1
|
Lata K, Nandy K, Geetika, Chattopadhyay K. Mechanistic Cooperation of the Two Pore-Forming Transmembrane Motifs Regulates the β-Barrel Pore Formation by Listeriolysin O. Biochemistry 2025; 64:917-927. [PMID: 39869763 DOI: 10.1021/acs.biochem.4c00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Listeriolysin O (LLO) is a potent membrane-damaging pore-forming toxin (PFT) secreted by the bacterial pathogen Listeria monocytogenes. LLO belongs to the family of cholesterol-dependent cytolysins (CDCs), which specifically target cholesterol-containing cell membranes to form oligomeric pores and induce membrane damage. CDCs, including LLO, harbor designated pore-forming motifs. In the soluble monomeric state, these motifs are present as helical segments (two transmembrane helices (TMHs); TMH1 and TMH2), and in the course of oligomeric pore formation, they convert into transmembrane β-hairpins to form the β-barrel scaffold of the CDC pores. Despite their well-established role in forming the β-barrel pore scaffold, precise structural implications of the two distinct TMH motifs and their membrane-insertion mechanism still remain obscure. Here, we show that the two TMH motifs of LLO contribute differently to maintaining the structural integrity of the toxin. While the deletion of TMH1 imposed a more serious defect, truncation of TMH2 was found to have a less severe effect on the structural integrity. Despite showing membrane-binding and oligomerization ability, the TMH2-deleted LLO variant displayed drastically abrogated pore-forming activity, presumably due to compromised membrane-insertion efficacy of the pore-forming TMH motifs. When probed for the membrane-insertion mechanism, we found slower membrane-insertion kinetics for TMH2 than for TMH1. Interestingly, deletion of TMH2 arrested membrane insertion of TMH1, thus suggesting a stringent cooperation between the two TMH motifs in regulating the pore-formation mechanism of LLO. Taken together, our study provides new mechanistic insights regarding the membrane-damaging action of LLO, in the CDC family of PFTs.
Collapse
Affiliation(s)
- Kusum Lata
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab 140306, India
| | - Koyel Nandy
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab 140306, India
| | - Geetika
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab 140306, India
| | - Kausik Chattopadhyay
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab 140306, India
| |
Collapse
|
2
|
Neupane R, Malla S, Karthikeyan C, Asbhy CR, Boddu SHS, Jayachandra Babu R, Tiwari AK. Endocytic highways: Navigating macropinocytosis and other endocytic routes for precision drug delivery. Int J Pharm 2025; 673:125356. [PMID: 39956408 DOI: 10.1016/j.ijpharm.2025.125356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/22/2024] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Drug molecules can reach intracellular targets by different mechanisms, such as passive diffusion, active transport, and endocytosis. Endocytosis is the process by which cells engulf extracellular material by forming a vesicle and transporting it into the cells. In addition to its biological functions, endocytosis plays a vital role in the internalization of the therapeutic molecules. Clathrin-mediated endocytosis, caveolar endocytosis, and macropinocytosis are the most researched routes in the field of drug delivery. In addition to conventional small therapeutic molecules, the use of nanoformulations and large molecules, such as nucleic acids, peptides, and antibodies, have broadened the field of drug delivery. Although the majority of small therapeutic molecules can enter cells via passive diffusion, large molecules, and advanced targeted delivery systems, such as nanoparticles, are internalized by the endocytic route. Therefore, it is imperative to understand the characteristics of the endocytic routes in greater detail to design therapeutic molecules or formulations for successful delivery to the intracellular targets. This review highlights the prospects and limitations of the major endocytic routes for drug delivery, with a major emphasis on macropinocytosis. Since macropinocytosis is a non-selective uptake of extracellular matrix, the selective induction of macropinocytosis, using compounds that induce macropinocytosis and modulate macropinosome trafficking pathways, could be a potential approach for the intracellular delivery of diverse therapeutic modalities. Furthermore, we have summarized the characteristics associated with the formulations or drug carriers that can affect the endocytic routes for cellular internalization. The techniques that are used to study the intracellular uptake processes of therapeutic molecules are briefly discussed. Finally, the major limitations for intracellular targeting, endo-lysosomal degradation, and different approaches that have been used in overcoming these limitations, are highlighted in this review.
Collapse
Affiliation(s)
- Rabin Neupane
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA
| | - Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA
| | - Chandrabose Karthikeyan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak 484887, India
| | - Charles R Asbhy
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John's University, Queens, NY 10049, USA
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - R Jayachandra Babu
- Department of Drug Discovery and Development, Auburn University, AL 36849, USA
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
3
|
Khlebnikova A, Kirshina A, Zakharova N, Ivanov R, Reshetnikov V. Current Progress in the Development of mRNA Vaccines Against Bacterial Infections. Int J Mol Sci 2024; 25:13139. [PMID: 39684849 DOI: 10.3390/ijms252313139] [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: 11/11/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Bacterial infections have accompanied humanity for centuries. The discovery of the first antibiotics and the subsequent golden era of their discovery temporarily shifted the balance in this confrontation to the side of humans. Nevertheless, the excessive and improper use of antibacterial drugs and the evolution of bacteria has gotten the better of humans again. Therefore, today, the search for new antibacterial drugs or the development of alternative approaches to the prevention and treatment of bacterial infections is relevant and topical again. Vaccination is one of the most effective strategies for the prevention of bacterial infections. The success of new-generation vaccines, such as mRNA vaccines, in the fight against viral infections has prompted many researchers to design mRNA vaccines against bacterial infections. Nevertheless, the biology of bacteria and their interactions with the host's immunity are much more complex compared to viruses. In this review, we discuss structural features and key mechanisms of evasion of an immune response for nine species of bacterial pathogens against which mRNA vaccines have been developed and tested in animals. We focus on the results of experiments involving the application of mRNA vaccines against various bacterial pathogens in animal models and discuss possible options for improving the vaccines' effectiveness. This is one of the first comprehensive reviews of the use of mRNA vaccines against bacterial infections in vivo to improve our knowledge.
Collapse
Affiliation(s)
- Alina Khlebnikova
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Anna Kirshina
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Natalia Zakharova
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Roman Ivanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Vasiliy Reshetnikov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| |
Collapse
|
4
|
Holmes CL, Albin OR, Mobley HLT, Bachman MA. Bloodstream infections: mechanisms of pathogenesis and opportunities for intervention. Nat Rev Microbiol 2024:10.1038/s41579-024-01105-2. [PMID: 39420097 DOI: 10.1038/s41579-024-01105-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 10/19/2024]
Abstract
Bloodstream infections (BSIs) are common in hospitals, often life-threatening and increasing in prevalence. Microorganisms in the blood are usually rapidly cleared by the immune system and filtering organs but, in some cases, they can cause an acute infection and trigger sepsis, a systemic response to infection that leads to circulatory collapse, multiorgan dysfunction and death. Most BSIs are caused by bacteria, although fungi also contribute to a substantial portion of cases. Escherichia coli, Staphylococcus aureus, coagulase-negative Staphylococcus, Klebsiella pneumoniae and Candida albicans are leading causes of BSIs, although their prevalence depends on patient demographics and geographical region. Each species is equipped with unique factors that aid in the colonization of initial sites and dissemination and survival in the blood, and these factors represent potential opportunities for interventions. As many pathogens become increasingly resistant to antimicrobials, new approaches to diagnose and treat BSIs at all stages of infection are urgently needed. In this Review, we explore the prevalence of major BSI pathogens, prominent mechanisms of BSI pathogenesis, opportunities for prevention and diagnosis, and treatment options.
Collapse
Affiliation(s)
- Caitlyn L Holmes
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Owen R Albin
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Harry L T Mobley
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael A Bachman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
5
|
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
|
6
|
Gul A, Pewe LL, Willems P, Mayer R, Thery F, Asselman C, Aernout I, Verbeke R, Eggermont D, Van Moortel L, Upton E, Zhang Y, Boucher K, Miret-Casals L, Demol H, De Smedt SC, Lentacker I, Radoshevich L, Harty JT, Impens F. Immunopeptidomics Mapping of Listeria monocytogenes T Cell Epitopes in Mice. Mol Cell Proteomics 2024; 23:100829. [PMID: 39147027 PMCID: PMC11414675 DOI: 10.1016/j.mcpro.2024.100829] [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: 02/09/2024] [Revised: 07/21/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024] Open
Abstract
Listeria monocytogenes is a foodborne intracellular bacterial model pathogen. Protective immunity against Listeria depends on an effective CD8+ T cell response, but very few T cell epitopes are known in mice as a common animal infection model for listeriosis. To identify epitopes, we screened for Listeria immunopeptides presented in the spleen of infected mice by mass spectrometry-based immunopeptidomics. We mapped more than 6000 mouse self-peptides presented on MHC class I molecules, including 12 high confident Listeria peptides from 12 different bacterial proteins. Bacterial immunopeptides with confirmed fragmentation spectra were further tested for their potential to activate CD8+ T cells, revealing VTYNYINI from the putative cell wall surface anchor family protein LMON_0576 as a novel bona fide peptide epitope. The epitope showed high biological potency in a prime boost model and can be used as a research tool to probe CD8+ T cell responses in the mouse models of Listeria infection. Together, our results demonstrate the power of immunopeptidomics for bacterial antigen identification.
Collapse
Affiliation(s)
- Adillah Gul
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lecia L Pewe
- Department of Pathology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA
| | - Patrick Willems
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Center for Plant Systems Biology, VIB, Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Rupert Mayer
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Fabien Thery
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Caroline Asselman
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Ilke Aernout
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Rein Verbeke
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Denzel Eggermont
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Laura Van Moortel
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Ellen Upton
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Yifeng Zhang
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA
| | - Katie Boucher
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Laia Miret-Casals
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Hans Demol
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA; Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA.
| | - John T Harty
- Department of Pathology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium.
| |
Collapse
|
7
|
Popoff MR, Ladant D. Toxins, Pathogenicity, Anti-Toxins, a Bicentennial Contribution. Toxins (Basel) 2024; 16:97. [PMID: 38393174 PMCID: PMC10891685 DOI: 10.3390/toxins16020097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
The bicentenary of Louis Pasteur's birth raises the opportunity to revisit the activity and influence of L [...].
Collapse
Affiliation(s)
- Michel R. Popoff
- Unité des Toxines Bactériennes, Institut Pasteur, Université Paris Cité, CNRS UMR 2001 INSERM U1306, F-75015 Paris, France
| | - Daniel Ladant
- Unité de Biochimie des Interactions Macromoléculaires, Institut Pasteur, Université Paris Cité, CNRS UMR 3528, F-75015 Paris, France;
| |
Collapse
|
8
|
Cong Z, Xiong Y, Lyu L, Fu B, Guo D, Sha Z, Yang B, Wu H. The relationship between Listeria infections and host immune responses: Listeriolysin O as a potential target. Biomed Pharmacother 2024; 171:116129. [PMID: 38194738 DOI: 10.1016/j.biopha.2024.116129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
Listeria monocytogenes (Lm), a foodborne bacterium, can infect people and has a high fatality rate in immunocompromised individuals. Listeriolysin O (LLO), the primary virulence factor of Lm, is critical in regulating the pathogenicity of Lm. This review concludes that LLO may either directly or indirectly activate a number of host cell viral pathophysiology processes, such as apoptosis, pyroptosis, autophagy, necrosis and necroptosis. We describe the invasion of host cells by Lm and the subsequent removal of Lm by CD8 T cells and CD4 T cells upon receipt of the LLO epitopes from major histocompatibility complex class I (MHC-I) and major histocompatibility complex class II (MHC-II). The development of several LLO-based vaccines that make use of the pore-forming capabilities of LLO and the immune response of the host cells is then described. Finally, we conclude by outlining the several natural substances that have been shown to alter the three-dimensional conformation of LLO by binding to particular amino acid residues of LLO, which reduces LLO pathogenicity and may be a possible pharmacological treatment for Lm.
