1
|
Stevens MJA, Wambui J, Corti S, Nüesch-Inderbinen M, Stephan R. Weak phospholipase-positive Listeria monocytogenes: a challenge for the diagnostic laboratory. Diagn Microbiol Infect Dis 2025; 112:116802. [PMID: 40120294 DOI: 10.1016/j.diagmicrobio.2025.116802] [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: 02/26/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Listeria monocytogenes isolates from environmental samples of a food producing company harboured a C424G mutation in the prfA gene that results in a G141A substitution in PrfA. This mutation results in strongly decreased phospholipase activity and is therefore challenging for culture-based detection of L. monocytogenes and may even lead to misidentification.
Collapse
Affiliation(s)
- Marc J A Stevens
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Joseph Wambui
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Sabrina Corti
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | | | - Roger Stephan
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Alejandro-Navarreto X, Cahoon LA, Freitag NE. Characterization of the Listeria monocytogenes PieRS regulon distinguishes the function of the critical secretion chaperone PrsA2 from other regulon members. Infect Immun 2025:e0035724. [PMID: 40422080 DOI: 10.1128/iai.00357-24] [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: 08/17/2024] [Accepted: 04/25/2025] [Indexed: 05/28/2025] Open
Abstract
Listeria monocytogenes (Lm) is a gram-positive pathogen that is widespread throughout the environment and known for its ability to infect mammalian hosts following the ingestion of contaminated food. Lm uses a variety of mechanisms to survive challenging conditions experienced both during life in the outside environment and inside of the infected host. We recently described a novel two-component signaling system known as PieRS that regulates the secretion of the chaperone PrsA2, which is essential for bacterial virulence, as well as its related homolog PrsA1 and a variety of gene products of unknown function. Here, we examine the roles of the less characterized PieRS-regulated gene products and contrast their functions with PrsA2 in terms of bacterial survival under stress conditions and virulence in mice. Characterization of targeted in-frame deletion mutants of PieRS regulon members indicates-in contrast to prsA2 mutants-minimal contributions to stress survival and bacterial virulence. Modest contributions of select regulon members were associated with Lm colonization of the gastrointestinal tract. The PieRS regulon thus consists of gene products that contribute to Lm physiology in ways that are clearly distinct from PrsA2 and the chaperone's essential function for both stress survival and bacterial virulence.
Collapse
Affiliation(s)
| | - Laty A Cahoon
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nancy E Freitag
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
3
|
Pham HG, Tran KN, Gomelsky L, Roy T, Gigley JP, Gomelsky M. Robust Inducible Gene Expression in Intracellular Listeria monocytogenes In Vivo. ACS Synth Biol 2025; 14:1397-1404. [PMID: 40277175 DOI: 10.1021/acssynbio.5c00131] [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] [Indexed: 04/26/2025]
Abstract
Attenuated strains of the intracellular pathogen Listeria monocytogenes can deliver genetically encoded payloads inside tumor cells. L. monocytogenes preferentially accumulates and propagates in immune-suppressed tumor microenvironments. To maximize the payload impact in tumors and minimize damage to healthy tissues, it is desirable to induce payload synthesis when bacteria are eliminated from the healthy tissues but are grown to high numbers intratumorally. Here, we have engineered a tightly controlled gene expression system for intracellular L. monocytogenes inducible with a cumin derivative, cumate. Upon cumate addition, expression of a reporter gene is increased in L. monocytogenes growing in vitro by 80-fold and in intracellular L. monocytogenes in murine tumors by 75-fold. This study demonstrates the feasibility of activating gene expression in intracellular bacteria in live animals using an edible inducer. The system is expected to enhance the efficacy and safety of the attenuated L. monocytogenes strains as antitumor payload delivery bacterial drones.
Collapse
Affiliation(s)
- Huong Giang Pham
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming 82071, United States
| | - Kiet N Tran
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming 82071, United States
| | - Larissa Gomelsky
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming 82071, United States
| | - Tathagato Roy
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming 82071, United States
| | - Jason P Gigley
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming 82071, United States
| | - Mark Gomelsky
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming 82071, United States
| |
Collapse
|
4
|
Engelgeh T, Wamp S, Rothe P, Herrmann J, Fischer MA, Müller R, Halbedel S. ClpP2 proteasomes and SpxA1 determine Listeria monocytogenes tartrolon B hyper-resistance. PLoS Genet 2025; 21:e1011621. [PMID: 40184427 PMCID: PMC11970672 DOI: 10.1371/journal.pgen.1011621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 02/13/2025] [Indexed: 04/06/2025] Open
Abstract
The foodborne bacterium Listeria monocytogenes is transmitted to humans from various environmental sources through consumption of contaminated plant and animal-based food. L. monocytogenes uses ATP-binding cassette (ABC)-type drug transporters to resist antimicrobial compounds produced by competitors co-residing in its environmental reservoirs. We have shown previously that the TimAB transporter confers resistance of L. monocytogenes to tartrolon B, a boron containing macrodiolide produced by myxo- and proteobacterial species. Tartrolon B acts as a potassium ionophore and is sensed by TimR, the transcriptional repressor of timABR operon. We here have isolated tartrolon B resistant suppressor mutations outside the timABR locus. These mutations inactivated the clpP2 gene, which encodes the main proteolytic component of house-keeping Clp proteases. Deletion of clpP2 impaired growth and virulence but caused tartrolon B hyper-resistance. This phenotype was timAB-dependent, but neither production nor degradation of TimAB was affected upon clpP2 inactivation. Combinatorial deletions of the genes encoding the three Clp ATPases showed that ClpCP2 and ClpXP2 proteasomes jointly promote tartrolon B hyper-resistance. Genetic follow-up experiments identified the ClpP2 substrate and transcription factor SpxA1 and its protease adaptor YjbH as further tartrolon B resistance determinants. SpxA1 activates transcription of the cydABCD operon encoding cytochrome oxidase and in accordance with this transposon mutants with impaired cytochrome oxidase function were depleted from a transposon mutant library during tartrolon B exposure. Our work demonstrates novel roles of Clp proteasomes, SpxA1 and cytochrome oxidase CydAB in the resistance against compounds dissipating transmembrane ion gradients and helps to better understand the genetic and chemical basis of the manifold ecological interactions of an important human pathogen in its natural ecologic niches.
Collapse
Affiliation(s)
- Tim Engelgeh
- FG11 Division of Enteropathogenic bacteria and Legionella, Robert Koch Institute, Wernigerode, Germany
| | - Sabrina Wamp
- FG11 Division of Enteropathogenic bacteria and Legionella, Robert Koch Institute, Wernigerode, Germany
| | - Patricia Rothe
- FG11 Division of Enteropathogenic bacteria and Legionella, Robert Koch Institute, Wernigerode, Germany
| | - Jennifer Herrmann
- Department of Microbial Natural Products, Helmholtz Centre for Infection Research and Department of Pharmaceutical Biotechnology, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) and Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Martin A. Fischer
- FG13 Division of Nosocomial Pathogens and Antibiotic Resistances, Robert Koch Institute, Wernigerode, Germany
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Centre for Infection Research and Department of Pharmaceutical Biotechnology, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) and Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Sven Halbedel
- FG11 Division of Enteropathogenic bacteria and Legionella, Robert Koch Institute, Wernigerode, Germany
- Institute for Medical Microbiology and Hospital Hygiene, Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
5
|
Li W, Huang C, Yuan W, Xu T, Shan L, Xia W, Ren Y, Zhang Z, Chen J. Effects of the histone-like protein on biofilm formation and pathogenicity of Listeria monocytogenes. Int J Biol Macromol 2025; 304:140908. [PMID: 39938827 DOI: 10.1016/j.ijbiomac.2025.140908] [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: 12/04/2024] [Revised: 01/20/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025]
Abstract
Histone-like protein HU is essential for DNA recombination, repair, and transcriptional regulation in bacteria. However, the physiological roles of HU proteins in Listeria monocytogenes (LmHU) remain unexplored. Given the significant biofilm-forming ability of this foodborne pathogen and its associated cross-contamination risks, identifying novel control targets is critical. LmHU, as the sole double-stranded DNA-binding protein in L. monocytogenes, is a promising candidate. This study systematically explored its contributions to biofilm formation, motility, and the regulation of virulence factors. The results indicated that high levels of LmHU in vivo promoted cell cohesion, leading to a chain-like structure among L. monocytogenes. Additionally, LmHU could be secreted into the biofilm matrix, reinforcing the structure by interacting with extracellular polymeric substances. However, elevated LmHU levels inhibited bacterial motility, flagellar synthesis, and host invasion in Caco-2 cells. RNA-seq analysis revealed 374 differentially expressed genes in the Lmhu mutant relative to the wild-type strain, supporting these findings. Further enrichment analysis and validation experiments suggested that Lmhu overexpression impaired trehalose utilization. These results indicate that LmHU may serve as a potential target for novel disinfectants or therapies to reduce risks associated with L. monocytogenes in food safety and public health.
