, Mississippi State University, Starkville, Mississippi 39762, United States
| | - Omowunmi A. Sadik
- Department
of Chemistry, Center for Research
in Advanced Sensing Technologies & Environmental Sustainability
(CREATES), State University of New York
at Binghamton, PO Box 6000, Binghamton, New York 13902, United States
| |
Collapse
|
36
|
Wang Y, Guo W, Wu X, Zhang Y, Mannion C, Brouchkov A, Man YG, Chen T. Oncolytic Bacteria and their potential role in bacterium-mediated tumour therapy: a conceptual analysis. J Cancer 2019; 10:4442-4454. [PMID: 31528208 PMCID: PMC6746139 DOI: 10.7150/jca.35648] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022] Open
Abstract
As the human microbiota has been confirmed to be of great significance in maintaining health, the dominant bacteria in them have been applied as probiotics to treat various diseases. After the detection of bacteria in tumours, which had previously been considered a sterile region, these bacteria have been isolated and genetically modified for use in tumour therapy. In this review, we sum up the main types of bacteria used in tumour therapy and reveal the mechanisms of both wild type and engineered bacteria in eliminating tumour cells, providing potential possibilities for newly detected, genetically modified, tumour-associated bacteria in anti-tumour therapy.
Collapse
Affiliation(s)
- Yuqing Wang
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Wenxuan Guo
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - XiaoLi Wu
- JiangXi university of traditional Chinese medicine, College of basic medicine, Nanchang 330000, PR China
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Ciaran Mannion
- Hackensack University Medical Center, Hackensack, NJ, USA
| | - Anatoli Brouchkov
- Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
- Tyumen State University, Volodarskogo 6, Tyumen 625003, Russia
| | - Yan-Gao Man
- Department of Pathology, Hackensack Meridian Health-Hackensack University Medical Center, NJ, USA
| | - Tingtao Chen
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, PR China
| |
Collapse
|
37
|
|
38
|
Establishment of Listeria monocytogenes in the Gastrointestinal Tract. Microorganisms 2019; 7:microorganisms7030075. [PMID: 30857361 PMCID: PMC6463042 DOI: 10.3390/microorganisms7030075] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/05/2019] [Accepted: 03/05/2019] [Indexed: 12/15/2022] Open
Abstract
Listeria monocytogenes is a Gram positive foodborne pathogen that can colonize the gastrointestinal tract of a number of hosts, including humans. These environments contain numerous stressors such as bile, low oxygen and acidic pH, which may impact the level of colonization and persistence of this organism within the GI tract. The ability of L. monocytogenes to establish infections and colonize the gastrointestinal tract is directly related to its ability to overcome these stressors, which is mediated by the efficient expression of several stress response mechanisms during its passage. This review will focus upon how and when this occurs and how this impacts the outcome of foodborne disease.
Collapse
|
39
|
Chen C, Nguyen BN, Mitchell G, Margolis SR, Ma D, Portnoy DA. The Listeriolysin O PEST-like Sequence Co-opts AP-2-Mediated Endocytosis to Prevent Plasma Membrane Damage during Listeria Infection. Cell Host Microbe 2019; 23:786-795.e5. [PMID: 29902442 DOI: 10.1016/j.chom.2018.05.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/23/2018] [Accepted: 05/11/2018] [Indexed: 10/14/2022]
Abstract
Listeriolysin O (LLO) is a cholesterol-dependent cytolysin that mediates escape of Listeria monocytogenes from a phagosome, enabling growth of the bacteria in the host cell cytosol. LLO contains a PEST-like sequence that prevents it from killing infected cells, but the mechanism involved is unknown. We found that the LLO PEST-like sequence was necessary to mediate removal of LLO from the interior face of the plasma membrane, where it coalesces into discrete puncta. LLO interacts with Ap2a2, an adaptor protein involved in endocytosis, via its PEST-like sequence, and Ap2a2-dependent endocytosis is required to prevent LLO-induced cytotoxicity. An unrelated PEST-like sequence from a human G protein-coupled receptor (GPCR), which also interacts with Ap2a2, could functionally complement the PEST-like sequence in L. monocytogenes LLO. These data revealed that LLO co-opts the host endocytosis machinery to protect the integrity of the host plasma membrane during L. monocytogenes infection.