Collapse
Affiliation(s)
- Zixuan Cong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Lyu Lyu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Beibei Fu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Zhou Sha
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Bo Yang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
9
|
Meireles D, Pombinho R, Cabanes D. Signals behind Listeria monocytogenes virulence mechanisms. Gut Microbes 2024; 16:2369564. [PMID: 38979800 PMCID: PMC11236296 DOI: 10.1080/19490976.2024.2369564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/13/2024] [Indexed: 07/10/2024] Open
Abstract
The tight and coordinated regulation of virulence gene expression is crucial to ensure the survival and persistence of bacterial pathogens in different contexts within their hosts. Considering this, bacteria do not express virulence factors homogenously in time and space, either due to their associated fitness cost or to their detrimental effect at specific infection stages. To efficiently infect and persist into their hosts, bacteria have thus to monitor environmental cues or chemical cell-to-cell signaling mechanisms that allow their transition from the external environment to the host, and therefore adjust gene expression levels, intrinsic biological activities, and appropriate behaviors. Listeria monocytogenes (Lm), a major Gram-positive facultative intracellular pathogen, stands out for its adaptability and capacity to thrive in a wide range of environments. Because of that, Lm presents itself as a significant concern in food safety and public health, that can lead to potentially life-threatening infections in humans. A deeper understanding of the intricate bacterial virulence mechanisms and the signals that control them provide valuable insights into the dynamic interplay between Lm and the host. Therefore, this review addresses the role of some crucial signals behind Lm pathogenic virulence mechanisms and explores how the ability to assimilate and interpret these signals is fundamental for pathogenesis, identifying potential targets for innovative antimicrobial strategies.
Collapse
Affiliation(s)
- Diana Meireles
- Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Group of Molecular Microbiology, IBMC, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar – ICBAS, Porto, Portugal
| | - Rita Pombinho
- Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Group of Molecular Microbiology, IBMC, Porto, Portugal
| | - Didier Cabanes
- Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Group of Molecular Microbiology, IBMC, Porto, Portugal
| |
Collapse
|
10
|
Magri Z, Poltorak A. You had me at PELO: a "Ribosome Rescuer" induces NLR inflammasome assembly. Cell Mol Immunol 2024; 21:1-2. [PMID: 37369783 PMCID: PMC10757713 DOI: 10.1038/s41423-023-01059-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Affiliation(s)
- Zoie Magri
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Alexander Poltorak
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, 136 Harrison Avenue, Boston, MA, 02111, USA.
- Department of Immunology, Tufts University School of Medicine, Boston, MA, 02111, USA.
| |
Collapse
|
11
|
Lee UH, Park SJ, Ju SA, Lee SC, Kim BS, Ahn B, Yi J, Park J, Won YW, Han IS, Lee BJ, Cho WJ, Park JW. DRG2 in macrophages is crucial for initial inflammatory response and protection against Listeria monocytogenes infection. Clin Immunol 2023; 257:109819. [PMID: 37918467 DOI: 10.1016/j.clim.2023.109819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023]
Abstract
Innate immune response is critical for the control of Listeria monocytogenes infection. Here, we identified developmentally regulated GTP-binding protein 2 (DRG2) in macrophages as a major regulator of the innate immune response against L. monocytogenes infection. Both whole-body DRG2 knockout (KO) mice and macrophage-specific DRG2 KO mice had low levels of IL-6 during early infection and increased susceptibility to L. monocytogenes infection. Following an initial impaired inflammatory response of macrophages upon i.p. L. monocytogenes infection, DRG2-/- mice showed delayed recruitment of neutrophils and monocytes into the peritoneal cavity, which led to elevated bacterial burden, inflammatory cytokine production at a late infection time point, and liver micro-abscesses. DRG2 deficiency decreased the transcriptional activity of NF-κB and impaired the inflammatory response of both bone marrow-derived and peritoneal macrophages upon L. monocytogenes stimulation. Our findings reveal that DRG2 in macrophages is critical for the initial inflammatory response and protection against L. monocytogenes infection.
Collapse
Affiliation(s)
- Unn Hwa Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Sang Jin Park
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Seong A Ju
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Sang Chul Lee
- CRONEX Co., Ltd., Hwaseong-si, Gyeonggi-do 18333, Republic of Korea
| | - Byung Sam Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Byungyong Ahn
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 44610, Republic of Korea; RopheLBio, B102, Seoul Forest M Tower, Seoul 04778, Republic of Korea
| | - Jawoon Yi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Young-Wook Won
- Department of Biomedical Engineering, University of North Texas, TX 76203-5017, USA; RopheLBio, B102, Seoul Forest M Tower, Seoul 04778, Republic of Korea
| | - In Seob Han
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea; Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Wha Ja Cho
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea; Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan 44610, Republic of Korea.
| |
Collapse
|
12
|
Wiśniewski P, Chajęcka-Wierzchowska W, Zadernowska A. High-Pressure Processing-Impacts on the Virulence and Antibiotic Resistance of Listeria monocytogenes Isolated from Food and Food Processing Environments. Foods 2023; 12:3899. [PMID: 37959018 PMCID: PMC10650155 DOI: 10.3390/foods12213899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
High-pressure processing (HPP) is one of the non-thermal methods of food preservation considered to be safe but may cause an increase/decrease in virulence potential and antibiotic resistance. The aim of the present study was to evaluate the survival of L. monocytogenes isolates after high-pressure processing (200 and 400 MPa for 5 min) and to determine changes in phenotypic and genotypic antibiotic resistance and virulence after this treatment. The 400 MPa treatment was shown to be effective in reducing pathogens to safe levels; however, the potential for cell recovery during storage was observed. In addition, studies on changes in virulence indicated possibilities related to a decrease in actA gene expression, overexpression of the hly and osfX gene, and an increase in biofilm-forming ability. The studies on changes in antibiotic resistance of isolates showed that all isolates showing initial susceptibility to lincomycin, fosfomycin, trimethoprim/sulfamethoxazole, and tetracycline became resistant to these antibiotics, which was associated with an increase in the values of minimum inhibitory concentrations. An increase in the expression of antibiotic resistance genes (mainly tetA_1, tetA_3, tetC) was also observed (mainly after the application of 200 MPa pressure), which was isolate dependent. However, it is noteworthy that the induced changes were permanent, i.e., they persisted even after the restoration of optimal environmental conditions. The results presented in our work indicate that the stress occurring during HPP can affect both phenotypic and genotypic changes in the virulence and antibiotic resistance potential of pathogens isolated from food and food processing environments. The potential associated with cell recovery and persistence of changes may influence the spread of virulent isolates of pathogens with increased antibiotic resistance in the food and food processing environment.
Collapse
Affiliation(s)
- Patryk Wiśniewski
- Department of Food Microbiology, Meat Technology and Chemistry, Faculty of Food Science, University of Warmia and Mazury, Plac Cieszyński 1, 10-726 Olsztyn, Poland; (W.C.-W.); (A.Z.)
| | | | | |
Collapse
|
13
|
Petrišič N, Adamek M, Kežar A, Hočevar SB, Žagar E, Anderluh G, Podobnik M. Structural basis for the unique molecular properties of broad-range phospholipase C from Listeria monocytogenes. Nat Commun 2023; 14:6474. [PMID: 37838694 PMCID: PMC10576769 DOI: 10.1038/s41467-023-42134-4] [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: 03/13/2023] [Accepted: 09/28/2023] [Indexed: 10/16/2023] Open
Abstract
Listeriosis is one of the most serious foodborne diseases caused by the intracellular bacterium Listeria monocytogenes. Its two major virulence factors, broad-range phospholipase C (LmPC-PLC) and the pore-forming toxin listeriolysin O (LLO), enable the bacterium to spread in the host by destroying cell membranes. Here, we determine the crystal structure of LmPC-PLC and complement it with the functional analysis of this enzyme. This reveals that LmPC-PLC has evolved several structural features to regulate its activity, including the invariant position of the N-terminal tryptophan (W1), the structurally plastic active site, Zn2+-dependent activity, and the tendency to form oligomers with impaired enzymatic activity. We demonstrate that the enzymatic activity of LmPC-PLC can be specifically inhibited by its propeptide added in trans. Furthermore, we show that the phospholipase activity of LmPC-PLC facilitates the pore-forming activity of LLO and affects the morphology of LLO oligomerization on lipid membranes, revealing the multifaceted synergy of the two virulence factors.
Collapse
Affiliation(s)
- Nejc Petrišič
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
- PhD Program 'Biosciences', Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Maksimiljan Adamek
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Andreja Kežar
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Samo B Hočevar
- Department of Analytical Chemistry, National Institute of Chemistry, Ljubljana, Slovenia
| | - Ema Žagar
- Department of Polymer Chemistry and Technology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia.
| |
Collapse
|
14
|
Ding YD, Shu LZ, He RS, Chen KY, Deng YJ, Zhou ZB, Xiong Y, Deng H. Listeria monocytogenes: a promising vector for tumor immunotherapy. Front Immunol 2023; 14:1278011. [PMID: 37868979 PMCID: PMC10587691 DOI: 10.3389/fimmu.2023.1278011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Cancer receives enduring international attention due to its extremely high morbidity and mortality. Immunotherapy, which is generally expected to overcome the limits of traditional treatments, serves as a promising direction for patients with recurrent or metastatic malignancies. Bacteria-based vectors such as Listeria monocytogenes take advantage of their unique characteristics, including preferential infection of host antigen presenting cells, intracellular growth within immune cells, and intercellular dissemination, to further improve the efficacy and minimize off-target effects of tailed immune treatments. Listeria monocytogenes can reshape the tumor microenvironment to bolster the anti-tumor effects both through the enhancement of T cells activity and a decrease in the frequency and population of immunosuppressive cells. Modified Listeria monocytogenes has been employed as a tool to elicit immune responses against different tumor cells. Currently, Listeria monocytogenes vaccine alone is insufficient to treat all patients effectively, which can be addressed if combined with other treatments, such as immune checkpoint inhibitors, reactivated adoptive cell therapy, and radiotherapy. This review summarizes the recent advances in the molecular mechanisms underlying the involvement of Listeria monocytogenes vaccine in anti-tumor immunity, and discusses the most concerned issues for future research.