Collapse
Affiliation(s)
- Wenqian Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Can Huang
- School of Public Health, Qingdao University, Qingdao 266000, China
| | - Wanjuan Yuan
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Tiangang Xu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Lei Shan
- Department of Food Science and Nutrition, University of Minnesota, Saint Paul MN55108, USA
| | - Wanpeng Xia
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ying Ren
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Zhenfeng Zhang
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Jingyu Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; China Agricultural University-Sichuan Advanced Agricultural & Industrial Institute, Chengdu 611430, China.
| |
Collapse
|
6
|
Radhakrishnan P, Theriot JA. Listeria monocytogenes cell-to-cell spread bypasses nutrient limitation for replicating intracellular bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.635960. [PMID: 39975404 PMCID: PMC11838505 DOI: 10.1101/2025.01.31.635960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Listeria monocytogenes is an intracellular bacterial pathogen that obtains nutrients from the mammalian host cell to fuel its replication in cytosol. Sparse infection of epithelial monolayers by L. monocytogenes results in the formation of distinct infectious foci, where each focus originates from the initial infection of a single host cell followed by multiple rounds of active bacterial cell-to-cell spread into neighboring host cells in the monolayer. We used time-lapse microscopy to measure changes in bacterial growth rate in individual foci over time and found that intracellular bacteria initially replicate exponentially, but then bacterial growth rate slows later in infection, particularly in the center of the infectious focus. We found that the intracellular replication rate of L. monocytogenes is measurably decreased by limiting host cell glucose availability, by decreasing the rate of intracellular bacterial oligopeptide import, and, most interestingly, by alterations in host cell junctional proteins that limit bacterial spread into neighboring cells without directly affecting bacterial growth or metabolism. By measuring the carrying capacity of individual host cells, we found that the nutritional density of cytoplasm is comparable to rich medium. Taken together, our results indicate that the rate of intracellular L. monocytogenes replication is governed by a balance of the rate of nutrient depletion by the bacteria, the rate of nutrient replenishment by the metabolically active host cells, and the rate of bacterial cell-to-cell spread which enables the bacteria to seek out "greener pastures" before nutrient availability in a single host cell becomes limiting.
Collapse
Affiliation(s)
- Prathima Radhakrishnan
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195-1800
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195-1800
| |
Collapse
|
7
|
Lavergne M, Schaerer R, De Grandis S, Bouheraoua S, Adenuga O, Muralt T, Schaerer T, Chèvre L, Failla A, Matthey P, Stumpe M, Kressler D, Mantel PY, Walch M. Executioner caspases degrade essential mediators of pathogen-host interactions to inhibit growth of intracellular Listeria monocytogenes. Cell Death Dis 2025; 16:55. [PMID: 39885151 PMCID: PMC11782612 DOI: 10.1038/s41419-025-07365-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/20/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Cell death mediated by executioner caspases is essential during organ development and for organismal homeostasis. The mechanistic role of activated executioner caspases in antibacterial defense during infections with intracellular bacteria, such as Listeria monocytogenes, remains elusive. Cell death upon intracellular bacterial infections is considered altruistic to deprive the pathogens of their protective niche. To establish infections in a human host, Listeria monocytogenes deploy virulence mediators, including membranolytic listeriolysin O (LLO) and the invasion associated protein p60 (Iap), allowing phagosomal escape, intracellular replication and cell-to-cell spread. Here, by means of chemical and genetical modifications, we show that the executioner caspases-3 and -7 efficiently inhibit growth of intracellular Listeria monocytogenes in host cells. Comprehensive proteomics revealed multiple caspase-3 substrates in the Listeria secretome, including LLO, Iap and various other proteins crucially involved in pathogen-host interactions. Listeria secreting caspase-uncleavable LLO or Iap gained significant growth advantage in epithelial cells. With that, we uncovered an underappreciated defense barrier and a non-canonical role of executioner caspases to degrade virulence mediators, thus impairing intracellular Listeria growth.
Collapse
Affiliation(s)
- Marilyne Lavergne
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Raffael Schaerer
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Sara De Grandis
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Safaa Bouheraoua
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Oluwadamilola Adenuga
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Tanja Muralt
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Tiffany Schaerer
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Léa Chèvre
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Alessandro Failla
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Patricia Matthey
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Michael Stumpe
- Faculty of Science and Medicine, Department of Biology, Metabolomics and Proteomics Platform, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Dieter Kressler
- Faculty of Science and Medicine, Department of Biology, Metabolomics and Proteomics Platform, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Pierre-Yves Mantel
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), CH-7265, Davos Wolfgang, Switzerland
| | - Michael Walch
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland.
| |
Collapse
|
8
|
Mandinyenya T, Wambui J, Muchaamba F, Stevens MJA, Tasara T. Nisin resistance is increased through GtcA mutation induced loss of cell wall teichoic acid N-acetylglucosamine modifications in Listeria monocytogenes. Int J Food Microbiol 2025; 428:110954. [PMID: 39566380 DOI: 10.1016/j.ijfoodmicro.2024.110954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/04/2024] [Accepted: 10/19/2024] [Indexed: 11/22/2024]
Abstract
Nisin resistance development is one of food safety challenges posed by Listeria monocytogenes, an important foodborne pathogen that causes human listeriosis. The GtcA flippase enzyme is functionally crucial in two separate pathways that glycosylate cell envelope wall teichoic acids (WTA) with N-acetylglucosamine (NAG) and lipoteichoic acids (LTA) with galactose, respectively. This study investigated phenotypic roles and molecular mechanisms underlying GtcA involvement in L. monocytogenes nisin resistance. A GtcAA65V mutation was linked with increased nisin resistance in a food processing environment associated L. monocytogenes strain. Examination of nisin stress survival and growth phenotypes among L. monocytogenes gtcA mutants in different genetic backgrounds showed that GtcA function promoted sensitivity and loss of its function through genetic deletion (ΔgtcA) and a natural GtcAA65V mutation increased nisin resistance. Individual contributions of GtcA WTA NAG and LTA galactose glycosylation functions to nisin resistance modulation were examined through nisin sensitivity analysis of genetic deletion mutants and L. monocytogenes strains complemented using functionally altered GtcA mutants. This revealed WTA NAG glycosylation to be the main functional mechanism that determines GtcA dependent nisin phenotypic sensitization. An examination for mechanisms underlying GtcA involvement in nisin sensitivity revealed that the loss of GtcA function induces changes in the cell envelope carbohydrate composition profiles reducing cell surface hydrophobicity. Overall, our results showed that cell envelope WTA NAG glycosylation promotes nisin susceptibility through facilitation of hydrophobic interactions between nisin and the Listeria cell envelope. There may be practical implications from our observations since nisin resistance could be gained in food associated L. monocytogenes strains that develop phage resistance through acquisition of mutations in genes that cause loss of cell envelope WTA NAG modifications.
Collapse
Affiliation(s)
- Toruvandepi Mandinyenya
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 272, 8057 Zurich, Switzerland
| | - Joseph Wambui
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 272, 8057 Zurich, Switzerland
| | - Francis Muchaamba
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 272, 8057 Zurich, Switzerland
| | - Marc J A Stevens
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 272, 8057 Zurich, Switzerland
| | - Taurai Tasara
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 272, 8057 Zurich, Switzerland.
| |
Collapse
|
9
|
Hafner L, Gadin E, Huang L, Frouin A, Laporte F, Gaultier C, Vieira A, Maudet C, Varet H, Moura A, Bracq-Dieye H, Tessaud-Rita N, Maury M, Dazas M, Legendre R, Gastineau P, Tsai YH, Coppée JY, Charlier C, Patin E, Chikhi R, Rocha EPC, Leclercq A, Disson O, Aschard H, Lecuit M. Differential stress responsiveness determines intraspecies virulence heterogeneity and host adaptation in Listeria monocytogenes. Nat Microbiol 2024; 9:3345-3361. [PMID: 39578578 DOI: 10.1038/s41564-024-01859-8] [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: 03/21/2024] [Accepted: 10/14/2024] [Indexed: 11/24/2024]
Abstract
Microbial pathogenesis is mediated by the expression of virulence genes. However, as microbes with identical virulence gene content can differ in their pathogenic potential, other virulence determinants must be involved. Here, by combining comparative genomics and transcriptomics of a large collection of isolates of the model pathogen Listeria monocytogenes, time-lapse microscopy, in vitro evolution and in vivo experiments, we show that the individual stress responsiveness of L. monocytogenes isolates determines their respective levels of virulence in vivo and reflects their degree of host adaptation. The transcriptional signature that accounts for the heterogeneity in the virulence of L. monocytogenes species is mediated by the stress response regulator SigB and driven by differential stress responsiveness. The tuning of SigB pathway responsiveness is polygenic and influenced by multiple, individually rare gene variations. This study reveals an overarching determinant of microbial virulence, challenging the paradigm of accessory virulence gene content as the major determinant of intraspecies virulence heterogeneity.
Collapse
Affiliation(s)
- Lukas Hafner
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Enzo Gadin
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Lei Huang
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Arthur Frouin
- Statistical Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS USR375, Paris, France
| | - Fabien Laporte
- Statistical Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS USR375, Paris, France
| | - Charlotte Gaultier
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Afonso Vieira
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Claire Maudet
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Hugo Varet
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Alexandra Moura
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
| | - Hélène Bracq-Dieye
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
| | - Nathalie Tessaud-Rita
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
| | - Mylène Maury
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
| | - Melody Dazas
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Rachel Legendre
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Pauline Gastineau
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Yu-Huan Tsai
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Jean-Yves Coppée
- Transcriptome et Epigenome Platform, Biomics, Center for Technological Resources and Research, Institut Pasteur, Université Paris Cité, Paris, France
| | - Caroline Charlier
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
- Necker-Enfants Malades University Hospital, Division of Infectious Diseases and Tropical Medicine, Institut Imagine, APHP, Paris, France
| | - Etienne Patin
- Human Evolutionary Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR2000, Paris, France
| | - Rayan Chikhi
- Sequence Bioinformatics Group, Institut Pasteur, Université Paris Cité, Paris, France
| | - Eduardo P C Rocha
- Microbial Evolutionary Genomics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3525, Paris, France
| | - Alexandre Leclercq
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
| | - Olivier Disson
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Hugues Aschard
- Statistical Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS USR375, Paris, France
| | - Marc Lecuit
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France.