Collapse
Affiliation(s)
- Chen Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Brittney N Nguyen
- Graduate Group in Microbiology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gabriel Mitchell
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Shally R Margolis
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Darren Ma
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
40
|
Expression of the VP1 protein of FMDV integrated chromosomally with mutant Listeria monocytogenes strain induced both humoral and cellular immune responses. Appl Microbiol Biotechnol 2019; 103:1919-1929. [PMID: 30627793 DOI: 10.1007/s00253-018-09605-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/26/2018] [Accepted: 12/28/2018] [Indexed: 02/07/2023]
Abstract
Live vector-based vaccine is a modern approach to overcome the drawbacks of inactivated foot-and-mouth disease (FMD) vaccines such as improper inactivation during manufacture. Listeria monocytogenes (LM), an intracellular microorganism with immune-stimulatory properties, is appropriate to be utilized as a live bacterial vaccine vector. FMDV-VP1 protein has the capability to induce both cellular and humoral immune responses since it is considered the most immunogenic part of FMDV capsid and has the most of antigenic sites for viral neutralization. The codon-optimized vp1 gene was ligated to the integrative pCW702 plasmid to construct the target cassette. The antigen cassette was integrated successfully into the chromosome of mutant LM strain via homologous recombination for more stability to generate a candidate vaccine strain LM△actAplcB-vp1. Safety evaluation of recombinant LM△actAplcB-vp1 revealed it could be eliminated from the internal organs within 3 days as a safe candidate vaccine. Mice groups were immunized I.V. twice with the recombinant LM△actAplcB-vp1 at an interval of 2 weeks. Antigen-specific IgG antibodies and the level of CD4+- and CD8+-specific secreted cytokines were estimated to evaluate the immunogenicity of the candidate vaccine. The rapid onset immune response was detected, strong IgG humoral immune response within 14 days post immunization and augmented again after the booster dose. Cellular immunity data after 9 days post the prime dose indicated elevation in CD4+ and CD8+ secreted cytokine level with another elevation after the booster dose. This is the first report to explain the ability of attenuated mutant LM to be a promising live vector for FMDV vaccine.
Collapse
|
41
|
Esbelin J, Santos T, Ribière C, Desvaux M, Viala D, Chambon C, Hébraud M. Comparison of three methods for cell surface proteome extraction of Listeria monocytogenes biofilms. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 22:779-787. [PMID: 30457927 DOI: 10.1089/omi.2018.0144] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The cell surface proteome of the foodborne pathogen Listeria monocytogenes, the etiological agent of listeriosis, is critical for understanding the physiological processes associated with stress resistance and persistence in the environment. In this context, the most widespread mode of growth for bacterial cells in natural and industrial environments is in biofilms. Cell surface proteins are, however, challenging to characterize because of their low abundance and poor solubility. Moreover, cell surface protein extracts are usually contaminated with cytoplasmic proteins that constitute the main signal in proteomic analysis. This study aimed to compare the efficiency of three methods to extract and explore surface proteins of L. monocytogenes growing in a biofilm: trypsin shaving, biotinylation, and cell fractionation. Peptide separation and identification were performed by shotgun proteomics using high-performance liquid chromatography combined with tandem mass spectrometry (LC-MS/MS). The biotinylation method was the most effective in extracting surface proteins, with the lowest rate of contamination by cytoplasmic proteins. Although presenting a higher contamination rate in cytoplasmic proteins, the other two techniques allowed the identification of additional surface proteins. Seven proteins were commonly retrieved by the three methods. The extracted proteins belong to several functional classes, involved in virulence, transport, or metabolic pathways. Finally, the three extraction methods seemed complementary and their combined use improved the exploration of the bacterial surface proteome. These new findings collectively inform future discovery and translational proteomics for clinical, environmental health, and industrial applications.