Collapse
Affiliation(s)
- Yi-Dan Ding
- Medical College, Nanchang University, Nanchang, China
| | - Lin-Zhen Shu
- Medical College, Nanchang University, Nanchang, China
| | - Rui-Shan He
- Medical College, Nanchang University, Nanchang, China
| | - Kai-Yun Chen
- Office of Clinical Trials Administration, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan-Juan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
| | - Zhi-Bin Zhou
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
| | - Ying Xiong
- Department of General Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Moran J, Feltham L, Bagnall J, Goldrick M, Lord E, Nettleton C, Spiller DG, Roberts I, Paszek P. Live-cell imaging reveals single-cell and population-level infection strategies of Listeria monocytogenes in macrophages. Front Immunol 2023; 14:1235675. [PMID: 37675103 PMCID: PMC10478088 DOI: 10.3389/fimmu.2023.1235675] [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: 06/06/2023] [Accepted: 08/01/2023] [Indexed: 09/08/2023] Open
Abstract
Pathogens have developed intricate strategies to overcome the host's innate immune responses. In this paper we use live-cell microscopy with a single bacterium resolution to follow in real time interactions between the food-borne pathogen L. monocytogenes and host macrophages, a key event controlling the infection in vivo. We demonstrate that infection results in heterogeneous outcomes, with only a subset of bacteria able to establish a replicative invasion of macrophages. The fate of individual bacteria in the same host cell was independent from the host cell and non-cooperative, being independent from co-infecting bacteria. A higher multiplicity of infection resulted in a reduced probability of replication of the overall bacterial population. By use of internalisation assays and conditional probabilities to mathematically describe the two-stage invasion process, we demonstrate that the higher MOI compromises the ability of macrophages to phagocytose bacteria. We found that the rate of phagocytosis is mediated via the secreted Listeriolysin toxin (LLO), while the probability of replication of intracellular bacteria remained constant. Using strains expressing fluorescent reporters to follow transcription of either the LLO-encoding hly or actA genes, we show that replicative bacteria exhibited higher PrfA regulon expression in comparison to those bacteria that did not replicate, however elevated PrfA expression per se was not sufficient to increase the probability of replication. Overall, this demonstrates a new role for the population-level, but not single cell, PrfA-mediated activity to regulate outcomes of host pathogen interactions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ian Roberts
- School of Biology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Pawel Paszek
- School of Biology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
16
|
Wanford JJ, Odendall C. Ca 2+-calmodulin signalling at the host-pathogen interface. Curr Opin Microbiol 2023; 72:102267. [PMID: 36716574 DOI: 10.1016/j.mib.2023.102267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/19/2022] [Accepted: 01/04/2023] [Indexed: 01/29/2023]
Abstract
Multiple eukaryotic cell processes are modulated by calcium ions (Ca2+). As such, Ca2+ is emerging as a crucial regulator of innate immunity in multicellular organisms. In particular, recent studies have identified roles of Ca2+ signalling at the host-bacteria interface. Following microbial exposure, Ca2+ signals mobilised from the extracellular milieu or intracellular stores are transduced into cell physiological responses. However, during infection with host-adapted pathogens, Ca2+ signals are often atypical, due to the activities of virulence factors, with varied consequences for both the pathogen and the host cell. In this review, we describe the Ca2+-dependent host factors regulating antibacterial immunity, in addition to bacterial effectors that promote, inhibit, or co-opt Ca2+-calmodulin signalling to promote infection.
Collapse
Affiliation(s)
- Joseph J Wanford
- School of Immunology and Microbial Sciences, Kings College London, London, UK
| | - Charlotte Odendall
- School of Immunology and Microbial Sciences, Kings College London, London, UK.
| |
Collapse
|
17
|
Abstract
Listeria monocytogenes is a Gram-positive facultative intracellular pathogen that can cause severe invasive infections upon ingestion with contaminated food. Clinically, listerial disease, or listeriosis, most often presents as bacteremia, meningitis or meningoencephalitis, and pregnancy-associated infections manifesting as miscarriage or neonatal sepsis. Invasive listeriosis is life-threatening and a main cause of foodborne illness leading to hospital admissions in Western countries. Sources of contamination can be identified through international surveillance systems for foodborne bacteria and strains' genetic data sharing. Large-scale whole genome studies have increased our knowledge on the diversity and evolution of L. monocytogenes, while recent pathophysiological investigations have improved our mechanistic understanding of listeriosis. In this article, we present an overview of human listeriosis with particular focus on relevant features of the causative bacterium, epidemiology, risk groups, pathogenesis, clinical manifestations, and treatment and prevention.
Collapse
Affiliation(s)
- Merel M Koopmans
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Matthijs C Brouwer
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - José A Vázquez-Boland
- Infection Medicine, Edinburgh Medical School (Biomedical Sciences), University of Edinburgh, Edinburgh, United Kingdom
| | - Diederik van de Beek
- Amsterdam UMC, University of Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Pittner NA, Solomon RN, Bui DC, McBride JW. Ehrlichia effector SLiM-icry: Artifice of cellular subversion. Front Cell Infect Microbiol 2023; 13:1150758. [PMID: 36960039 PMCID: PMC10028187 DOI: 10.3389/fcimb.2023.1150758] [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: 01/24/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
As an obligately intracellular bacterial pathogen that selectively infects the mononuclear phagocyte, Ehrlichia chaffeensis has evolved sophisticated mechanisms to subvert innate immune defenses. While the bacterium accomplishes this through a variety of mechanisms, a rapidly expanding body of evidence has revealed that E. chaffeensis has evolved survival strategies that are directed by the versatile, intrinsically disordered, 120 kDa tandem repeat protein (TRP120) effector. E. chaffeensis establishes infection by manipulating multiple evolutionarily conserved cellular signaling pathways through effector-host interactions to subvert innate immune defenses. TRP120 activates these pathways using multiple functionally distinct, repetitive, eukaryote-mimicking short linear motifs (SLiMs) located within the tandem repeat domain that have evolved in nihilo. Functionally, the best characterized TRP120 SLiMs mimic eukaryotic ligands (SLiM-icry) to engage pathway-specific host receptors and activate cellular signaling, thereby repurposing these pathways to promote infection. Moreover, E. chaffeensis TRP120 contains SLiMs that are targets of post-translational modifications such as SUMOylation in addition to many other validated SLiMs that are curated in the eukaryotic linear motif (ELM) database. This review will explore the extracellular and intracellular roles TRP120 SLiM-icry plays during infection - mediated through a variety of SLiMs - that enable E. chaffeensis to subvert mononuclear phagocyte innate defenses.
Collapse
Affiliation(s)
- Nicholas A. Pittner
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Regina N. Solomon
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Duc-Cuong Bui
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
19
|
The Application of Cinnamon Twig Extract as an Inhibitor of Listeriolysin O against Listeria monocytogenes Infection. Molecules 2023; 28:molecules28041625. [PMID: 36838612 PMCID: PMC9962927 DOI: 10.3390/molecules28041625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 02/11/2023] Open
Abstract
As a major virulence factor of Listeria monocytogenes (L. monocytogenes), listeriolysin O (LLO) can assist in the immune escape of L. monocytogenes, which is critical for the pathogen to evade host immune recognition, leading to various infectious diseases. Cinnamon twig (CT), as a traditional medicine, has been widely used in clinics for multiple functions and it has exhibited excellent safety, efficacy and stability. There are few reports on the effects of the extracts of traditional medicine on bacterial virulence factors. CT has not been reported to be effective in the treatment of L. monocytogenes infection. Therefore, this study aims to explore the preventive effect of CT against L. monocytogenes infection in vivo and in vitro by targeting LLO. Firstly, a hemolysis assay and a cell viability determination are used to detect the effect of CT extract on the inhibition of the cytolytic activity of LLO. The potential mechanism through which CT extract inhibits LLO activity is predicted through network pharmacology, molecular docking assay, real-time polymerase chain reaction (RT-PCR), Western blotting and circular dichroism (CD) analysis. The experimental therapeutic effect of CT extract is examined in a mouse model infected with L. monocytogenes. Then, the ingredients are identified through a high-performance liquid chromatography (HPLC) and thin layer chromatography (TLC) analysis. Here we find that CT extract, containing mainly cinnamic acid, cinnamaldehyde, β-sitosterol, taxifolin, catechin and epicatechin, shows a potential inhibition of LLO-mediated hemolysis without any antimicrobial activity. The results of the mechanism research show that CT extract treatment can simultaneously inhibit LLO expression and oligomerization. Furthermore, the addition of CT extract led to a remarkable alleviation of LLO-induced cytotoxicity. After treatment with CT extract, the mortality, bacterial load, pathological damage and inflammatory responses of infected mice are significantly reduced when compared with the untreated group. This study suggests that CT extract can be a novel and multicomponent inhibitor of LLO with multiple strategies against L. monocytogenes infection, which could be further developed into a novel treatment for infections caused by L. monocytogenes.
Collapse
|
20
|
Karthikeyan R, Gayathri P, Ramasamy S, Suvekbala V, Jagannadham MV, Rajendhran J. Transcriptome responses of intestinal epithelial cells induced by membrane vesicles of Listeria monocytogenes. CURRENT RESEARCH IN MICROBIAL SCIENCES 2023; 4:100185. [PMID: 36942003 PMCID: PMC10023947 DOI: 10.1016/j.crmicr.2023.100185] [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: 03/08/2023] Open
Abstract
Membrane vesicles (MVs) serve as an essential virulence factor in several pathogenic bacteria. The release of MVs by Listeria monocytogenes is only recently recognized; still, the enigmatic role of MVs in pathogenesis is yet to be established. We report the transcriptome response of Caco-2 cells upon exposure to MVs and the L. monocytogenes that leads to observe the up-regulation of autophagy-related genes in the early phase of exposure to MVs. Transcription of inflammatory cytokines is to the peak at the fourth hour of exposure. An array of differentially expressed genes was associated with actin cytoskeleton rearrangement, autophagy, cell cycle arrest, and induction of oxidative stress. At a later time point, transcriptional programs are generated upon interaction with MVs to evade innate immune signals, by modulating the expression of anti-inflammatory genes. KEGG pathway analysis is palpably confirming that MVs appear principally responsible for the induction of immune signaling pathways. Besides, MVs induced the expression of cell cycle regulatory genes, likely responsible for the ability to prolong host cell survival, thus protecting the replicative niche for L. monocytogenes. Notably, we identified several non-coding RNAs (ncRNAs), possibly involved in the regulation of early manipulation of the host gene expression, essential for the persistence of L. monocytogenes.
Collapse
Affiliation(s)
- Raman Karthikeyan
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625021, Tamil Nadu, India
| | - Pratapa Gayathri
- CSIR - Centre for Cellular and Molecular Biology, Tarnaka, Hyderabad 500007, India
| | - Subbiah Ramasamy
- Department of Biochemistry, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625021, Tamil Nadu, India
| | - Vemparthan Suvekbala
- EDII-Anna Business Incubation Research Foundation, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli 620024, India
| | - Medicharla V. Jagannadham
- CSIR - Centre for Cellular and Molecular Biology, Tarnaka, Hyderabad 500007, India
- Corresponding authors.
| | - Jeyaprakash Rajendhran
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625021, Tamil Nadu, India
- Corresponding authors.
| |
Collapse
|
21
|
Listeria monocytogenes-How This Pathogen Uses Its Virulence Mechanisms to Infect the Hosts. Pathogens 2022; 11:pathogens11121491. [PMID: 36558825 PMCID: PMC9783847 DOI: 10.3390/pathogens11121491] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Listeriosis is a serious food-borne illness, especially in susceptible populations, including children, pregnant women, and elderlies. The disease can occur in two forms: non-invasive febrile gastroenteritis and severe invasive listeriosis with septicemia, meningoencephalitis, perinatal infections, and abortion. Expression of each symptom depends on various bacterial virulence factors, immunological status of the infected person, and the number of ingested bacteria. Internalins, mainly InlA and InlB, invasins (invasin A, LAP), and other surface adhesion proteins (InlP1, InlP4) are responsible for epithelial cell binding, whereas internalin C (InlC) and actin assembly-inducing protein (ActA) are involved in cell-to-cell bacterial spread. L. monocytogenes is able to disseminate through the blood and invade diverse host organs. In persons with impaired immunity, the elderly, and pregnant women, the pathogen can also cross the blood-brain and placental barriers, which results in the invasion of the central nervous system and fetus infection, respectively. The aim of this comprehensive review is to summarize the current knowledge on the epidemiology of listeriosis and L. monocytogenes virulence mechanisms that are involved in host infection, with a special focus on their molecular and cellular aspects. We believe that all this information is crucial for a better understanding of the pathogenesis of L. monocytogenes infection.