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France.
- Necker-Enfants Malades University Hospital, Division of Infectious Diseases and Tropical Medicine, Institut Imagine, APHP, Paris, France.
| |
Collapse
|
10
|
Carvalho F, Carreaux A, Sartori-Rupp A, Tachon S, Gazi AD, Courtin P, Nicolas P, Dubois-Brissonnet F, Barbotin A, Desgranges E, Bertrand M, Gloux K, Schouler C, Carballido-López R, Chapot-Chartier MP, Milohanic E, Bierne H, Pagliuso A. Aquatic environment drives the emergence of cell wall-deficient dormant forms in Listeria. Nat Commun 2024; 15:8499. [PMID: 39358320 PMCID: PMC11447242 DOI: 10.1038/s41467-024-52633-7] [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: 11/21/2023] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
Stressed bacteria can enter a dormant viable but non-culturable (VBNC) state. VBNC pathogens pose an increased health risk as they are undetectable by growth-based techniques and can wake up back into a virulent state. Although widespread in bacteria, the mechanisms governing this phenotypic switch remain elusive. Here, we investigate the VBNC state transition in the human pathogen Listeria monocytogenes. We show that bacteria starved in mineral water become VBNC by converting into osmotically stable cell wall-deficient coccoid forms, a phenomenon that occurs in other Listeria species. We reveal the bacterial stress response regulator SigB and the autolysin NamA as major actors of VBNC state transition. We lastly show that VBNC Listeria revert to a walled and virulent state after passage in chicken embryos. Our study provides more detail on the VBNC state transition mechanisms, revealing wall-free bacteria naturally arising in aquatic environments as a potential survival strategy in hypoosmotic and oligotrophic conditions.
Collapse
Affiliation(s)
- Filipe Carvalho
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Alexis Carreaux
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | | | - Anastasia D Gazi
- Ultrastructural Bioimaging Facility, Institut Pasteur, Paris, France
| | - Pascal Courtin
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Pierre Nicolas
- INRAE, Université Paris-Saclay, MaIAGE, Jouy-en-Josas, France
| | | | - Aurélien Barbotin
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Emma Desgranges
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Matthieu Bertrand
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Karine Gloux
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Rut Carballido-López
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Eliane Milohanic
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Hélène Bierne
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Alessandro Pagliuso
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
| |
Collapse
|
11
|
Elbakush AM, Trunschke O, Shafeeq S, Römling U, Gomelsky M. Maple compounds prevent biofilm formation in Listeria monocytogenes via sortase inhibition. Front Microbiol 2024; 15:1436476. [PMID: 39351304 PMCID: PMC11439720 DOI: 10.3389/fmicb.2024.1436476] [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: 05/22/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
The Pss exopolysaccharide (EPS) enhances the ability of the foodborne pathogen Listeria monocytogenes to colonize and persist on surfaces of fresh fruits and vegetables. Eradicating listeria within EPS-rich biofilms is challenging due to their increased tolerance to disinfectants, desiccation, and other stressors. Recently, we discovered that extracts of maple wood, including maple sap, are a potent source of antibiofilm agents. Maple lignans, such as nortrachelogenin-8'-O-β-D-glucopyranoside and lariciresinol, were found to inhibit the formation of, and promote the dispersion of pre-formed L. monocytogenes EPS biofilms. However, the mechanism remained unknown. Here, we report that these lignans do not affect Pss EPS synthesis or degradation. Instead, they promote EPS detachment, likely by interfering with an unidentified lectin that keeps EPS attached to the cell surfaces. Furthermore, the maple lignans inhibit the activity of L. monocytogenes sortase A (SrtA) in vitro. SrtA is a transpeptidase that covalently anchors surface proteins, including the Pss-specific lectin, to the cell wall peptidoglycan. Consistent with this, deletion of the srtA gene results in Pss EPS detachment from listerial cells. We also identified several additional maple compounds, including epicatechin gallate, isoscopoletin, scopoletin, and abscisic acid, which inhibit L. monocytogenes SrtA activity in vitro and prevent biofilm formation. Molecular modelling indicates that, despite their structural diversity, these compounds preferentially bind to the SrtA active site. Since maple products are abundant and safe for consumption, our finding that they prevent biofilm formation in L. monocytogenes offers a viable source for protecting fresh produce from this foodborne pathogen.
Collapse
Affiliation(s)
- Ahmed M Elbakush
- Department of Molecular Biology, University of Wyoming, Laramie, WY, United States
| | - Oliver Trunschke
- Department of Molecular Biology, University of Wyoming, Laramie, WY, United States
| | - Sulman Shafeeq
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Ute Römling
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Mark Gomelsky
- Department of Molecular Biology, University of Wyoming, Laramie, WY, United States
| |
Collapse
|
12
|
Kader Chowdhury QMM, Islam S, Narayanan L, Ogunleye SC, Wang S, Thu D, Freitag NE, Lawrence ML, Abdelhamed H. An insight into the role of branched-chain α-keto acid dehydrogenase (BKD) complex in branched-chain fatty acid biosynthesis and virulence of Listeria monocytogenes. J Bacteriol 2024; 206:e0003324. [PMID: 38899896 PMCID: PMC11270904 DOI: 10.1128/jb.00033-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Listeria monocytogenes is a foodborne bacterial pathogen that causes listeriosis. Positive regulatory factor A (PrfA) is a pleiotropic master activator of virulence genes of L. monocytogenes that becomes active upon the entry of the bacterium into the cytosol of infected cells. L. monocytogenes can survive and multiply at low temperatures; this is accomplished through the maintenance of appropriate membrane fluidity via branched-chain fatty acid (BCFA) synthesis. Branched-chain α-keto acid dehydrogenase (BKD), which is composed of four polypeptides encoded by lpd, bkdA1, bkdA2, and bkdB, is known to play a vital role in BCFA biosynthesis. Here, we constructed BKD-deficient Listeria strains by in-frame deletion of lpd, bkdA1, bkdA2, and bkdB genes. To determine the role in in vivo and in vitro, mouse model challenges, plaque assay in murine L2 fibroblast, and intracellular replication in J744A.1 macrophage were conducted. BKD-deficient strains exhibited defects in BCFA composition, virulence, and PrfA-regulon function within the host cells. Transcriptomics analysis revealed that the transcript level of the PrfA-regulon was lower in ΔbkdA1 strain than those in the wild-type. This study demonstrates that L. monocytogenes strains lacking BKD complex components were defective in PrfA-regulon function, and full activation of wild-type prfA may not occur within host cells in the absence of BKD. Further study will investigate the consequences of BKD deletion on PrfA function through altering BCFA catabolism.IMPORTANCEListeria monocytogenes is the causative agent of listeriosis, a disease with a high mortality rate. In this study, we have shown that the deletion of BKD can impact the function of PrfA and the PrfA-regulon. The production of virulence proteins within host cells is necessary for L. monocytogenes to promote its intracellular survival and is likely dependent on membrane integrity. We thus report a link between L. monocytogenes membrane integrity and the function of PrfA. This knowledge will increase our understanding of L. monocytogenes pathogenesis, which may provide insight into the development of antimicrobial agents.
Collapse
Affiliation(s)
- Q M Monzur Kader Chowdhury
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Shamima Islam
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Lakshmi Narayanan
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Seto C. Ogunleye
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Shangshang Wang
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, Mississippi, USA
| | - Dinh Thu
- Tyson Foods, R&D Ingredient Solutions, Springdale, Arkansas, USA
| | - Nancy E. Freitag
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mark L. Lawrence
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Hossam Abdelhamed
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| |
Collapse
|
13
|
Liu C, Qian R, Shi W, Kou L, Wang J, Ma X, Ren H, Gao S, Ren J. EⅡB Mutation Reduces the Pathogenicity of Listeria monocytogenes by Negatively Regulating Biofilm Formation Ability, Infective Capacity, and Virulence Gene Expression. Vet Sci 2024; 11:301. [PMID: 39057985 PMCID: PMC11281496 DOI: 10.3390/vetsci11070301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
To explore the role of the membrane permease ⅡB (EⅡB) gene of Listeria pathogenicity island 4 (LIPI-4) in the virulence of Listeria monocytogenes, both an EⅡB deletion strain (∆EⅡB) and a complemented strain were constructed. In vitro experiments demonstrated that EⅡB deletion affected the biofilm formation ability of the wild-type strain (Lm928). Moreover, this deletion decreased the intracellular proliferation abilities of L. monocytogenes. Mice infected with ∆EⅡB survived longer and experienced less weight loss on days 1, 2, and 3 post-infection. The bacterial load in the liver tissue of ∆EⅡB-infected mice was significantly reduced, and a considerable decrease in the blood levels of inflammatory cytokines IL-β, IL-6, IL-10, and TNF-α were observed. Following EⅡB deletion, 65% (13/20) of genes were downregulated, 25% (5/20) were upregulated, and 10% (2/20) showed no change. These findings suggest that EⅡB deletion may reduce both the in vivo and in vitro virulence levels as well as the biofilm formation ability of Lm928 by downregulating the transcription levels of genes associated with virulence and biofilm formation. These findings provide a foundation for further examining the pathogenic mechanisms of LIPI-4 and EⅡB in L. monocytogenes.