Collapse
Affiliation(s)
- Julia Esbelin
- 1 Université Clermont Auvergne, INRA, UMR Microbiologie Environnement Digestif Santé (MEDiS), Saint-Genès Champanelle, France
| | - Tiago Santos
- 1 Université Clermont Auvergne, INRA, UMR Microbiologie Environnement Digestif Santé (MEDiS), Saint-Genès Champanelle, France
| | - Céline Ribière
- 1 Université Clermont Auvergne, INRA, UMR Microbiologie Environnement Digestif Santé (MEDiS), Saint-Genès Champanelle, France
| | - Mickaël Desvaux
- 1 Université Clermont Auvergne, INRA, UMR Microbiologie Environnement Digestif Santé (MEDiS), Saint-Genès Champanelle, France
| | - Didier Viala
- 2 INRA, Plate-Forme d'Exploration du Métabolisme composante protéomique (PFEMcp), Saint-Genès Champanelle, France
| | - Christophe Chambon
- 2 INRA, Plate-Forme d'Exploration du Métabolisme composante protéomique (PFEMcp), Saint-Genès Champanelle, France
| | - Michel Hébraud
- 1 Université Clermont Auvergne, INRA, UMR Microbiologie Environnement Digestif Santé (MEDiS), Saint-Genès Champanelle, France.,2 INRA, Plate-Forme d'Exploration du Métabolisme composante protéomique (PFEMcp), Saint-Genès Champanelle, France
| |
Collapse
|
42
|
Szappanos D, Tschismarov R, Perlot T, Westermayer S, Fischer K, Platanitis E, Kallinger F, Novatchkova M, Lassnig C, Müller M, Sexl V, Bennett KL, Foong-Sobis M, Penninger JM, Decker T. The RNA helicase DDX3X is an essential mediator of innate antimicrobial immunity. PLoS Pathog 2018; 14:e1007397. [PMID: 30475900 PMCID: PMC6283616 DOI: 10.1371/journal.ppat.1007397] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 12/06/2018] [Accepted: 10/11/2018] [Indexed: 12/30/2022] Open
Abstract
DExD/H box RNA helicases, such as the RIG-I-like receptors (RLR), are important components of the innate immune system. Here we demonstrate a pivotal and sex-specific role for the heterosomal isoforms of the DEAD box RNA helicase DDX3 in the immune system. Mice lacking DDX3X during hematopoiesis showed an altered leukocyte composition in bone marrow and spleen and a striking inability to combat infection with Listeria monocytogenes. Alterations in innate immune responses resulted from decreased effector cell availability and function as well as a sex-dependent impairment of cytokine synthesis. Thus, our data provide further in vivo evidence for an essential contribution of a non-RLR DExD/H RNA helicase to innate immunity and suggest it may contribute to sex-related differences in resistance to microbes and resilience to inflammatory disease.