Collapse
|
22
|
Xu L, Lu G, Zhan B, Wei L, Deng X, Zhang Q, Shen X, Wang J, Feng H. Uncovering the efficacy and mechanisms of Genkwa flos and bioactive ingredient genkwanin against L. monocytogenes infection. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115571. [PMID: 35870686 DOI: 10.1016/j.jep.2022.115571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Genkwa flos (yuanhua in Chinese), the dried flower buds of the plant Daphne genkwa Siebold & Zucc., as a traditional herb widely used for the treatment of inflammation-related symptoms and diseases, with the efficacies of diuretic, phlegm-resolving and cough suppressant. AIM OF THE STUDY Traditional Chinese Medicine (TCM) is presumed to be of immense potential against pathogens infection. Whereas, the potential efficacy and mechanisms of Genkwa flos against L. monocytogenes infection has not been extensively explored. The present study aimed to identify the bioactive ingredients of Genkwa flos against L. monocytogenes infection and to delineate the underlying mechanisms of action. MATERIALS AND METHODS Bioinformatics approach at protein network level was employed to investigate the therapeutic mechanisms of Genkwa flos against L. monocytogenes infection. And hemolysis inhibition assay, cytoprotection test, western blotting, oligomerization assay and molecular docking analysis were applied to substantiate the multiple efficacies of Genkwa flos and the bioactive ingredient genkwanin. Histopathological analysis and biochemistry detection were conducted to evaluate the in vivo protective effect of genkwanin. RESULTS Network pharmacology and experimental validation revealed that Traditional Chinese Medicine (TCM) Genkwa flos exhibited anti-L. monocytogenes potency and was found to inhibit the hemolytic activity of LLO. Bioactive ingredient genkwanin interfered with the pore-forming activity of LLO by engaging the active residues Tyr414, Tyr98, Asn473, Val100, Tyr440 and Val438, and thereby attenuated LLO-mediated cytotoxicity. Consistent with the bioinformatics prediction, exposed to genkwanin could upregulate the Nrf2 level and promote the translocation of Nrf2. In vivo, genkwanin oral administration (80 mg/kg) significantly protected against systemic L. monocytogenes infection, as evidenced by reduced myeloperoxidase (MPO) and malondialdehyde (MDA) levels, increased mice survival rate by 30% and decreased pathogen colonization. CONCLUSION Our study demonstrated that Genkwa flos is a potential anti-L. monocytogenes TCM, highlighted the therapeutic potential of Genkwa flos active ingredient genkwanin by targeting the pore-forming cytolysin LLO and acting as a promising antioxidative candidate against L. monocytogenes infection.
Collapse
Affiliation(s)
- Lei Xu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Gejin Lu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Yujinxiang Street 573, Changchun, Jilin, 130122, China.
| | - Baihe Zhan
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Lijuan Wei
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China; Hebei Veterinary Medicine Technology Innovation Center, Shijiazhuang, 050041, Hebei, China.
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Qiaoling Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Xue Shen
- Department of Food Science, College of Food Science and Engineering, Jilin University, Changchun, China.
| | - Jianfeng Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Haihua Feng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
23
|
Mayer RL, Verbeke R, Asselman C, Aernout I, Gul A, Eggermont D, Boucher K, Thery F, Maia TM, Demol H, Gabriels R, Martens L, Bécavin C, De Smedt SC, Vandekerckhove B, Lentacker I, Impens F. Immunopeptidomics-based design of mRNA vaccine formulations against Listeria monocytogenes. Nat Commun 2022; 13:6075. [PMID: 36241641 PMCID: PMC9562072 DOI: 10.1038/s41467-022-33721-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/29/2022] [Indexed: 12/24/2022] Open
Abstract
Listeria monocytogenes is a foodborne intracellular bacterial pathogen leading to human listeriosis. Despite a high mortality rate and increasing antibiotic resistance no clinically approved vaccine against Listeria is available. Attenuated Listeria strains offer protection and are tested as antitumor vaccine vectors, but would benefit from a better knowledge on immunodominant vector antigens. To identify novel antigens, we screen for Listeria peptides presented on the surface of infected human cell lines by mass spectrometry-based immunopeptidomics. In between more than 15,000 human self-peptides, we detect 68 Listeria immunopeptides from 42 different bacterial proteins, including several known antigens. Peptides presented on different cell lines are often derived from the same bacterial surface proteins, classifying these antigens as potential vaccine candidates. Encoding these highly presented antigens in lipid nanoparticle mRNA vaccine formulations results in specific CD8+ T-cell responses and induces protection in vaccination challenge experiments in mice. Our results can serve as a starting point for the development of a clinical mRNA vaccine against Listeria and aid to improve attenuated Listeria vaccines and vectors, demonstrating the power of immunopeptidomics for next-generation bacterial vaccine development.
Collapse
Affiliation(s)
- Rupert L Mayer
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- VIB Proteomics Core, VIB, Ghent, Belgium
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, Vienna, Austria
| | - Rein Verbeke
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Caroline Asselman
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Ilke Aernout
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Adillah Gul
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Denzel Eggermont
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Katie Boucher
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- VIB Proteomics Core, VIB, Ghent, Belgium
| | - Fabien Thery
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Teresa M Maia
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- VIB Proteomics Core, VIB, Ghent, Belgium
| | - Hans Demol
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- VIB Proteomics Core, VIB, Ghent, Belgium
| | - Ralf Gabriels
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lennart Martens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | | | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Bart Vandekerckhove
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, 9000, Ghent, Belgium
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.
- VIB Proteomics Core, VIB, Ghent, Belgium.
| |
Collapse
|
24
|
Invading Bacterial Pathogens Activate Transcription Factor EB in Epithelial Cells through the Amino Acid Starvation Pathway of mTORC1 Inhibition. Mol Cell Biol 2022; 42:e0024122. [PMID: 36005752 PMCID: PMC9476939 DOI: 10.1128/mcb.00241-22] [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] [Indexed: 11/20/2022] Open
Abstract
Upon pathogen infection, intricate innate signaling cascades are induced to initiate the transcription of immune effectors, including cytokines and chemokines. Transcription factor EB (TFEB), a master regulator of lysosomal biogenesis and autophagy genes, was found recently to be a novel regulator of innate immunity in both Caenorhabditis elegans and mammals. Despite TFEB participating in critical mechanisms of pathogen recognition and in the transcriptional response to infection in mammalian macrophages, little is known about its roles in the infected epithelium or infected nonimmune cells in general. Here, we demonstrate that TFEB is activated in nonimmune cells upon infection with bacterial pathogens through a pathway dependent on mTORC1 inhibition and RAG-GTPase activity, reflecting the importance of membrane damage and amino acid starvation responses during infection. Additionally, we present data demonstrating that although TFEB does not affect bacterial killing or load in nonimmune cells, it alters the host transcriptome upon infection, thus promoting an antibacterial transcriptomic landscape. Elucidating the roles of TFEB in infected nonimmune cells and the upstream signaling cascade provides critical insight into understanding how cells recognize and respond to bacterial pathogens.
Collapse
|
25
|
Osek J, Lachtara B, Wieczorek K. Listeria monocytogenes in foods-From culture identification to whole-genome characteristics. Food Sci Nutr 2022; 10:2825-2854. [PMID: 36171778 PMCID: PMC9469866 DOI: 10.1002/fsn3.2910] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/06/2022] [Accepted: 04/19/2022] [Indexed: 12/03/2022] Open
Abstract
Listeria monocytogenes is an important foodborne pathogen, which is able to persist in the food production environments. The presence of these bacteria in different niches makes them a potential threat for public health. In the present review, the current information on the classical and alternative methods used for isolation and identification of L. monocytogenes in food have been described. Although these techniques are usually simple, standardized, inexpensive, and are routinely used in many food testing laboratories, several alternative molecular-based approaches for the bacteria detection in food and food production environments have been developed. They are characterized by the high sample throughput, a short time of analysis, and cost-effectiveness. However, these methods are important for the routine testing toward the presence and number of L. monocytogenes, but are not suitable for characteristics and typing of the bacterial isolates, which are crucial in the study of listeriosis infections. For these purposes, novel approaches, with a high discriminatory power to genetically distinguish the strains during epidemiological studies, have been developed, e.g., whole-genome sequence-based techniques such as NGS which provide an opportunity to perform comparison between strains of the same species. In the present review, we have shown a short description of the principles of microbiological, alternative, and modern methods of detection of L. monocytogenes in foods and characterization of the isolates for epidemiological purposes. According to our knowledge, similar comprehensive papers on such subject have not been recently published, and we hope that the current review may be interesting for research communities.
Collapse
Affiliation(s)
- Jacek Osek
- Department of Hygiene of Food of Animal OriginNational Veterinary Research InstitutePuławyPoland
| | - Beata Lachtara
- Department of Hygiene of Food of Animal OriginNational Veterinary Research InstitutePuławyPoland
| | - Kinga Wieczorek
- Department of Hygiene of Food of Animal OriginNational Veterinary Research InstitutePuławyPoland
| |
Collapse
|
26
|
Sibanda T, Buys EM. Listeria monocytogenes Pathogenesis: The Role of Stress Adaptation. Microorganisms 2022; 10:microorganisms10081522. [PMID: 36013940 PMCID: PMC9416357 DOI: 10.3390/microorganisms10081522] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/08/2022] [Accepted: 07/19/2022] [Indexed: 12/13/2022] Open
Abstract
Adaptive stress tolerance responses are the driving force behind the survival ability of Listeria monocytogenes in different environmental niches, within foods, and ultimately, the ability to cause human infections. Although the bacterial stress adaptive responses are primarily a necessity for survival in foods and the environment, some aspects of the stress responses are linked to bacterial pathogenesis. Food stress-induced adaptive tolerance responses to acid and osmotic stresses can protect the pathogen against similar stresses in the gastrointestinal tract (GIT) and, thus, directly aid its virulence potential. Moreover, once in the GIT, the reprogramming of gene expression from the stress survival-related genes to virulence-related genes allows L. monocytogenes to switch from an avirulent to a virulent state. This transition is controlled by two overlapping and interlinked transcriptional networks for general stress response (regulated by Sigma factor B, (SigB)) and virulence (regulated by the positive regulatory factor A (PrfA)). This review explores the current knowledge on the molecular basis of the connection between stress tolerance responses and the pathogenesis of L. monocytogenes. The review gives a detailed background on the currently known mechanisms of pathogenesis and stress adaptation. Furthermore, the paper looks at the current literature and theories on the overlaps and connections between the regulatory networks for SigB and PrfA.
Collapse
Affiliation(s)
- Thulani Sibanda
- Department of Consumer and Food Sciences, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa;
- Department of Applied Biology and Biochemistry, National University of Science and Technology, Bulawayo P.O. Box AC939, Zimbabwe
| | - Elna M. Buys
- Department of Consumer and Food Sciences, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa;
- Correspondence:
| |
Collapse
|
27
|
Kaempferol-Driven Inhibition of Listeriolysin O Pore Formation and Inflammation Suppresses Listeria monocytogenes Infection. Microbiol Spectr 2022; 10:e0181022. [PMID: 35856678 PMCID: PMC9431489 DOI: 10.1128/spectrum.01810-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Listeria monocytogenes remains a nonnegligible cause of foodborne infection, posing a critical threat to public health. Under the global antibiotic crisis, novel alternative approaches are urgently needed. The indispensable role of listeriolysin O (LLO) in the intracellular life cycle, barrier penetration, colonization, and systemic dissemination of L. monocytogenes renders it a potent drug target, which means curbing L. monocytogenes via interfering with LLO-associated pathogenic mechanisms. Here, we identified kaempferol, a natural small molecule compound, as an effective LLO inhibitor that engaged the residues Glu437, Ile468, and Tyr469 of LLO, thereby suppressing LLO-mediated membrane perforation and barrier disruption. Moreover, we found that kaempferol also suppressed host-derived inflammation in a distinct way independent of LLO inhibition. The in vivo study revealed that kaempferol treatment significantly reduced bacterial burden and cytokine burst in target organs, thereby effectively protecting mice from systemic L. monocytogenes infection. Our findings present kaempferol as a potential therapeutic application for L. monocytogenes infection, which is less likely to induce drug resistance than antibiotics because of its superiority of interfering with the pathogenesis process rather than exerting pressure on bacterial viability. IMPORTANCE Currently, we are facing a global crisis of antibiotic resistance, and novel alternative approaches are urgently needed to curb L. monocytogenes infection. Our study demonstrated that kaempferol alleviated L. monocytogenes infection via suppressing LLO pore formation and inflammation response, which might represent a novel antimicrobial-independent strategy to curb listeriosis.