Collapse
Affiliation(s)
| | | | | | | | - Jing Wang
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (C.L.); (R.Q.); (W.S.); (L.K.); (H.R.); (S.G.); (J.R.)
| | - Xun Ma
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (C.L.); (R.Q.); (W.S.); (L.K.); (H.R.); (S.G.); (J.R.)
| | | | | | | |
Collapse
|
14
|
Chatterjee A, Kaval KG, Garsin DA. Role of ethanolamine utilization and bacterial microcompartment formation in Listeria monocytogenes intracellular infection. Infect Immun 2024; 92:e0016224. [PMID: 38752742 PMCID: PMC11237587 DOI: 10.1128/iai.00162-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 04/18/2024] [Indexed: 05/28/2024] Open
Abstract
Ethanolamine (EA) affects the colonization and pathogenicity of certain human bacterial pathogens in the gastrointestinal tract. However, EA can also affect the intracellular survival and replication of host cell invasive bacteria such as Listeria monocytogenes (LMO) and Salmonella enterica serovar Typhimurium (S. Typhimurium). The EA utilization (eut) genes can be categorized as regulatory, enzymatic, or structural, and previous work in LMO showed that loss of genes encoding functions for the enzymatic breakdown of EA inhibited LMO intracellular replication. In this work, we sought to further characterize the role of EA utilization during LMO infection of host cells. Unlike what was previously observed for S. Typhimurium, in LMO, an EA regulator mutant (ΔeutV) was equally deficient in intracellular replication compared to an EA metabolism mutant (ΔeutB), and this was consistent across Caco-2, RAW 264.7, and THP-1 cell lines. The structural genes encode proteins that self-assemble into bacterial microcompartments (BMCs) that encase the enzymes necessary for EA metabolism. For the first time, native EUT BMCs were fluorescently tagged, and EUT BMC formation was observed in vitro and in vivo. Interestingly, BMC formation was observed in bacteria infecting Caco-2 cells, but not the macrophage cell lines. Finally, the cellular immune response of Caco-2 cells to infection with eut mutants was examined, and it was discovered that ΔeutB and ΔeutV mutants similarly elevated the expression of inflammatory cytokines. In conclusion, EA sensing and utilization during LMO intracellular infection are important for optimal LMO replication and immune evasion but are not always concomitant with BMC formation.IMPORTANCEListeria monocytogenes (LMO) is a bacterial pathogen that can cause severe disease in immunocompromised individuals when consumed in contaminated food. It can replicate inside of mammalian cells, escaping detection by the immune system. Therefore, understanding the features of this human pathogen that contribute to its infectiousness and intracellular lifestyle is important. In this work we demonstrate that genes encoding both regulators and enzymes of EA metabolism are important for optimal growth inside mammalian cells. Moreover, the formation of specialized compartments to enable EA metabolism were visualized by tagging with a fluorescent protein and found to form when LMO infects some mammalian cell types, but not others. Interestingly, the formation of the compartments was associated with features consistent with an early stage of the intracellular infection. By characterizing bacterial metabolic pathways that contribute to survival in host environments, we hope to positively impact knowledge and facilitate new treatment strategies.
Collapse
Affiliation(s)
- Ayan Chatterjee
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center, Houston, Texas, USA
| | - Karan Gautam Kaval
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center, Houston, Texas, USA
| | - Danielle A. Garsin
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
15
|
Pham HG, Tran KN, Gomelsky L, Roy T, Gigley JP, Gomelsky M. Robust inducible gene expression in intracellular Listeria monocytogenes in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596178. [PMID: 38853860 PMCID: PMC11160606 DOI: 10.1101/2024.05.28.596178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Attenuated strains of the intracellular pathogen Listeria monocytogenes can deliver genetically encoded payloads inside tumor cells. L. monocytogenes preferentially accumulates and propagates inside immune-suppressed tumor microenvironments. To maximize the payload impact in tumors and minimize damage to healthy tissues, it is desirable to induce payload synthesis when bacteria are eliminated from the healthy tissues but are grown to high numbers intratumorally. Here, we have engineered a tightly controlled gene expression system for intracellular L. monocytogenes inducible with a cumin derivative, cumate. Upon cumate addition, expression of a reporter gene is increased in L. monocytogenes growing in vitro by 80-fold, and in intracellular L. monocytogenes in murine tumors by 10-fold. This study demonstrates the feasibility of activating gene expression in intracellular bacteria in live animals using an edible inducer. The system is expected to enhance the efficacy and safety of the attenuated L. monocytogenes strains as antitumor payload delivery bacterial drones.
Collapse
|
16
|
Tullius MV, Bowen RA, Back PS, Masleša-Galić S, Nava S, Horwitz MA. LVS Δ capB-vectored multiantigenic melioidosis vaccines protect against lethal respiratory Burkholderia pseudomallei challenge in highly sensitive BALB/c mice. mBio 2024; 15:e0018624. [PMID: 38511933 PMCID: PMC11005352 DOI: 10.1128/mbio.00186-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/05/2024] [Indexed: 03/22/2024] Open
Abstract
Melioidosis, caused by the intracellular bacterial pathogen and Tier 1 select agent Burkholderia pseudomallei (Bp), is a highly fatal disease endemic in tropical areas. No licensed vaccine against melioidosis exists. In preclinical vaccine studies, demonstrating protection against respiratory infection in the highly sensitive BALB/c mouse has been especially challenging. To address this challenge, we have used a safe yet potent live attenuated platform vector, LVS ΔcapB, previously used successfully to develop vaccines against the Tier 1 select agents of tularemia, anthrax, and plague, to develop a melioidosis vaccine. We have engineered melioidosis vaccines (rLVS ΔcapB/Bp) expressing multiple immunoprotective Bp antigens among type VI secretion system proteins Hcp1, Hcp2, and Hcp6, and membrane protein LolC. Administered intradermally, rLVS ΔcapB/Bp vaccines strongly protect highly sensitive BALB/c mice against lethal respiratory Bp challenge, but protection is overwhelmed at very high challenge doses. In contrast, administered intranasally, rLVS ΔcapB/Bp vaccines remain strongly protective against even very high challenge doses. Under some conditions, the LVS ΔcapB vector itself provides significant protection against Bp challenge, and consistent with this, both the vector and vaccines induce humoral immune responses to Bp antigens. Three-antigen vaccines expressing Hcp6-Hcp1-Hcp2 or Hcp6-Hcp1-LolC are among the most potent and provide long-term protection and protection even with a single intranasal immunization. Protection via the intranasal route was either comparable to or statistically significantly better than the single-deletional Bp mutant Bp82, which served as a positive control. Thus, rLVS ΔcapB/Bp vaccines are exceptionally promising safe and potent melioidosis vaccines. IMPORTANCE Melioidosis, a major neglected disease caused by the intracellular bacterial pathogen Burkholderia pseudomallei, is endemic in many tropical areas of the world and causes an estimated 165,000 cases and 89,000 deaths in humans annually. Moreover, B. pseudomallei is categorized as a Tier 1 select agent of bioterrorism, largely because inhalation of low doses can cause rapidly fatal pneumonia. No licensed vaccine is available to prevent melioidosis. Here, we describe a safe and potent melioidosis vaccine that protects against lethal respiratory challenge with B. pseudomallei in a highly sensitive small animal model-even a single immunization is highly protective, and the vaccine gives long-term protection. The vaccine utilizes a highly attenuated replicating intracellular bacterium as a vector to express multiple key proteins of B. pseudomallei; this vector platform has previously been used successfully to develop potent vaccines against other Tier 1 select agent diseases including tularemia, anthrax, and plague.
Collapse
Affiliation(s)
- Michael V. Tullius
- Division of Infectious Diseases, Department of Medicine, Center for Health Sciences, School of Medicine, University of California, Los Angeles, California, USA
| | - Richard A. Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Peter S. Back
- Division of Infectious Diseases, Department of Medicine, Center for Health Sciences, School of Medicine, University of California, Los Angeles, California, USA
| | - Saša Masleša-Galić
- Division of Infectious Diseases, Department of Medicine, Center for Health Sciences, School of Medicine, University of California, Los Angeles, California, USA
| | - Susana Nava
- Division of Infectious Diseases, Department of Medicine, Center for Health Sciences, School of Medicine, University of California, Los Angeles, California, USA
| | - Marcus A. Horwitz
- Division of Infectious Diseases, Department of Medicine, Center for Health Sciences, School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
17
|
Ghatani K, Prasad Sha S, Thapa S, Chakraborty P, Sarkar S. Bifidobacterial Genome Editing for Potential Probiotic Development. GENOME EDITING IN BACTERIA (PART 1) 2024:62-87. [DOI: 10.2174/9789815165678124010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Genome editing is a promising tool in the era of modern biotechnology that
can alter the DNA of many organisms. It is now extensively used in various industries
to obtain the well-desired and enhanced characteristics to improve the yield and
nutritional quality of products. The positive health attributes of Bifidobacteria, such as
prevention of diarrhoea, reduction of ulcerative colitis, prevention of necrotizing
enterocolitis, etc., have shown promising reports in many clinical trials. The potential
use of Bifidobacteria as starter or adjunct cultures has become popular. Currently,
Bifidobacterium bifidum, B. adolescentis, B. breve, B. infantis, B. longum, and B. lactis
find a significant role in the development of probiotic fermented dairy products.