Collapse
Affiliation(s)
- Daniel Szappanos
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Roland Tschismarov
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Thomas Perlot
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter, Vienna, Austria
| | - Sandra Westermayer
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Katrin Fischer
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Ekaterini Platanitis
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Fabian Kallinger
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Maria Novatchkova
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter, Vienna, Austria
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Caroline Lassnig
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Keiryn L. Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Michelle Foong-Sobis
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter, Vienna, Austria
| | - Josef M. Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter, Vienna, Austria
| | - Thomas Decker
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna Biocenter, Vienna, Austria
| |
Collapse
|
43
|
Li J, Zhang Y, Zhang Y, Mao F, Xiao S, Xiang Z, Ma H, Yu Z. A Lysin motif (LysM)-containing protein from Hong Kong oyster, Crassostrea hongkongensis functions as a pattern recognition protein and an antibacterial agent. Gene 2018; 674:134-142. [DOI: 10.1016/j.gene.2018.06.091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 06/11/2018] [Accepted: 06/26/2018] [Indexed: 12/26/2022]
|
44
|
King MT, Huh I, Shenai A, Brooks TM, Brooks CL. Structural basis of V HH-mediated neutralization of the food-borne pathogen Listeria monocytogenes. J Biol Chem 2018; 293:13626-13635. [PMID: 29976754 DOI: 10.1074/jbc.ra118.003888] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/01/2018] [Indexed: 12/12/2022] Open
Abstract
Listeria monocytogenes causes listeriosis, a potentially fatal food-borne disease. The condition is especially harmful to pregnant women. Listeria outbreaks can originate from diverse foods, highlighting the need for novel strategies to improve food safety. The first step in Listeria invasion is internalization of the bacteria, which is mediated by the interaction of the internalin family of virulence factors with host cell receptors. A crucial interaction for Listeria invasion of the placenta, and thus a target for therapeutic intervention, is between internalin B (InlB) and the receptor c-Met. Single-domain antibodies (VHH, also called nanobodies, or sdAbs) from camel heavy-chain antibodies are a novel solution for preventing Listeria infections. The VHH R303, R330, and R326 all bind InlB with high affinity; however, the molecular mechanism behind their mode of action was unknown. We demonstrate that despite a high degree of sequence and structural diversity, the VHH bind a single epitope on InlB. A combination of gentamicin protection assays and florescent microscopy establish that InlB-specific VHH inhibit Listeria invasion of HeLa cells. A high-resolution X-ray structure of VHH R303 in complex with InlB showed that the VHH binds at the c-Met interaction site on InlB, thereby acting as a competitive inhibitor preventing bacterial invasion. These results point to the potential of VHH as a novel class of therapeutics for the prevention of listeriosis.
Collapse
Affiliation(s)
- Moeko Toride King
- From the Department of Chemistry, California State University, Fresno, California 93740
| | - Ian Huh
- From the Department of Chemistry, California State University, Fresno, California 93740
| | - Akhilesh Shenai
- From the Department of Chemistry, California State University, Fresno, California 93740
| | - Teresa M Brooks
- From the Department of Chemistry, California State University, Fresno, California 93740
| | - Cory L Brooks
- From the Department of Chemistry, California State University, Fresno, California 93740
| |
Collapse
|
45
|
Vertical Transmission of Listeria monocytogenes: Probing the Balance between Protection from Pathogens and Fetal Tolerance. Pathogens 2018; 7:pathogens7020052. [PMID: 29799503 PMCID: PMC6027155 DOI: 10.3390/pathogens7020052] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/16/2018] [Accepted: 05/22/2018] [Indexed: 12/19/2022] Open
Abstract
Protection of the developing fetus from pathogens is one of the many critical roles of the placenta. Listeria monocytogenes is one of a select number of pathogens that can cross the placental barrier and cause significant harm to the fetus, leading to spontaneous abortion, stillbirth, preterm labor, and disseminated neonate infection despite antibiotic treatment. Such severe outcomes serve to highlight the importance of understanding how L. monocytogenes mediates infiltration of the placental barrier. Here, we review what is currently known regarding vertical transmission of L. monocytogenes as a result of cell culture and animal models of infection. In vitro cell culture and organ models have been useful for the identification of L. monocytogenes virulence factors that contribute to placental invasion. Examples include members of the Internalin family of bacterial surface proteins such as Interalin (Inl)A, InlB, and InlP that promote invasion of cells at the maternal-fetal interface. A number of animal models have been used to interrogate L. monocytogenes vertical transmission, including mice, guinea pigs, gerbils, and non-human primates; each of these models has advantages while still not providing a comprehensive understanding of L. monocytogenes invasion of the human placenta and/or fetus. These models do, however, allow for the molecular investigation of the balance between fetal tolerance and immune protection from L. monocytogenes during pregnancy.