Collapse
|
28
|
Rukit J, Boonmee A, Kijpornyongpan T, Tulsook K, Baranyi J, Chaturongakul S. Roles of Alternative Sigma Factors in Invasion and Growth Characteristics of Listeria monocytogenes 10403S Into Human Epithelial Colorectal Adenocarcinoma Caco-2 Cell. Front Microbiol 2022; 13:901484. [PMID: 35910626 PMCID: PMC9329085 DOI: 10.3389/fmicb.2022.901484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022] Open
Abstract
Listeria monocytogenes is a Gram-positive facultative intracellular bacterium with a broad host range. With its housekeeping sigma factor and four alternative ones (namely SigB, SigC, SigH, and SigL), L. monocytogenes can express genes in response to changing environments. However, the roles of these sigma factors in intracellular survival are still unclear. The objectives of this study were to characterize the role of each alternative σ factor on L. monocytogenes invasion and growth inside human epithelial colorectal adenocarcinoma Caco-2 cells. We used L. monocytogenes 10403S wild type and its 15 alternative sigma factor deletion mutants at a multiplicity of infection of 100 and 1 in invasion and intracellular growth assays in the Caco-2 cells, respectively. At 1.5, 2, 4, 6, 8, 10, and 12 h post-infection, Caco-2 cells were lysed, and intracellular L. monocytogenes were enumerated on brain-heart infusion agar. Colony-forming and growth rates were compared among strains. The results from phenotypic characterization confirmed that (i) SigB is the key factor for L. monocytogenes invasion and (ii) having only SigA (ΔsigBCHL strain) is sufficient to invade and multiply in the host cell at similar levels as the wild type. Our previous study suggested the negative role of SigL in bile stress response. In this study, we have shown that additional deletion of the rpoN (or sigL) gene to ΔsigB, ΔsigC, or ΔsigH could restore the impaired invasion efficiencies of the single mutant, suggesting the absence of SigL could enhance host invasion. Therefore, we further investigated the role of SigL during extracellular and intracellular life cycles. Using RNA sequencing, we identified 118 and 16 SigL-dependent genes during the extracellular and intracellular life cycles, respectively. The sigL gene itself was induced by fivefolds prior to the invasion, and 5.3 folds during Caco-2 infection, further suggesting the role of SigL in intracellular growth.
Collapse
Affiliation(s)
- Junyaluck Rukit
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Atsadang Boonmee
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Teeratas Kijpornyongpan
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN, United States
| | - Kan Tulsook
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - József Baranyi
- Institute of Nutrition, University of Debrecen, Debrecen, Hungary
| | - Soraya Chaturongakul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Molecular Medical Biosciences Cluster, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
- *Correspondence: Soraya Chaturongakul,
| |
Collapse
|
29
|
Saravanakumar K, SivaSantosh S, Sathiyaseelan A, Naveen KV, AfaanAhamed MA, Zhang X, Priya VV, MubarakAli D, Wang MH. Unraveling the hazardous impact of diverse contaminants in the marine environment: Detection and remedial approach through nanomaterials and nano-biosensors. JOURNAL OF HAZARDOUS MATERIALS 2022; 433:128720. [PMID: 35366447 DOI: 10.1016/j.jhazmat.2022.128720] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/28/2022] [Accepted: 03/14/2022] [Indexed: 06/14/2023]
Abstract
Marine pollution is one of the most underlooked forms of pollution as it affects most aquatic lives and public health in the coastal area. The diverse form of the hazardous pollutant in the marine ecosystem leads the serious genetic level disorders and diseases which include cancer, diabetes, arthritis, reproductive, and neurological diseases such as Parkinson's, Alzheimer's, and several microbial infections. Therefore, a recent alarming study on these pollutants, the microplastics have been voiced out in many countries worldwide, it was even found to be in the human placenta. In recent times, nanomaterials have demonstrated their potential in the detection and remediation of sensitive contaminants. In this review, we presented a comprehensive overview of the source, and distribution of diverse marine pollution on both aquatic and human health by summarizing the concentration of diverse pollutions (heavy metals, pesticides, microbial toxins, and micro/nano plastics) in marine samples such as soil, water, and seafood. Followed by emphasizing its ecotoxicological impact on aquatic animal life and coastal public health. Also discussed are the applicability and advancements of nanomaterials and nano-based biosensors in the detection, prevention, and remediation of diverse pollution in the marine ecosystem.
Collapse
Affiliation(s)
- Kandasamy Saravanakumar
- Department of Bio-Health convergence, Kangwon National University, Chuncheon 200-701, Republic of Korea.
| | | | - Anbazhagan Sathiyaseelan
- Department of Bio-Health convergence, Kangwon National University, Chuncheon 200-701, Republic of Korea.
| | - Kumar Vishven Naveen
- Department of Bio-Health convergence, Kangwon National University, Chuncheon 200-701, Republic of Korea.
| | - Mohamed Ali AfaanAhamed
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, Tamil Nadu 600048, India.
| | - Xin Zhang
- Department of Bio-Health convergence, Kangwon National University, Chuncheon 200-701, Republic of Korea.
| | - Veeraraghavan Vishnu Priya
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India.
| | - Davoodbasha MubarakAli
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, Tamil Nadu 600048, India.
| | - Myeong-Hyeon Wang
- Department of Bio-Health convergence, Kangwon National University, Chuncheon 200-701, Republic of Korea.
| |
Collapse
|
30
|
Li Y, Ma X, Yue Y, Zhang K, Cheng K, Feng Q, Ma N, Liang J, Zhang T, Zhang L, Chen Z, Wang X, Ren L, Zhao X, Nie G. Rapid Surface Display of mRNA Antigens by Bacteria-Derived Outer Membrane Vesicles for a Personalized Tumor Vaccine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109984. [PMID: 35315546 DOI: 10.1002/adma.202109984] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/21/2022] [Indexed: 06/14/2023]
Abstract
Therapeutic mRNA vaccination is an attractive approach to trigger antitumor immunity. However, the mRNA delivery technology for customized tumor vaccine is still limited. In this work, bacteria-derived outer membrane vesicles (OMVs) are employed as an mRNA delivery platform by genetically engineering with surface decoration of RNA binding protein, L7Ae, and lysosomal escape protein, listeriolysin O (OMV-LL). OMV-LL can rapidly adsorb box C/D sequence-labelled mRNA antigens through L7Ae binding (OMV-LL-mRNA) and deliver them into dendritic cells (DCs), following by the cross-presentation via listeriolysin O-mediated endosomal escape. OMV-LL-mRNA significantly inhibits melanoma progression and elicits 37.5% complete regression in a colon cancer model. OMV-LL-mRNA induces a long-term immune memory and protects the mice from tumor challenge after 60 days. In summary, this platform provides a delivery technology distinct from lipid nanoparticles (LNPs) for personalized mRNA tumor vaccination, and with a "Plug-and-Display" strategy that enables its versatile application in mRNA vaccines.
Collapse
Affiliation(s)
- Yao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, College of Materials, Xiamen University, Xiamen, Fujian, 361005, China
| | - Xiaotu Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Yale Yue
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Kaiyue Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Qingqing Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Nana Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Tianjiao Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Lizhuo Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Zhiqiang Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xinwei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Lei Ren
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, College of Materials, Xiamen University, Xiamen, Fujian, 361005, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Xiamen, Beijing, 100101, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Xiamen, Beijing, 100049, China
- The GBA National Institute for Nanotechnology Innovation, Guangdong, 510700, China
| |
Collapse
|
31
|
Herb M, Gluschko A, Farid A, Krönke M. When the Phagosome Gets Leaky: Pore-Forming Toxin-Induced Non-Canonical Autophagy (PINCA). Front Cell Infect Microbiol 2022; 12:834321. [PMID: 35372127 PMCID: PMC8968195 DOI: 10.3389/fcimb.2022.834321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Macrophages remove bacteria from the extracellular milieu via phagocytosis. While most of the engulfed bacteria are degraded in the antimicrobial environment of the phagolysosome, several bacterial pathogens have evolved virulence factors, which evade degradation or allow escape into the cytosol. To counter this situation, macrophages activate LC3-associated phagocytosis (LAP), a highly bactericidal non-canonical autophagy pathway, which destroys the bacterial pathogens in so called LAPosomes. Moreover, macrophages can also target intracellular bacteria by pore-forming toxin-induced non-canonical autophagy (PINCA), a recently described non-canonical autophagy pathway, which is activated by phagosomal damage induced by bacteria-derived pore-forming toxins. Similar to LAP, PINCA involves LC3 recruitment to the bacteria-containing phagosome independently of the ULK complex, but in contrast to LAP, this process does not require ROS production by Nox2. As last resort of autophagic targeting, macrophages activate xenophagy, a selective form of macroautophagy, to recapture bacteria, which evaded successful targeting by LAP or PINCA through rupture of the phagosome. However, xenophagy can also be hijacked by bacterial pathogens for their benefit or can be completely inhibited resulting in intracellular growth of the bacterial pathogen. In this perspective, we discuss the molecular differences and similarities between LAP, PINCA and xenophagy in macrophages during bacterial infections.
Collapse
Affiliation(s)
- Marc Herb
- Faculty of Medicine and University Hospital of Cologne, Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Alexander Gluschko
- Faculty of Medicine and University Hospital of Cologne, Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Alina Farid
- Faculty of Medicine and University Hospital of Cologne, Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Martin Krönke
- Faculty of Medicine and University Hospital of Cologne, Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- German Center for Infection Research, Bonn-Cologne, Germany
| |
Collapse
|
32
|
Molecular characterization and hematological analysis of Listeria monocytogenes infection in dairy cows in Punjab (Pakistan). Arch Microbiol 2022; 204:201. [PMID: 35239048 DOI: 10.1007/s00203-022-02804-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 11/02/2022]
Abstract
Listeriosis is an emerging bacterial disease of animals and humans worldwide, caused by Listeria monocytogenes. The infected dairy cows continuously shed the microbes in their milk, a human being's concern. This study was designed to molecular characterize the Listeria monocytogenes isolated from symptomatic cow's milk of tehsils Samundri, Gujar khan, and Alipur of Punjab. A total of 175 milk samples were collected, pre-enriched and cultured on PALCAM agar. The affirmation of the hlyA gene of Listeria monocytogenes was performed by polymerase chain reaction (PCR) and 3.43% of isolates were found positive. The phylogenetic analysis showed a resemblance of our isolates of Listeria monocytogenes with India (KP965733), the USA (DQ812484), and 3 of our isolates made a clade. The leucocytes and neutrophils count were found significantly increased in listeriosis affected cows. The Chi-square test showed a significant association between poor quality silage feeding and listeriosis. The presence of L.monocytogenes in cow's milk indicates a potential threat to humans. It is further recommended that it should be consistently monitored to ensure food safety.