However, Bifidobacteria, one of the first colonizers of the human GI tract and an
indicator of the health status of an individual, has opened new avenues for research
and, thereby, its application. Besides this, the GRAS/QPS (Generally Regarded as
Safe/Qualified Presumption of Safety) status of Bifidobacteria makes it safe for use.
They belong to the subgroup (which are the fermentative types that are primarily found
in the natural cavities of humans and animals) of Actinomycetes. B. lactis has been used
industrially in fermented foods, such as yogurt, cheese, beverages, sausages, infant
formulas, and cereals. In the present book chapter, the authors tried to explore the
origin, health attributes, and various genetic engineering tools for genome editing of
Bifidobacteria for the development of starter culture for dairy and non-dairy industrial
applications as well as probiotics.
Collapse
Affiliation(s)
- Kriti Ghatani
- Department of Food Technology, University of North Bengal, Raja Rammohunpur, Darjeeling,
West Bengal, 734013, India
| | - Shankar Prasad Sha
- Department of Botany, Food Microbiology Lab, Kurseong College, University of North Bengal,
Dow Hill Road, Kurseong, Darjeeling 7342003, West Bengal, India
| | - Subarna Thapa
- Department of Food Technology, University of North Bengal, Raja Rammohunpur, Darjeeling,
West Bengal, 734013, India
| | - Priya Chakraborty
- Department of Food Technology, University of North Bengal, Raja Rammohunpur, Darjeeling,
West Bengal, 734013, India
| | - Sagnik Sarkar
- Department of Food Technology, University of North Bengal, Raja Rammohunpur, Darjeeling,
West Bengal, 734013, India
| |
Collapse
|
18
|
Wu J, McAuliffe O, O'Byrne CP. A novel RofA-family transcriptional regulator, GadR, controls the development of acid resistance in Listeria monocytogenes. mBio 2023; 14:e0171623. [PMID: 37882515 PMCID: PMC10746197 DOI: 10.1128/mbio.01716-23] [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: 07/04/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
IMPORTANCE The ability to survive the acidic conditions found in the stomach is crucial for the food-borne pathogen Listeria monocytogenes to gain access to the mammalian gastrointestinal tract. Little is currently known about how acid resistance is regulated in this pathogen and why this trait is highly variable between strains. Here, we used comparative genomics to identify a novel RofA-family transcriptional regulator, GadR, that controls the development of acid resistance. The RofA family of regulators was previously found only in a small group of bacterial pathogens, including streptococci, where they regulate virulence properties. We show that gadR encodes the dominant regulator of acid resistance in L. monocytogenes and that its sequence variability accounts for previously observed differences between strains in this trait. Together, these findings significantly advance our understanding of how this important pathogen copes with acid stress and suggest a potential molecular target to aid its control in the food chain.
Collapse
Affiliation(s)
- Jialun Wu
- Bacterial Stress Response Group, Microbiology, Ryan Institute, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Olivia McAuliffe
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
- School of Biological Sciences, Queen’s University Belfast, Belfast, Northern Ireland
| | - Conor P. O'Byrne
- Bacterial Stress Response Group, Microbiology, Ryan Institute, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| |
Collapse
|
19
|
Smith HB, Lee K, Freeman MJ, Stevenson DM, Amador-Noguez D, Sauer JD. Listeria monocytogenes requires DHNA-dependent intracellular redox homeostasis facilitated by Ndh2 for survival and virulence. Infect Immun 2023; 91:e0002223. [PMID: 37754681 PMCID: PMC10580952 DOI: 10.1128/iai.00022-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023] Open
Abstract
Listeria monocytogenes is a remarkably well-adapted facultative intracellular pathogen that can thrive in a wide range of ecological niches. L. monocytogenes maximizes its ability to generate energy from diverse carbon sources using a respiro-fermentative metabolism that can function under both aerobic and anaerobic conditions. Cellular respiration maintains redox homeostasis by regenerating NAD+ while also generating a proton motive force. The end products of the menaquinone (MK) biosynthesis pathway are essential to drive both aerobic and anaerobic cellular respirations. We previously demonstrated that intermediates in the MK biosynthesis pathway, notably 1,4-dihydroxy-2-naphthoate (DHNA), are required for the survival and virulence of L. monocytogenes independent of their role in respiration. Furthermore, we found that restoration of NAD+/NADH ratio through expression of water-forming NADH oxidase could rescue phenotypes associated with DHNA deficiency. Here, we extend these findings to demonstrate that endogenous production or direct supplementation of DHNA restored both the cellular redox homeostasis and metabolic output of fermentation in L. monocytogenes. Furthermore, exogenous supplementation of DHNA rescues the in vitro growth and ex vivo virulence of L. monocytogenes DHNA-deficient mutants. Finally, we demonstrate that exogenous DHNA restores redox balance in L. monocytogenes specifically through the recently annotated NADH dehydrogenase Ndh2, independent of its role in the extracellular electron transport pathway. These data suggest that the production of DHNA may represent an additional layer of metabolic adaptability by L. monocytogenes to drive energy metabolism in the absence of respiration-favorable conditions.
Collapse
Affiliation(s)
- Hans B. Smith
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kijeong Lee
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Matthew J. Freeman
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David M. Stevenson
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Daniel Amador-Noguez
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
20
|
Anderson TS, McCormick AL, Daugherity EA, Oladejo M, Okpalanwaka IF, Smith SL, Appiah D, Wood LM, Lowe DB. Listeria-based vaccination against the pericyte antigen RGS5 elicits anti-vascular effects and colon cancer protection. Oncoimmunology 2023; 12:2260620. [PMID: 37781234 PMCID: PMC10540654 DOI: 10.1080/2162402x.2023.2260620] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/14/2023] [Indexed: 10/03/2023] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality despite efforts to improve standard interventions. As CRC patients can benefit from immunotherapeutic strategies that incite effector T cell action, cancer vaccines represent a safe and promising therapeutic approach to elicit protective and durable immune responses against components of the tumor microenvironment (TME). In this study, we investigate the pre-clinical potential of a Listeria monocytogenes (Lm)-based vaccine targeting the CRC-associated vasculature. CRC survival and progression are reliant on functioning blood vessels to effectively mediate various metabolic processes and oxygenate underlying tissues. We, therefore, advance the strategy of initiating immunity in syngeneic mouse models against the endogenous pericyte antigen RGS5, which is a critical mediator of pathological vascularization. Overall, Lm-based vaccination safely induced potent anti-tumor effects that consisted of recruiting functional Type-1-associated T cells into the TME and reducing tumor blood vessel content. This study underscores the promising clinical potential of targeting RGS5 against vascularized tumors like CRC.
Collapse
Affiliation(s)
- Trevor S. Anderson
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Amanda L. McCormick
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Elizabeth A. Daugherity
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Izuchukwu F. Okpalanwaka
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Savanna L. Smith
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Duke Appiah
- Department of Public Health, School of Population and Public Health, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Laurence M. Wood
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| | - Devin B. Lowe
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, USA
| |
Collapse
|
21
|
Goedseels M, Michiels CW. Cell Envelope Modifications Generating Resistance to Hop Beta Acids and Collateral Sensitivity to Cationic Antimicrobials in Listeria monocytogenes. Microorganisms 2023; 11:2024. [PMID: 37630584 PMCID: PMC10457916 DOI: 10.3390/microorganisms11082024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Hop beta acids (HBAs) are characteristic compounds from the hop plant that are of interest for their strong antimicrobial activity. In this work, we report a resistance mechanism against HBA in the foodborne pathogen Listeria monocytogenes. Using an evolution experiment, we isolated two HBA-resistant mutants with mutations in the mprF gene, which codes for the Multiple Peptide Resistance Factor, an enzyme that confers resistance to cationic peptides and antibiotics in several Gram-positive bacteria by lysinylating membrane phospholipids. Besides the deletion of mprF, the deletion of dltA, which mediates the alanylation of teichoic acids, resulted in increased HBA resistance, suggesting that resistance may be caused by a reduction in positive charges on the cell surface. Additionally, we found that this resistance is maintained at low pH, indicating that the resistance mechanism is not solely based on electrostatic interactions of HBA with the cell surface. Finally, we showed that the HBA-resistant mutants display collateral sensitivity to the cationic antimicrobials polymyxin B and nisin, which may open perspectives for combining antimicrobials to prevent resistance development.