Collapse
|
46
|
Bastounis EE, Yeh YT, Theriot JA. Matrix stiffness modulates infection of endothelial cells by Listeria monocytogenes via expression of cell surface vimentin. Mol Biol Cell 2018; 29:1571-1589. [PMID: 29718765 PMCID: PMC6080647 DOI: 10.1091/mbc.e18-04-0228] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Extracellular matrix stiffness (ECM) is one of the many mechanical forces acting on mammalian adherent cells and an important determinant of cellular function. While the effect of ECM stiffness on many aspects of cellular behavior has been studied previously, how ECM stiffness might mediate susceptibility of host cells to infection by bacterial pathogens is hitherto unexplored. To address this open question, we manufactured hydrogels of varying physiologically relevant stiffness and seeded human microvascular endothelial cells (HMEC-1) on them. We then infected HMEC-1 with the bacterial pathogen Listeria monocytogenes (Lm) and found that adhesion of Lm to host cells increases monotonically with increasing matrix stiffness, an effect that requires the activity of focal adhesion kinase (FAK). We identified cell surface vimentin as a candidate surface receptor mediating stiffness-dependent adhesion of Lm to HMEC-1 and found that bacterial infection of these host cells is decreased when the amount of surface vimentin is reduced. Our results provide the first evidence that ECM stiffness can mediate the susceptibility of mammalian host cells to infection by a bacterial pathogen.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305
| | - Yi-Ting Yeh
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Julie A Theriot
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
47
|
Pietrocola G, Arciola CR, Rindi S, Montanaro L, Speziale P. Streptococcus agalactiae Non-Pilus, Cell Wall-Anchored Proteins: Involvement in Colonization and Pathogenesis and Potential as Vaccine Candidates. Front Immunol 2018; 9:602. [PMID: 29686667 PMCID: PMC5900788 DOI: 10.3389/fimmu.2018.00602] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/09/2018] [Indexed: 11/13/2022] Open
Abstract
Group B Streptococcus (GBS) remains an important etiological agent of several infectious diseases including neonatal septicemia, pneumonia, meningitis, and orthopedic device infections. This pathogenicity is due to a variety of virulence factors expressed by Streptococcus agalactiae. Single virulence factors are not sufficient to provoke a streptococcal infection, which is instead promoted by the coordinated activity of several pathogenicity factors. Such determinants, mostly cell wall-associated and secreted proteins, include adhesins that mediate binding of the pathogen to host extracellular matrix/plasma ligands and cell surfaces, proteins that cooperate in the invasion of and survival within host cells and factors that neutralize phagocytosis and/or modulate the immune response. The genome-based approaches and bioinformatics tools and the extensive use of biophysical and biochemical methods and animal model studies have provided a great wealth of information on the molecular structure and function of these virulence factors. In fact, a number of new GBS surface-exposed or secreted proteins have been identified (GBS immunogenic bacterial adhesion protein, leucine-rich repeat of GBS, serine-rich repeat proteins), the three-dimensional structures of known streptococcal proteins (αC protein, C5a peptidase) have been solved and an understanding of the pathogenetic role of "old" and new determinants has been better defined in recent years. Herein, we provide an update of our current understanding of the major surface cell wall-anchored proteins from GBS, with emphasis on their biochemical and structural properties and the pathogenetic roles they may have in the onset and progression of host infection. We also focus on the antigenic profile of these compounds and discuss them as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Giampiero Pietrocola
- Unit of Biochemistry, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Carla Renata Arciola
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy.,Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Simonetta Rindi
- Unit of Biochemistry, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Lucio Montanaro
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy.,Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Pietro Speziale
- Unit of Biochemistry, Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy
| |
Collapse
|
48
|
Bacteriocinogenic Enterococcus faecium inhibits the virulence property of Listeria monocytogenes. Lebensm Wiss Technol 2018. [DOI: 10.1016/j.lwt.2017.10.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
49
|
|
50
|
Prevalence and methodologies for detection, characterization and subtyping of Listeria monocytogenes and L. ivanovii in foods and environmental sources. FOOD SCIENCE AND HUMAN WELLNESS 2017. [DOI: 10.1016/j.fshw.2017.06.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|