Collapse
|
33
|
Antimicrobial Activity of Ohelo Berry (Vaccinium calycinum) Juice against Listeria monocytogenes and Its Potential for Milk Preservation. Microorganisms 2022; 10:microorganisms10030548. [PMID: 35336123 PMCID: PMC8953330 DOI: 10.3390/microorganisms10030548] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
Listeria monocytogenes is a foodborne pathogen and causes illnesses with a high mortality rate in susceptible populations. Several dairy-related outbreaks have been attributed to contamination by L. monocytogenes, which requires antimicrobial interventions to enhance the safety of these products. This study aimed to determine the antimicrobial activity of the ohelo berry (Vaccinium calycinum), a Hawaiian wild relative of cranberry, against L. monocytogenes in culture media and milk products. The effect of ohelo berry juice at its sub-inhibitory concentrations on the physicochemical properties, biofilm formation, and gene expression of L. monocytogenes was also investigated. The minimum inhibitory concentration of ohelo berry juice against L. monocytogenes was 12.5%. The sub-inhibitory concentration of ohelo berry juice (6.25%) significantly increased the auto-aggregation and decreased the hydrophobicity, swimming motility, swarming motility, and biofilm formation capability of L. monocytogenes. The relative expression of genes for motility (flaA), biofilm formation and disinfectant resistance (sigB), invasion (iap), listeriolysin (hly), and phospholipase (plcA) was significantly downregulated in L. monocytogenes treated by the 6.25% juice. L. monocytogenes was significantly inhibited in whole and skim milk supplemented with 50% ohelo berry juice, regardless of the fat content. These findings highlight the potential of ohelo berry as a natural preservative and functional food to prevent L. monocytogenes infection.
Collapse
|
34
|
Tanishita Y, Sekiya H, Inohara N, Tsuchiya K, Mitsuyama M, Núñez G, Hara H. Listeria toxin promotes phosphorylation of the inflammasome adaptor ASC through Lyn and Syk to exacerbate pathogen expansion. Cell Rep 2022; 38:110414. [PMID: 35196496 DOI: 10.1016/j.celrep.2022.110414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 10/08/2021] [Accepted: 01/31/2022] [Indexed: 12/20/2022] Open
Abstract
Inflammasome activation exacerbates infectious disease caused by pathogens such as Listeria monocytogenes, Staphylococcus aureus, and severe acute respiratory syndrome coronavirus 2. Although these pathogens activate host inflammasomes to regulate pathogen expansion, the mechanisms by which pathogen toxins contribute to inflammasome activation remain poorly understood. Here we show that activation of inflammasomes by Listeria infection is promoted by amino acid residue T223 of listeriolysin O (LLO) independently of its pore-forming activity. LLO T223 is critical for phosphorylation of the inflammasome adaptor ASC at amino acid residue Y144 through Lyn-Syk signaling, which is essential for ASC oligomerization. Notably, a Listeria mutant expressing LLO T223A is impaired in inducing ASC phosphorylation and inflammasome activation. Furthermore, the virulence of LLO T223A mutant is markedly attenuated in vivo due to impaired ability to activate the inflammasome. Our results reveal a function of a pathogen toxin that exacerbates infection by promoting phosphorylation of ASC.
Collapse
Affiliation(s)
- Yuko Tanishita
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hisateru Sekiya
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Naohiro Inohara
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Masao Mitsuyama
- Department of Microbiology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8501, Japan
| | - Gabriel Núñez
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hideki Hara
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
35
|
Sriwastva MK, Deng Z, Wang B, Teng Y, Kumar A, Sundaram K, Mu J, Lei C, Dryden GW, Xu F, Zhang L, Yan J, Zhang X, Park JW, Merchant ML, Egilmez NK, Zhang H. Exosome-like nanoparticles from Mulberry bark prevent DSS-induced colitis via the AhR/COPS8 pathway. EMBO Rep 2022; 23:e53365. [PMID: 34994476 PMCID: PMC8892346 DOI: 10.15252/embr.202153365] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/02/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Bark protects the tree against environmental insults. Here, we analyzed whether this defensive strategy could be utilized to broadly enhance protection against colitis. As a proof of concept, we show that exosome-like nanoparticles (MBELNs) derived from edible mulberry bark confer protection against colitis in a mouse model by promoting heat shock protein family A (Hsp70) member 8 (HSPA8)-mediated activation of the AhR signaling pathway. Activation of this pathway in intestinal epithelial cells leads to the induction of COP9 Constitutive Photomorphogenic Homolog Subunit 8 (COPS8). Utilizing a gut epithelium-specific knockout of COPS8, we demonstrate that COPS8 acts downstream of the AhR pathway and is required for the protective effect of MBELNs by inducing an array of anti-microbial peptides. Our results indicate that MBELNs represent an undescribed mode of inter-kingdom communication in the mammalian intestine through an AhR-COPS8-mediated anti-inflammatory pathway. These data suggest that inflammatory pathways in a microbiota-enriched intestinal environment are regulated by COPS8 and that edible plant-derived ELNs may hold the potential as new agents for the prevention and treatment of gut-related inflammatory disease.
Collapse
Affiliation(s)
- Mukesh K Sriwastva
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Zhong‐Bin Deng
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Bomei Wang
- Department of Translational OncologyGenentechSan FranciscoCaliforniaUSA
| | - Yun Teng
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Anil Kumar
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Kumaran Sundaram
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Jingyao Mu
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Chao Lei
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Gerald W Dryden
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
- Robley Rex Veterans Affairs Medical CenterLouisvilleKYUSA
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKYUSA
| | - Fangyi Xu
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Lifeng Zhang
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Jun Yan
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Xiang Zhang
- KBRIN Bioinformatics CoreUniversity of LouisvilleLouisvilleKYUSA
| | - Juw Won Park
- KBRIN Bioinformatics CoreUniversity of LouisvilleLouisvilleKYUSA
- Department of Computer Engineering and Computer ScienceUniversity of LouisvilleLouisvilleKYUSA
| | - Michael L Merchant
- Kidney Disease Program and Clinical Proteomics CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Nejat K Egilmez
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Huang‐Ge Zhang
- Department of Microbiology & ImmunologyBrown Cancer CenterUniversity of LouisvilleLouisvilleKYUSA
- Robley Rex Veterans Affairs Medical CenterLouisvilleKYUSA
| |
Collapse
|
36
|
Recent Advancements in Tracking Bacterial Effector Protein Translocation. Microorganisms 2022; 10:microorganisms10020260. [PMID: 35208715 PMCID: PMC8876096 DOI: 10.3390/microorganisms10020260] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/17/2022] Open
Abstract
Bacteria-host interactions are characterized by the delivery of bacterial virulence factors, i.e., effectors, into host cells where they counteract host immunity and exploit host responses allowing bacterial survival and spreading. These effectors are translocated into host cells by means of dedicated secretion systems such as the type 3 secretion system (T3SS). A comprehensive understanding of effector translocation in a spatio-temporal manner is of critical importance to gain insights into an effector’s mode of action. Various approaches have been developed to understand timing and order of effector translocation, quantities of translocated effectors and their subcellular localization upon translocation into host cells. Recently, the existing toolset has been expanded by newly developed state-of-the art methods to monitor bacterial effector translocation and dynamics. In this review, we elaborate on reported methods and discuss recent advances and shortcomings in this area of tracking bacterial effector translocation.
Collapse
|
37
|
Zhang Y, Wang H, Shang K, Wang X, Xu P. Reliable detection of Listeria monocytogenes by a portable paper-based multi-biocatalyst platform integrating three biomarkers: Gene hly, acetoin, and listeriolysin O protein. J Electroanal Chem (Lausanne) 2022. [DOI: 10.1016/j.jelechem.2021.115975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
38
|
Benton JT, Bayly-Jones C. Challenges and approaches to studying pore-forming proteins. Biochem Soc Trans 2021; 49:2749-2765. [PMID: 34747994 PMCID: PMC8892993 DOI: 10.1042/bst20210706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/19/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023]
Abstract
Pore-forming proteins (PFPs) are a broad class of molecules that comprise various families, structural folds, and assembly pathways. In nature, PFPs are most often deployed by their host organisms to defend against other organisms. In humans, this is apparent in the immune system, where several immune effectors possess pore-forming activity. Furthermore, applications of PFPs are found in next-generation low-cost DNA sequencing, agricultural crop protection, pest control, and biosensing. The advent of cryoEM has propelled the field forward. Nevertheless, significant challenges and knowledge-gaps remain. Overcoming these challenges is particularly important for the development of custom, purpose-engineered PFPs with novel or desired properties. Emerging single-molecule techniques and methods are helping to address these unanswered questions. Here we review the current challenges, problems, and approaches to studying PFPs.
Collapse
Affiliation(s)
- Joshua T. Benton
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Charles Bayly-Jones
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
39
|
Quereda JJ, Morón-García A, Palacios-Gorba C, Dessaux C, García-del Portillo F, Pucciarelli MG, Ortega AD. Pathogenicity and virulence of Listeria monocytogenes: A trip from environmental to medical microbiology. Virulence 2021; 12:2509-2545. [PMID: 34612177 PMCID: PMC8496543 DOI: 10.1080/21505594.2021.1975526] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 01/02/2023] Open
Abstract
Listeria monocytogenes is a saprophytic gram-positive bacterium, and an opportunistic foodborne pathogen that can produce listeriosis in humans and animals. It has evolved an exceptional ability to adapt to stress conditions encountered in different environments, resulting in a ubiquitous distribution. Because some food preservation methods and disinfection protocols in food-processing environments cannot efficiently prevent contaminations, L. monocytogenes constitutes a threat to human health and a challenge to food safety. In the host, Listeria colonizes the gastrointestinal tract, crosses the intestinal barrier, and disseminates through the blood to target organs. In immunocompromised individuals, the elderly, and pregnant women, the pathogen can cross the blood-brain and placental barriers, leading to neurolisteriosis and materno-fetal listeriosis. Molecular and cell biology studies of infection have proven L. monocytogenes to be a versatile pathogen that deploys unique strategies to invade different cell types, survive and move inside the eukaryotic host cell, and spread from cell to cell. Here, we present the multifaceted Listeria life cycle from a comprehensive perspective. We discuss genetic features of pathogenic Listeria species, analyze factors involved in food contamination, and review bacterial strategies to tolerate stresses encountered both during food processing and along the host's gastrointestinal tract. Then we dissect host-pathogen interactions underlying listerial pathogenesis in mammals from a cell biology and systemic point of view. Finally, we summarize the epidemiology, pathophysiology, and clinical features of listeriosis in humans and animals. This work aims to gather information from different fields crucial for a comprehensive understanding of the pathogenesis of L. monocytogenes.
Collapse
Affiliation(s)
- Juan J. Quereda
- Departamento de Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. Valencia, Spain
| | - Alvaro Morón-García
- Departamento de Biología Celular. Facultad de Ciencias Biológicas, Universidad Complutense de Madrid. Madrid, Spain
| | - Carla Palacios-Gorba
- Departamento de Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities. Valencia, Spain
| | - Charlotte Dessaux
- Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología (CNB)- Consejo Superior De Investigaciones Científicas (CSIC), Madrid, Spain
| | - Francisco García-del Portillo
- Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología (CNB)- Consejo Superior De Investigaciones Científicas (CSIC), Madrid, Spain
| | - M. Graciela Pucciarelli
- Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología (CNB)- Consejo Superior De Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Biología Molecular ‘Severo Ochoa’. Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid. Madrid, Spain
| | - Alvaro D. Ortega
- Departamento de Biología Celular. Facultad de Ciencias Biológicas, Universidad Complutense de Madrid. Madrid, Spain
- Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología (CNB)- Consejo Superior De Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
40
|
Li S, Xu X, Wei L, Wang L, Lv Q. Acacetin Alleviates Listeria monocytogenes Virulence Both In Vitro and In Vivo via the Inhibition of Listeriolysin O. Foodborne Pathog Dis 2021; 19:115-125. [PMID: 34809484 DOI: 10.1089/fpd.2021.0021] [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: 11/12/2022] Open
Abstract
Listeria monocytogenes is a ubiquitous Gram-positive foodborne pathogen that is responsible for listeriosis in both humans and several animal species. The bacterium secretes a pore-forming cholesterol-dependent cytolysin, listeriolysin O (LLO), a major virulence factor involved in the activation of cellular processes. The ability of LLO to lyse erythrocytes is a measure of LLO activity. We used hemolytic activity assay to screen the LLO inhibitors. Acacetin was found to be an LLO inhibitor, which is a di-hydroxy and mono-methoxy flavone present in various plants, including Black locust, Damiana, and Silver birch. As the features of acacetin are of low toxicity and have less acquired resistance, it comes to a hotspot in drug development. In our study, we report that acacetin antagonized the hemolytic activity of L. monocytogenes culture supernatants and purified LLO by directly interfering with the formation of oligomers without inhibiting the bacterial growth and the expression of LLO. Acacetin also relieved the injury of alveolar epithelial cells by inhibiting LLO activity. Further, acacetin significantly promoted the clearance of L. monocytogenes and alleviated the histopathological damage, thereby raising survival rate, which conferred mice with effective protection against L. monocytogenes infection. Using molecular docking and dynamics simulation, we further proved the mechanism of acacetin antagonizing LLO pore-forming activity by direct binding to the second membrane-inserting helix bundle (HB2) of LLO domain 3. These data suggested that acacetin recedes the virulence of L. monocytogenes both in vivo and in vitro, and this study provided a promising candidate and potential alternative for the prevention and treatment of L. monocytogenes infections.