Collapse
Affiliation(s)
| | - Chris W. Michiels
- Department of Microbial and Molecular Systems, KU Leuven, B-3000 Leuven, Belgium;
| |
Collapse
|
22
|
Sun L, Van Loey A, Buvé C, Michiels CW. Experimental Evolution Reveals a Novel Ene Reductase That Detoxifies α,β-Unsaturated Aldehydes in Listeria monocytogenes. Microbiol Spectr 2023; 11:e0487722. [PMID: 37036358 PMCID: PMC10269891 DOI: 10.1128/spectrum.04877-22] [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: 11/30/2022] [Accepted: 03/17/2023] [Indexed: 04/11/2023] Open
Abstract
The plant essential oil component trans-cinnamaldehyde (t-CIN) exhibits antibacterial activity against a broad range of foodborne pathogenic bacteria, including L. monocytogenes, but its mode of action is not fully understood. In this study, several independent mutants of L. monocytogenes with increased t-CIN tolerance were obtained via experimental evolution. Whole-genome sequencing (WGS) analysis revealed single-nucleotide-variation mutations in the yhfK gene, encoding an oxidoreductase of the short-chain dehydrogenases/reductases superfamily, in each mutant. The deletion of yhfK conferred increased sensitivity to t-CIN and several other α,β-unsaturated aldehydes, including trans-2-hexenal, citral, and 4-hydroxy-2-nonenal. The t-CIN tolerance of the deletion mutant was restored via genetic complementation with yhfK. Based on a gas chromatography-mass spectrometry (GC-MS) analysis of the culture supernatants, it is proposed that YhfK is an ene reductase that converts t-CIN to 3-phenylpropanal by reducing the C=C double bond of the α,β-unsaturated aldehyde moiety. YhfK homologs are widely distributed in Bacteria, and the deletion of the corresponding homolog in Bacillus subtilis also caused increased sensitivity to t-CIN and trans-2-hexenal, suggesting that this protein may have a conserved function to protect bacteria against toxic α,β-unsaturated aldehydes in their environments. IMPORTANCE While bacterial resistance against clinically used antibiotics has been well studied, less is known about resistance against other antimicrobials, such as natural compounds that could replace traditional food preservatives. In this work, we report that the food pathogen Listeria monocytogenes can rapidly develop an elevated tolerance against t-cinnamaldehyde, a natural antimicrobial from cinnamon, by single base pair changes in the yhfK gene. The enzyme encoded by this gene is an oxidoreductase, but its substrates and precise role were hitherto unknown. We demonstrate that the enzyme reduces the double bond in t-cinnamaldehyde and thereby abolishes its antibacterial activity. Furthermore, the mutations linked to t-CIN tolerance increased bacterial sensitivity to a related compound, suggesting that they modify the substrate specificity of the enzyme. Since the family of oxidoreductases to which YhfK belongs is of great interest in the mediation of stereospecific reactions in biocatalysis, our work may also have unanticipated application potential in this field.
Collapse
Affiliation(s)
- Lei Sun
- Department of Microbial and Molecular Systems and Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Leuven, Belgium
| | - Ann Van Loey
- Department of Microbial and Molecular Systems and Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Leuven, Belgium
| | - Carolien Buvé
- Department of Microbial and Molecular Systems and Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Leuven, Belgium
| | - Chris W. Michiels
- Department of Microbial and Molecular Systems and Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Vaval Taylor DM, Xayarath B, Freitag NE. Two Permeases Associated with the Multifunctional CtaP Cysteine Transport System in Listeria monocytogenes Play Distinct Roles in Pathogenesis. Microbiol Spectr 2023; 11:e0331722. [PMID: 37199604 PMCID: PMC10269559 DOI: 10.1128/spectrum.03317-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 04/05/2023] [Indexed: 05/19/2023] Open
Abstract
The soil-dwelling bacterium Listeria monocytogenes survives a multitude of conditions when residing in the outside environment and as a pathogen within host cells. Key to survival within the infected mammalian host is the expression of bacterial gene products necessary for nutrient acquisition. Similar to many bacteria, L. monocytogenes uses peptide import to acquire amino acids. Peptide transport systems play an important role in nutrient uptake as well as in additional functions that include bacterial quorum sensing and signal transduction, recycling of peptidoglycan fragments, adherence to eukaryotic cells, and alterations in antibiotic susceptibility. It has been previously described that CtaP, encoded by lmo0135, is a multifunctional protein associated with activities that include cysteine transport, resistance to acid, membrane integrity, and bacterial adherence to host cells. ctaP is located next to two genes predicted to encode membrane-bound permeases lmo0136 and lmo0137, termed CtpP1 and CtpP2, respectively. Here, we show that CtpP1 and CtpP2 are required for bacterial growth in the presence of low concentrations of cysteine and for virulence in mouse infection models. Taken together, the data identify distinct nonoverlapping roles for two related permeases that are important for the growth and survival of L. monocytogenes within host cells. IMPORTANCE Bacterial peptide transport systems are important for nutrient uptake and may additionally function in a variety of other roles, including bacterial communication, signal transduction, and bacterial adherence to eukaryotic cells. Peptide transport systems often consist of a substrate-binding protein associated with a membrane-spanning permease. The environmental bacterial pathogen Listeria monocytogenes uses the substrate-binding protein CtaP not only for cysteine transport but also for resistance to acid, maintenance of membrane integrity, and bacterial adherence to host cells. In this study, we demonstrate complementary yet distinct functional roles for two membrane permeases, CtpP1 and CtpP2, that are encoded by genes linked to ctaP and that contribute to bacterial growth, invasion, and pathogenicity.
Collapse
Affiliation(s)
- Diandra M. Vaval Taylor
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Bobbi Xayarath
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Nancy E. Freitag
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
24
|
Pensinger DA, Gutierrez KV, Smith HB, Vincent WJB, Stevenson DS, Black KA, Perez-Medina KM, Dillard JP, Rhee KY, Amador-Noguez D, Huynh TN, Sauer JD. Listeria monocytogenes GlmR Is an Accessory Uridyltransferase Essential for Cytosolic Survival and Virulence. mBio 2023; 14:e0007323. [PMID: 36939339 PMCID: PMC10128056 DOI: 10.1128/mbio.00073-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/14/2023] [Indexed: 03/21/2023] Open
Abstract
The cytosol of eukaryotic host cells is an intrinsically hostile environment for bacteria. Understanding how cytosolic pathogens adapt to and survive in the cytosol is critical to developing novel therapeutic interventions against these pathogens. The cytosolic pathogen Listeria monocytogenes requires glmR (previously known as yvcK), a gene of unknown function, for resistance to cell-wall stress, cytosolic survival, inflammasome avoidance, and, ultimately, virulence in vivo. In this study, a genetic suppressor screen revealed that blocking utilization of UDP N-acetylglucosamine (UDP-GlcNAc) by a nonessential wall teichoic acid decoration pathway restored resistance to lysozyme and partially restored virulence of ΔglmR mutants. In parallel, metabolomic analysis revealed that ΔglmR mutants are impaired in the production of UDP-GlcNAc, an essential peptidoglycan and wall teichoic acid (WTA) precursor. We next demonstrated that purified GlmR can directly catalyze the synthesis of UDP-GlcNAc from GlcNAc-1P and UTP, suggesting that it is an accessory uridyltransferase. Biochemical analysis of GlmR orthologues suggests that uridyltransferase activity is conserved. Finally, mutational analysis resulting in a GlmR mutant with impaired catalytic activity demonstrated that uridyltransferase activity was essential to facilitate cell-wall stress responses and virulence in vivo. Taken together, these studies indicate that GlmR is an evolutionary conserved accessory uridyltransferase required for cytosolic survival and virulence of L. monocytogenes. IMPORTANCE Bacterial pathogens must adapt to their host environment in order to cause disease. The cytosolic bacterial pathogen Listeria monocytogenes requires a highly conserved protein of unknown function, GlmR (previously known as YvcK), to survive in the host cytosol. GlmR is important for resistance to some cell-wall stresses and is essential for virulence. The ΔglmR mutant is deficient in production of an essential cell-wall metabolite, UDP-GlcNAc, and suppressors that increase metabolite levels also restore virulence. Purified GlmR can directly catalyze the synthesis of UDP-GlcNAc, and this enzymatic activity is conserved in both Bacillus subtilis and Staphylococcus aureus. These results highlight the importance of accessory cell wall metabolism enzymes in responding to cell-wall stress in a variety of Gram-positive bacteria.
Collapse
Affiliation(s)
- Daniel A. Pensinger
- Department of Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Kimberly V. Gutierrez
- Department of Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Hans B. Smith
- Department of Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - William J. B. Vincent
- Department of Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - David S. Stevenson
- Department of Bacteriology, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | | | - Krizia M. Perez-Medina
- Department of Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Joseph P. Dillard
- Department of Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Kyu Y. Rhee
- Weill Cornell Medical College, New York, New York, USA
| | - Daniel Amador-Noguez
- Microbiology Doctoral Training Program, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Department of Bacteriology, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - TuAnh N. Huynh
- Microbiology Doctoral Training Program, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Department of Food Science, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin—Madison, Madison, Wisconsin, USA
| |
Collapse
|
25
|
Wohlfarth JC, Feldmüller M, Schneller A, Kilcher S, Burkolter M, Meile S, Pilhofer M, Schuppler M, Loessner MJ. L-form conversion in Gram-positive bacteria enables escape from phage infection. Nat Microbiol 2023; 8:387-399. [PMID: 36717719 PMCID: PMC9981463 DOI: 10.1038/s41564-022-01317-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 12/22/2022] [Indexed: 02/01/2023]
Abstract
At the end of a lytic bacteriophage replication cycle in Gram-positive bacteria, peptidoglycan-degrading endolysins that cause explosive cell lysis of the host can also attack non-infected bystander cells. Here we show that in osmotically stabilized environments, Listeria monocytogenes can evade phage predation by transient conversion to a cell wall-deficient L-form state. This L-form escape is triggered by endolysins disintegrating the cell wall from without, leading to turgor-driven extrusion of wall-deficient, yet viable L-form cells. Remarkably, in the absence of phage predation, we show that L-forms can quickly revert to the walled state. These findings suggest that L-form conversion represents a population-level persistence mechanism to evade complete eradication by phage attack. Importantly, we also demonstrate phage-mediated L-form switching of the urinary tract pathogen Enterococcus faecalis in human urine, which underscores that this escape route may be widespread and has important implications for phage- and endolysin-based therapeutic interventions.
Collapse
Affiliation(s)
- Jan C Wohlfarth
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Miki Feldmüller
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
| | - Alissa Schneller
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Samuel Kilcher
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Marco Burkolter
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Susanne Meile
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Martin Pilhofer
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
| | - Markus Schuppler
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Martin J Loessner
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
26
|
Smith HB, Lee K, Stevenson DM, Amador-Noguez D, Sauer JD. Listeria monocytogenes requires DHNA-dependent intracellular redox homeostasis facilitated by Ndh2 for survival and virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.524026. [PMID: 36711537 PMCID: PMC9882099 DOI: 10.1101/2023.01.13.524026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Listeria monocytogenes is a remarkably well-adapted facultative intracellular pathogen that can thrive in a wide range of ecological niches. L. monocytogenes maximizes its ability to generate energy from diverse carbon sources using a respiro-fermentative metabolism that can function under both aerobic and anaerobic conditions. Cellular respiration maintains redox homeostasis by regenerating NAD + while also generating a proton motive force (PMF). The end products of the menaquinone (MK) biosynthesis pathway are essential to drive both aerobic and anaerobic cellular respiration. We previously demonstrated that intermediates in the MK biosynthesis pathway, notably 1,4-dihydroxy-2-naphthoate (DHNA), are required for the survival and virulence of L. monocytogenes independent of their role in respiration. Furthermore, we found that restoration of NAD + /NADH ratio through expression of water-forming NADH oxidase (NOX) could rescue phenotypes associated with DHNA deficiency. Here we extend these findings to demonstrate that endogenous production or direct supplementation of DHNA restored both the cellular redox homeostasis and metabolic output of fermentation in L. monocytogenes . Further, exogenous supplementation of DHNA rescues the in vitro growth and ex vivo virulence of L. monocytogenes DHNA-deficient mutants. Finally, we demonstrate that exogenous DHNA restores redox balance in L. monocytogenes specifically through the recently annotated NADH dehydrogenase Ndh2, independent of the extracellular electron transport (EET) pathway. These data suggest that the production of DHNA may represent an additional layer of metabolic adaptability by L. monocytogenes to drive energy metabolism in the absence of respiration-favorable conditions.
Collapse
Affiliation(s)
- Hans B. Smith
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53706, United States of America
| | - Kijeong Lee
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53706, United States of America
| | - David M. Stevenson
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, 53706, United States of America
| | - Daniel Amador-Noguez
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, 53706, United States of America
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53706, United States of America
| |
Collapse
|
27
|
Zhang Y, Guo Q, Fang X, Yuan M, Hu W, Liang X, Liu J, Yang Y, Fang C. Destroying glutathione peroxidase improves the oxidative stress resistance and pathogenicity of Listeria monocytogenes. Front Microbiol 2023; 14:1122623. [PMID: 37032864 PMCID: PMC10073551 DOI: 10.3389/fmicb.2023.1122623] [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: 12/13/2022] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Glutathione peroxidase is abundant in eukaryotes as an important antioxidant enzyme. However, prokaryotic glutathione peroxidase has not been thoroughly studied. Listeria monocytogenes is a facultative intracellular pathogen that is capable of causing listeriosis in animals as well as humans. Despite the fact that L. monocytogenes encodes a putative glutathione peroxidase, GSH-Px (encoded by lmo0983)), the functions of the enzyme are still unknown. Here we revealed the unusual roles of L. monocytogenes GSH-Px in bacterial antioxidants and pathogenicity. Methods L. monocytogenes Lm850658 was taken as the parental strain to construct the gsh-px deletion strain and related complement strain. The effect of the gsh-px gene on the resistance of L. monocytogenes to oxidative stress was determined by measuring the concentrations of glutathione and assaying the stress survival rates under different oxidative conditions. In addition, the pathogenicity of L. monocytogenes was determined by cellular adhesion and invasion assays and mice virulence tests, and the expression of virulence factors was determined by Western blot. Results Deficiency of GSH-Px not only increased glutathione concentrations in L. monocytogenes but also enhanced its resistance to oxidative stress when exposed to copper and iron ions. In addition, the absence of gsh-px significantly improved the adhesion and invasion efficiency of L. monocytogenes to Caco-2 cells. More importantly, L. monocytogenes lacking GSH-Px could colonize and proliferate more efficiently in mice livers and spleens, enhancing the pathogenicity of L. monocytogenes by increasing the expression of virulence factors like InlA, InlB, and LLO. Discussion Taken together, we confirmed that GSH-Px of L. monocytogenes has a counter-intuitive effect on the antioxidant capacity and pathogenicity.
Collapse
|
28
|
Effect of gastric pH and bile acids on the survival of Listeria monocytogenes and Salmonella Typhimurium during simulated gastrointestinal digestion. INNOV FOOD SCI EMERG 2022. [DOI: 10.1016/j.ifset.2022.103161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
29
|
Fischer MA, Engelgeh T, Rothe P, Fuchs S, Thürmer A, Halbedel S. Listeria monocytogenes genes supporting growth under standard laboratory cultivation conditions and during macrophage infection. Genome Res 2022; 32:1711-1726. [PMID: 36114002 PMCID: PMC9528990 DOI: 10.1101/gr.276747.122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/04/2022] [Indexed: 11/24/2022]
Abstract
The Gram-positive bacterium Listeria monocytogenes occurs widespread in the environment and infects humans when ingested along with contaminated food. Such infections are particularly dangerous for risk group patients, for whom they represent a life-threatening disease. To invent novel strategies to control contamination and disease, it is important to identify those cellular processes that maintain pathogen growth inside and outside the host. Here, we have applied transposon insertion sequencing (Tn-Seq) to L. monocytogenes for the identification of such processes on a genome-wide scale. Our approach identified 394 open reading frames that are required for growth under standard laboratory conditions and 42 further genes, which become necessary during intracellular growth in macrophages. Most of these genes encode components of the translation machinery and act in chromosome-related processes, cell division, and biosynthesis of the cellular envelope. Several cofactor biosynthesis pathways and 29 genes with unknown functions are also required for growth, suggesting novel options for the development of antilisterial drugs. Among the genes specifically required during intracellular growth are known virulence factors, genes compensating intracellular auxotrophies, and several cell division genes. Our experiments also highlight the importance of PASTA kinase signaling for general viability and of glycine metabolism and chromosome segregation for efficient intracellular growth of L. monocytogenes.
Collapse
Affiliation(s)
- Martin A Fischer
- FG11 Division of Enteropathogenic Bacteria and Legionella, Robert Koch Institute, 38855 Wernigerode, Germany
| | - Tim Engelgeh
- FG11 Division of Enteropathogenic Bacteria and Legionella, Robert Koch Institute, 38855 Wernigerode, Germany
| | - Patricia Rothe
- FG11 Division of Enteropathogenic Bacteria and Legionella, Robert Koch Institute, 38855 Wernigerode, Germany
| | - Stephan Fuchs
- MF1 Bioinformatic Support, Robert Koch Institute, 13353 Berlin, Germany
| | - Andrea Thürmer
- MF2 Genome Sequencing, Robert Koch Institute, 13353 Berlin, Germany
| | - Sven Halbedel
- FG11 Division of Enteropathogenic Bacteria and Legionella, Robert Koch Institute, 38855 Wernigerode, Germany
| |
Collapse
|
30
|
Cahoon LA, Alejandro‐Navarreto X, Gururaja AN, Light SH, Alonzo F, Anderson WF, Freitag NE. Listeria monocytogenes two component system PieRS regulates secretion chaperones PrsA1 and PrsA2 and enhances bacterial translocation across the intestine. Mol Microbiol 2022; 118:278-293. [PMID: 35943959 PMCID: PMC9545042 DOI: 10.1111/mmi.14967] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022]
Abstract
Listeria monocytogenes (Lm) is a widespread environmental Gram-positive bacterium that can transition into a pathogen following ingestion by a susceptible host. To cross host barriers and establish infection, Lm is dependent upon the regulated secretion and activity of many proteins including PrsA2, a peptidyl-prolyl cis-trans isomerase with foldase activity. PrsA2 contributes to the stability and activity of a number of secreted virulence factors that are required for Lm invasion, replication, and cell-to-cell spread within the infected host. In contrast, a second related secretion chaperone, PrsA1, has thus far no identified contributions to Lm pathogenesis. Here we describe the characterization of a two-component signal transduction system PieRS that regulates the expression of a regulon that includes the secretion chaperones PrsA1 and PrsA2. PieRS regulated gene products are required for bacterial resistance to ethanol exposure and are important for bacterial survival during transit through the gastrointestinal tract. PrsA1 was also found to make a unique contribution to Lm survival in the GI tract, revealing for the first time a non-overlapping requirement for both secretion chaperones PrsA1 and PrsA2 during the process of intra-gastric infection.