Collapse
Affiliation(s)
- Shufang Li
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiangzhu Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lijuan Wei
- Laigang Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qianghua Lv
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
41
|
McDougal CE, Morrow ZT, Christopher T, Kim S, Carter D, Stevenson DM, Amador-Noguez D, Miller MJ, Sauer JD. Phagocytes produce prostaglandin E2 in response to cytosolic Listeria monocytogenes. PLoS Pathog 2021; 17:e1009493. [PMID: 34555127 PMCID: PMC8491950 DOI: 10.1371/journal.ppat.1009493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 10/05/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
Listeria monocytogenes is an intracellular bacterium that elicits robust CD8+ T-cell responses. Despite the ongoing development of L. monocytogenes-based platforms as cancer vaccines, our understanding of how L. monocytogenes drives robust CD8+ T-cell responses remains incomplete. One overarching hypothesis is that activation of cytosolic innate pathways is critical for immunity, as strains of L. monocytogenes that are unable to access the cytosol fail to elicit robust CD8+ T-cell responses and in fact inhibit optimal T-cell priming. Counterintuitively, however, activation of known cytosolic pathways, such as the inflammasome and type I IFN, lead to impaired immunity. Conversely, production of prostaglandin E2 (PGE2) downstream of cyclooxygenase-2 (COX-2) is essential for optimal L. monocytogenes T-cell priming. Here, we demonstrate that vacuole-constrained L. monocytogenes elicit reduced PGE2 production compared to wild-type strains in macrophages and dendritic cells ex vivo. In vivo, infection with wild-type L. monocytogenes leads to 10-fold increases in PGE2 production early during infection whereas vacuole-constrained strains fail to induce PGE2 over mock-immunized controls. Mice deficient in COX-2 specifically in Lyz2+ or CD11c+ cells produce less PGE2, suggesting these cell subsets contribute to PGE2 levels in vivo, while depletion of phagocytes with clodronate abolishes PGE2 production completely. Taken together, this work demonstrates that optimal PGE2 production by phagocytes depends on L. monocytogenes access to the cytosol, suggesting that one reason cytosolic access is required to prime CD8+ T-cell responses may be to facilitate production of PGE2.
Collapse
Affiliation(s)
- Courtney E. McDougal
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Wisconsin, United States of America
| | - Zachary T. Morrow
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Tighe Christopher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Seonyoung Kim
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Drake Carter
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - David M. Stevenson
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Daniel Amador-Noguez
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mark J. Miller
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
42
|
Hossain MI, Mizan MFR, Roy PK, Nahar S, Toushik SH, Ashrafudoulla M, Jahid IK, Lee J, Ha SD. Listeria monocytogenes biofilm inhibition on food contact surfaces by application of postbiotics from Lactobacillus curvatus B.67 and Lactobacillus plantarum M.2. Food Res Int 2021; 148:110595. [PMID: 34507740 DOI: 10.1016/j.foodres.2021.110595] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
Owing to their preservative and antimicrobial effects, postbiotics (metabolic byproducts of probiotics) are promising natural components for the food industry. Therefore, the present study aimed to investigate the efficacy of postbiotics collected from isolated Lactobacillus curvatus B.67 and Lactobacillus plantarum M.2 against Listeria monocytogenes pathogens in planktonic cells, motility, and biofilm states. The analysis of the metabolite composition of the postbiotics revealed various organic acids, along with a few well-known bacteriocin-encoding genes with potential antimicrobial effects. Postbiotics maintained their residual antimicrobial activity over the pH range 1-6 but lost all activity at neutral pH (pH 7). Full antimicrobial activity (100%) was observed during heat treatment, even under the autoclaving condition.Minimum inhibitory concentration (MICs) of L. curvatus B.67 and L. plantarum M.2 against L. monocytogenes were 80 and 70 mg/mL, respectively. However, four sub-MICs of the postbiotics (1/2, 1/4, 1/8, and 1/16 MIC) were tested for inhibition efficacy against L. monocytogenes during different experiment in this study. Swimming motility, biofilm formation, and expression levels of target genes related to biofilm formation, virulence, and quorum-sensing were significantly inhibited with increasing postbiotics concentration. Postbiotics from L. plantarum M.2 exhibited a higher inhibitory effect than the postbiotics from L. curvatus B.67. Nonetheless, both these postbiotics from Lactobacillus spp. could be used as effective bio-interventions for controlling L. monocytogenes biofilm in the food industry.
Collapse
Affiliation(s)
- Md Iqbal Hossain
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Md Furkanur Rahaman Mizan
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Pantu Kumar Roy
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Shamsun Nahar
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Sazzad Hossen Toushik
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Md Ashrafudoulla
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Iqbal Kabir Jahid
- Department of Microbiology, Jashore University of Science and Technology, Bangladesh
| | - Jihyun Lee
- Department of Food Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Sang-Do Ha
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea.
| |
Collapse
|
43
|
Ilangumaran Ponmalar I, Sarangi NK, Basu JK, Ayappa KG. Pore Forming Protein Induced Biomembrane Reorganization and Dynamics: A Focused Review. Front Mol Biosci 2021; 8:737561. [PMID: 34568431 PMCID: PMC8459938 DOI: 10.3389/fmolb.2021.737561] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 07/30/2021] [Indexed: 11/13/2022] Open
Abstract
Pore forming proteins are a broad class of pathogenic proteins secreted by organisms as virulence factors due to their ability to form pores on the target cell membrane. Bacterial pore forming toxins (PFTs) belong to a subclass of pore forming proteins widely implicated in bacterial infections. Although the action of PFTs on target cells have been widely investigated, the underlying membrane response of lipids during membrane binding and pore formation has received less attention. With the advent of superresolution microscopy as well as the ability to carry out molecular dynamics (MD) simulations of the large protein membrane assemblies, novel microscopic insights on the pore forming mechanism have emerged over the last decade. In this review, we focus primarily on results collated in our laboratory which probe dynamic lipid reorganization induced in the plasma membrane during various stages of pore formation by two archetypal bacterial PFTs, cytolysin A (ClyA), an α-toxin and listeriolysin O (LLO), a β-toxin. The extent of lipid perturbation is dependent on both the secondary structure of the membrane inserted motifs of pore complex as well as the topological variations of the pore complex. Using confocal and superresolution stimulated emission depletion (STED) fluorescence correlation spectroscopy (FCS) and MD simulations, lipid diffusion, cholesterol reorganization and deviations from Brownian diffusion are correlated with the oligomeric state of the membrane bound protein as well as the underlying membrane composition. Deviations from free diffusion are typically observed at length scales below ∼130 nm to reveal the presence of local dynamical heterogeneities that emerge at the nanoscale-driven in part by preferential protein binding to cholesterol and domains present in the lipid membrane. Interrogating the lipid dynamics at the nanoscale allows us further differentiate between binding and pore formation of β- and α-PFTs to specific domains in the membrane. The molecular insights gained from the intricate coupling that occurs between proteins and membrane lipids and receptors during pore formation are expected to improve our understanding of the virulent action of PFTs.
Collapse
Affiliation(s)
| | - Nirod K. Sarangi
- School of Chemical Science, Dublin City University, Dublin, Ireland
| | - Jaydeep K. Basu
- Department of Physics, Indian Institute of Science, Bangalore, India
| | - K. Ganapathy Ayappa
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
44
|
Walch P, Selkrig J, Knodler LA, Rettel M, Stein F, Fernandez K, Viéitez C, Potel CM, Scholzen K, Geyer M, Rottner K, Steele-Mortimer O, Savitski MM, Holden DW, Typas A. Global mapping of Salmonella enterica-host protein-protein interactions during infection. Cell Host Microbe 2021; 29:1316-1332.e12. [PMID: 34237247 PMCID: PMC8561747 DOI: 10.1016/j.chom.2021.06.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 02/24/2021] [Accepted: 05/21/2021] [Indexed: 11/16/2022]
Abstract
Intracellular bacterial pathogens inject effector proteins to hijack host cellular processes and promote their survival and proliferation. To systematically map effector-host protein-protein interactions (PPIs) during infection, we generated a library of 32 Salmonella enterica serovar Typhimurium (STm) strains expressing chromosomally encoded affinity-tagged effectors and quantified PPIs in macrophages and epithelial cells. We identified 446 effector-host PPIs, 25 of which were previously described, and validated 13 by reciprocal co-immunoprecipitation. While effectors converged on the same host cellular processes, most had multiple targets, which often differed between cell types. We demonstrate that SseJ, SseL, and SifA modulate cholesterol accumulation at the Salmonella-containing vacuole (SCV) partially via the cholesterol transporter Niemann-Pick C1 protein. PipB recruits the organelle contact site protein PDZD8 to the SCV, and SteC promotes actin bundling by phosphorylating formin-like proteins. This study provides a method for probing host-pathogen PPIs during infection and a resource for interrogating STm effector mechanisms.
Collapse
Affiliation(s)
- Philipp Walch
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Joel Selkrig
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Leigh A Knodler
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, USA; Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Mandy Rettel
- EMBL, Proteomics Core Facility, Heidelberg, Germany
| | - Frank Stein
- EMBL, Proteomics Core Facility, Heidelberg, Germany
| | - Keith Fernandez
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Cristina Viéitez
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; EMBL European Bioinformatics Institute, (EMBL-EBI), Hinxton, UK
| | - Clément M Potel
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Karoline Scholzen
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Bonn, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, TU Braunschweig, Braunschweig, Germany; Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Olivia Steele-Mortimer
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Mikhail M Savitski
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; EMBL, Proteomics Core Facility, Heidelberg, Germany
| | - David W Holden
- MRC Centre for Molecular Bacteriology and Infection, Imperial College, London, UK
| | - Athanasios Typas
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany.
| |
Collapse
|
45
|
Li M, Carpenter CE, Broadbent JR. Organic Acid Exposure Enhances Virulence in Some Listeria monocytogenes Strains Using the Galleria mellonella Infection Model. Front Microbiol 2021; 12:675241. [PMID: 34295317 PMCID: PMC8290484 DOI: 10.3389/fmicb.2021.675241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/14/2021] [Indexed: 01/01/2023] Open
Abstract
Prior research has suggested that the use of organic acids in the food industry may unintentionally enhance pathogenicity of Listeria monocytogenes strain N1-227 and R2-499. This study explored the connection between habituation to L-lactic acid or acetic acid and virulence in L. monocytogenes strains N1-227 and R2-499 using selected gene expression analysis and the in vivo Galleria mellonella wax worm model for infection. Expression of transcription factors (sigB and prfA) and genes related to acid resistance (gadD2, gadD3, and arcA) and bile resistance (bsh and bilE) or to virulence (inlA, inlB, hly, plcA, plcB, uhpT, and actA) was investigated by quantitative real-time PCR (qRT-PCR), while in vivo virulence was assessed by following the lethal time to 50% population mortality (LT50) of G. mellonella larvae after injection of untreated and habituated L. monocytogenes. Twenty minutes of habituation to the organic acids at pH 6.0 significantly increased expression of key acid and bile stress response genes in both strains, while expression of virulence genes was strain-dependent. The expression of transcription factor sigB was strain-dependent and there was no significant change in the expression of transcription factor prfA in both strains. Habituation to acid increased virulence of both strains as evidenced by decreased LT50 of G. mellonella larvae injected with Listeria habituated to either acid. In summary, habituation of both L. monocytogenes strains to organic acids up-regulated expression of several stress and virulence genes and concurrently increased virulence as measured using the G. mellonella model.
Collapse
Affiliation(s)
- Minghao Li
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| | - Charles E Carpenter
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| | - Jeff R Broadbent
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| |
Collapse
|
46
|
Ilangumaran Ponmalar I, Ayappa KG, Basu JK. Bacterial protein listeriolysin O induces nonmonotonic dynamics because of lipid ejection and crowding. Biophys J 2021; 120:3040-3049. [PMID: 34214525 DOI: 10.1016/j.bpj.2021.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 01/15/2023] Open
Abstract
Membrane-bound protein complexes involving pore forming toxins (PFTs) released by virulent bacteria are known to form transmembrane pores leading to host cell lysis. Developing alternative strategies against PFT mediated bacterial virulence factors requires an understanding of the cellular membrane response. However, membrane disruption and related lipid reorganization events during attack by PFTs remain largely unexplored. We report counterintuitive and nonmonotonic variations in lipid diffusion, measured using confocal fluorescence correlation spectroscopy, due to interplay of lipid ejection and crowding by membrane-bound oligomers of a prototypical cholesterol-dependent cytolysin, listeriolysin O (LLO). The observed dynamical crossover is correlated with concentration dependent transitions of LLO oligomeric state populations from rings to arc-like pore complexes, predicted using a proposed two-state free area-based diffusion model. At low PFT concentrations, a hitherto unexplored regime of increased lipid diffusivity is attributed to lipid ejection events because of a preponderance of ring-like pore states. At higher protein concentrations in which membrane-inserted arc-like pores dominate, lipid ejection is less efficient and the ensuing crowding results in a lowering of lipid diffusion. These variations in lipid dynamics are corroborated by macroscopic rheological response measurements of PFT bound vesicles. Our study correlates PFT oligomeric state transitions, membrane remodeling, and mechanical property variations, providing unique insights into the pore forming mechanisms of cholesterol-dependent cytolysins.
Collapse
Affiliation(s)
| | - K Ganapathy Ayappa
- Center for BioSystems Science and Engineering Bengaluru, India; Department of Chemical Engineering Bengaluru, India.
| | - Jaydeep K Basu
- Department of Physics, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
47
|
Jones K, Zhu J, Jenkinson CB, Kim DW, Pfeifer MA, Khang CH. Disruption of the Interfacial Membrane Leads to Magnaporthe oryzae Effector Re-location and Lifestyle Switch During Rice Blast Disease. Front Cell Dev Biol 2021; 9:681734. [PMID: 34222251 PMCID: PMC8248803 DOI: 10.3389/fcell.2021.681734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/13/2021] [Indexed: 11/13/2022] Open
Abstract
To cause the devastating rice blast disease, the hemibiotrophic fungus Magnaporthe oryzae produces invasive hyphae (IH) that are enclosed in a plant-derived interfacial membrane, known as the extra-invasive hyphal membrane (EIHM), in living rice cells. Little is known about when the EIHM is disrupted and how the disruption contributes to blast disease. Here we show that the disruption of the EIHM correlates with the hyphal growth stage in first-invaded susceptible rice cells. Our approach utilized GFP that was secreted from IH as an EIHM integrity reporter. Secreted GFP (sec-GFP) accumulated in the EIHM compartment but appeared in the host cytoplasm when the integrity of the EIHM was compromised. Live-cell imaging coupled with sec-GFP and various fluorescent reporters revealed that the loss of EIHM integrity preceded shrinkage and eventual rupture of the rice vacuole. The vacuole rupture coincided with host cell death, which was limited to the invaded cell with presumed closure of plasmodesmata. We report that EIHM disruption and host cell death are landmarks that delineate three distinct infection phases (early biotrophic, late biotrophic, and transient necrotrophic phases) within the first-invaded cell before reestablishment of biotrophy in second-invaded cells. M. oryzae effectors exhibited infection phase-specific localizations, including entry of the apoplastic effector Bas4 into the host cytoplasm through the disrupted EIHM during the late biotrophic phase. Understanding how infection phase-specific cellular dynamics are regulated and linked to host susceptibility will offer potential targets that can be exploited to control blast disease.
Collapse
Affiliation(s)
- Kiersun Jones
- Department of Plant Biology, University of Georgia, Athens, GA, United States
| | - Jie Zhu
- Department of Plant Biology, University of Georgia, Athens, GA, United States
| | - Cory B Jenkinson
- Department of Plant Biology, University of Georgia, Athens, GA, United States
| | - Dong Won Kim
- Department of Plant Biology, University of Georgia, Athens, GA, United States
| | - Mariel A Pfeifer
- Department of Plant Biology, University of Georgia, Athens, GA, United States
| | - Chang Hyun Khang
- Department of Plant Biology, University of Georgia, Athens, GA, United States
| |
Collapse
|
48
|
Lopez Chiloeches M, Bergonzini A, Frisan T. Bacterial Toxins Are a Never-Ending Source of Surprises: From Natural Born Killers to Negotiators. Toxins (Basel) 2021; 13:426. [PMID: 34204481 PMCID: PMC8235270 DOI: 10.3390/toxins13060426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
The idea that bacterial toxins are not only killers but also execute more sophisticated roles during bacteria-host interactions by acting as negotiators has been highlighted in the past decades. Depending on the toxin, its cellular target and mode of action, the final regulatory outcome can be different. In this review, we have focused on two families of bacterial toxins: genotoxins and pore-forming toxins, which have different modes of action but share the ability to modulate the host's immune responses, independently of their capacity to directly kill immune cells. We have addressed their immuno-suppressive effects with the perspective that these may help bacteria to avoid clearance by the host's immune response and, concomitantly, limit detrimental immunopathology. These are optimal conditions for the establishment of a persistent infection, eventually promoting asymptomatic carriers. This immunomodulatory effect can be achieved with different strategies such as suppression of pro-inflammatory cytokines, re-polarization of the immune response from a pro-inflammatory to a tolerogenic state, and bacterial fitness modulation to favour tissue colonization while preventing bacteraemia. An imbalance in each of those effects can lead to disease due to either uncontrolled bacterial proliferation/invasion, immunopathology, or both.
Collapse
Affiliation(s)
| | | | - Teresa Frisan
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden; (M.L.C.); (A.B.)
| |
Collapse
|
49
|
Wu M, Brown AC. Applications of Catechins in the Treatment of Bacterial Infections. Pathogens 2021; 10:546. [PMID: 34062722 PMCID: PMC8147231 DOI: 10.3390/pathogens10050546] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 01/08/2023] Open
Abstract
Tea is the second most commonly consumed beverage worldwide. Along with its aromatic and delicate flavors that make it an enjoyable beverage, studies report numerous health advantages in tea consumption, including applications in antimicrobial therapy. The antimicrobial properties of tea are related to catechin and its derivatives, which are natural flavonoids that are abundant in tea. Increasing evidence from in vitro studies demonstrated antimicrobial effects of catechins on both gram-positive and gram-negative bacteria, and proposed direct and indirect therapeutic mechanisms. Additionally, catechins were reported to be effective anti-virulence agents. Furthermore, a number of studies presented evidence that catechins display synergistic effects with certain antibiotics, thus potentiating the activity of antibiotics in resistant bacteria. Despite their numerous beneficial properties, catechins face many challenges in their development as therapeutic agents, including poor absorption, low bioavailability, and rapid degradation. The introduction of nanobiotechnology provides target-based and stable delivery, which enhances catechin bioavailability and optimizes drug efficacy. As further research continues to focus on overcoming the unresolved challenges, catechins are likely to see additional promising applications in our continual fight against bacterial infections.
Collapse
Affiliation(s)
| | - Angela C. Brown
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA 18015, USA;
| |
Collapse
|
50
|
Parussolo L, Sfaciotte RAP, Dalmina KA, Melo FD, Costa UMDA, Ferraz SM. Detection of virulence genes and antimicrobial susceptibility profile of Listeria monocytogenes isolates recovered from artisanal cheese produced in the Southern region of Brazil. AN ACAD BRAS CIENC 2021; 93:e20190200. [PMID: 33950134 DOI: 10.1590/0001-3765202120190200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/13/2019] [Indexed: 11/22/2022] Open
Abstract
Listeria monocytogenes is an opportunistic pathogen that causes listeriosis, a foodborne disease with low incidence but with high mortality rate in humans. This microorganism has been recovered from several dairy products, especially those produced with raw milk. The objective of this work was to investigate the presence of virulence genes, and also to define the antimicrobial susceptibility profile of L. monocytogenes isolates recovered from serrano artisanal cheese produced in Southern region of Brazil. Nine strains of L. monocytogenes (serotypes 1/2b and 4b) were evaluated through PCR to detect the presence of the virulence genes hly, inlA, inlC, inlJ, actA, plcB and iap, while antimicrobial susceptibility profile was determined via disk diffusion method. All strains exhibited the presence of the genes hly and plcB, whereas the other genes (iap, actA, inlA, inlC and inlJ) were only detected in eight strains. We verified that all strains were resistant to at least one antimicrobial agent and three of them showed multidrug resistance. These findings demonstrated the serrano artisanal cheese offers risks to consumers' health and point to a need of adaptations and monitoring of manufacturing process of this food, in order to prevent the dissemination of L. monocytogenes.
Collapse
Affiliation(s)
- Leandro Parussolo
- Universidade do Estado de Santa Catarina, Centro de Diagnóstico Microbiológico Animal, Centro de Ciências Agroveterinárias, Av. Luiz de Camões, 2090, Bairro Conta Dinheiro, 88520-000 Lages, SC, Brazil.,Instituto Federal de Santa Catarina, Av. Mauro Ramos, 950, Centro, 88020-300 Florianópolis, SC, Brazil
| | - Ricardo Antônio P Sfaciotte
- Universidade do Estado de Santa Catarina, Centro de Diagnóstico Microbiológico Animal, Centro de Ciências Agroveterinárias, Av. Luiz de Camões, 2090, Bairro Conta Dinheiro, 88520-000 Lages, SC, Brazil
| | - Karine Andrezza Dalmina
- Universidade do Estado de Santa Catarina, Centro de Diagnóstico Microbiológico Animal, Centro de Ciências Agroveterinárias, Av. Luiz de Camões, 2090, Bairro Conta Dinheiro, 88520-000 Lages, SC, Brazil
| | - Fernanda Danielle Melo
- Universidade do Estado de Santa Catarina, Centro de Diagnóstico Microbiológico Animal, Centro de Ciências Agroveterinárias, Av. Luiz de Camões, 2090, Bairro Conta Dinheiro, 88520-000 Lages, SC, Brazil
| | - Ubirajara M DA Costa
- Universidade do Estado de Santa Catarina, Centro de Diagnóstico Microbiológico Animal, Centro de Ciências Agroveterinárias, Av. Luiz de Camões, 2090, Bairro Conta Dinheiro, 88520-000 Lages, SC, Brazil
| | - Sandra Maria Ferraz
- Universidade do Estado de Santa Catarina, Centro de Diagnóstico Microbiológico Animal, Centro de Ciências Agroveterinárias, Av. Luiz de Camões, 2090, Bairro Conta Dinheiro, 88520-000 Lages, SC, Brazil
| |
Collapse
|