Collapse
Affiliation(s)
- Laty A. Cahoon
- Department of Microbiology and ImmunologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | | | - Avinash N. Gururaja
- Department of Microbiology and ImmunologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Sam H. Light
- Department of MicrobiologyUniversity of ChicagoChicagoIllinoisUSA
| | - Francis Alonzo
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
| | - Wayne F. Anderson
- Center for Genomics and Infectious Diseases, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Nancy E. Freitag
- Department of Microbiology and ImmunologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
31
|
Reich SJ, Stohr J, Goldbeck O, Fendrich B, Crauwels P, Riedel CU. Improved fluorescent Listeria spp. biosensors for analysis of antimicrobials by flow cytometry. Microbiologyopen 2022; 11:e1304. [PMID: 36031957 PMCID: PMC9245168 DOI: 10.1002/mbo3.1304] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/17/2022] [Indexed: 01/14/2023] Open
Abstract
The global increase in antibiotic resistance of pathogenic microorganisms requires the identification and characterization of novel antimicrobials. Bacterial biosensors expressing fluorescent proteins such as pHluorin variants are suitable for high-throughput screenings. Here, we present Listeria spp. pH-sensitive biosensors with improved fluorescence for single-cell analysis of antimicrobials by flow cytometry.
Collapse
Affiliation(s)
| | - Jonas Stohr
- Institute of Microbiology and BiotechnologyUniversity of UlmUlmGermany
| | - Oliver Goldbeck
- Institute of Microbiology and BiotechnologyUniversity of UlmUlmGermany
| | - Bastian Fendrich
- Institute of Microbiology and BiotechnologyUniversity of UlmUlmGermany
| | - Peter Crauwels
- Institute of Microbiology and BiotechnologyUniversity of UlmUlmGermany
| | | |
Collapse
|
32
|
Krause AL, Stinear TP, Monk IR. Barriers to genetic manipulation of Enterococci: Current Approaches and Future Directions. FEMS Microbiol Rev 2022; 46:6650352. [PMID: 35883217 PMCID: PMC9779914 DOI: 10.1093/femsre/fuac036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 01/09/2023] Open
Abstract
Enterococcus faecalis and Enterococcus faecium are Gram-positive commensal gut bacteria that can also cause fatal infections. To study clinically relevant multi-drug resistant E. faecalis and E. faecium strains, methods are needed to overcome physical (thick cell wall) and enzymatic barriers that limit the transfer of foreign DNA and thus prevent facile genetic manipulation. Enzymatic barriers to DNA uptake identified in E. faecalis and E. faecium include type I, II and IV restriction modification systems and CRISPR-Cas. This review examines E. faecalis and E. faecium DNA defence systems and the methods with potential to overcome these barriers. DNA defence system bypass will allow the application of innovative genetic techniques to expedite molecular-level understanding of these important, but somewhat neglected, pathogens.
Collapse
Affiliation(s)
- Alexandra L Krause
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, VIC 3000 Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, VIC 3000 Australia
| | - Ian R Monk
- Corresponding author: Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, VIC 3000 Australia. E-mail:
| |
Collapse
|
33
|
Molecular insights into intra-complex signal transmission during stressosome activation. Commun Biol 2022; 5:621. [PMID: 35760945 PMCID: PMC9237128 DOI: 10.1038/s42003-022-03549-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
The stressosome is a pseudo-icosahedral megadalton bacterial stress-sensing protein complex consisting of several copies of two STAS-domain proteins, RsbR and RsbS, and the kinase RsbT. Upon perception of environmental stress multiple copies of RsbT are released from the surface of the stressosome. Free RsbT activates downstream proteins to elicit a global cellular response, such as the activation of the general stress response in Gram-positive bacteria. The molecular events triggering RsbT release from the stressosome surface remain poorly understood. Here we present the map of Listeria innocua RsbR1/RsbS complex at resolutions of 3.45 Å for the STAS domain core in icosahedral symmetry and of 3.87 Å for the STAS domain and N-terminal sensors in D2 symmetry, respectively. The structure reveals a conformational change in the STAS domain linked to phosphorylation in RsbR. Docking studies indicate that allosteric RsbT binding to the conformationally flexible N-terminal sensor domain of RsbR affects the affinity of RsbS towards RsbT. Our results bring to focus the molecular events within the stressosome complex and further our understanding of this ubiquitous signaling hub. Cryo-EM structures of the stress-sensing complex in Listeria innocua reveal conformational changes that initiate the signaling response to environmental stress.
Collapse
|
34
|
Guerreiro DN, Wu J, McDermott E, Garmyn D, Dockery P, Boyd A, Piveteau P, O’Byrne CP. In Vitro Evolution of Listeria monocytogenes Reveals Selective Pressure for Loss of SigB and AgrA Function at Different Incubation Temperatures. Appl Environ Microbiol 2022; 88:e0033022. [PMID: 35583325 PMCID: PMC9195950 DOI: 10.1128/aem.00330-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/15/2022] [Indexed: 12/15/2022] Open
Abstract
The alternative sigma factor B (σB) contributes to the stress tolerance of the foodborne pathogen Listeria monocytogenes by upregulating the general stress response. We previously showed that σB loss-of-function mutations arise frequently in strains of L. monocytogenes and suggested that mild stresses might favor the selection of such mutations. In this study, we performed in vitro evolution experiments (IVEE) where L. monocytogenes was allowed to evolve over 30 days at elevated (42°C) or lower (30°C) incubation temperatures. Isolates purified throughout the IVEE revealed the emergence of sigB operon mutations at 42°C. However, at 30°C, independent alleles in the agr locus arose, resulting in the inactivation of Agr quorum sensing. Colonies of both sigB mutants and agr mutants exhibited a greyer coloration on 7-days-old agar plates than those of the parental strain. Scanning electron microscopy revealed a more complex colony architecture in the wild type than in the mutant strains. sigB mutant strains outcompeted the parental strain at 42°C but not at 30°C, while agr mutant strains showed a small increase in competitive fitness at 30°C. Analysis of 40,080 L. monocytogenes publicly available genome sequences revealed a high occurrence rate of premature stop codons in both the sigB and agrCA loci. An analysis of a local L. monocytogenes strain collection revealed 5 out of 168 strains carrying agrCA alleles. Our results suggest that the loss of σB or Agr confer an increased competitive fitness in some specific conditions and this likely contributes to the emergence of these alleles in strains of L. monocytogenes. IMPORTANCE To withstand environmental aggressions, L. monocytogenes upregulates a large regulon through the action of the alternative sigma factor B (σB). However, σB becomes detrimental for L. monocytogenes growth under mild stresses, which confer a competitive advantage to σB loss-of-function alleles. Temperatures of 42°C, a mild stress, are often employed in mutagenesis protocols of L. monocytogenes and promote the emergence of σB loss-of-function alleles in the sigB operon. In contrast, lower temperatures of 30°C promote the emergence of Agr loss-of-function alleles, a cell-cell communication mechanism in L. monocytogenes. Our findings demonstrate that loss-of-function alleles emerge spontaneously in laboratory-grown strains. These alleles rise in the population as a consequence of the trade-off between growth and survival imposed by the activation of σB in L. monocytogenes. Additionally, our results demonstrate the importance of identifying unwanted hitchhiker mutations in newly constructed mutant strains.
Collapse
Affiliation(s)
- Duarte N. Guerreiro
- Bacterial Stress Response Group, Microbiology, Ryan Institute, School of Biological and Chemical Sciences, National University of Ireland, Galway, Ireland
| | - Jialun Wu
- Bacterial Stress Response Group, Microbiology, Ryan Institute, School of Biological and Chemical Sciences, National University of Ireland, Galway, Ireland
| | - Emma McDermott
- Centre for Microscopy and Imaging, Anatomy School of Medicine, National University of Ireland, Galway, Ireland
| | - Dominique Garmyn
- Agroécologie, AgroSup Dijon, INRAE, University Bourgogne, University Bourgogne Franche-Comté, Dijon, France
| | - Peter Dockery
- Centre for Microscopy and Imaging, Anatomy School of Medicine, National University of Ireland, Galway, Ireland
| | - Aoife Boyd
- Pathogenic Mechanisms Research Group, Microbiology, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | | | - Conor P. O’Byrne
- Bacterial Stress Response Group, Microbiology, Ryan Institute, School of Biological and Chemical Sciences, National University of Ireland, Galway, Ireland
| |
Collapse
|
35
|
Wang M, Wamp S, Gibhardt J, Holland G, Schwedt I, Schmidtke KU, Scheibner K, Halbedel S, Commichau FM. Adaptation of Listeria monocytogenes to perturbation of c-di-AMP metabolism underpins its role in osmoadaptation and identifies a fosfomycin uptake system. Environ Microbiol 2022; 24:4466-4488. [PMID: 35688634 DOI: 10.1111/1462-2920.16084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022]
Abstract
The human pathogen Listeria monocytogenes synthesizes and degrades c-di-AMP using the diadenylate cyclase CdaA and the phosphodiesterases PdeA and PgpH respectively. c-di-AMP is essential because it prevents the uncontrolled uptake of osmolytes. Here, we studied the phenotypes of cdaA, pdeA, pgpH and pdeA pgpH mutants with defects in c-di-AMP metabolism and characterized suppressor mutants restoring their growth defects. The characterization of the pdeA pgpH mutant revealed that the bacteria show growth defects in defined medium, a phenotype that is invariably suppressed by mutations in cdaA. The previously reported growth defect of the cdaA mutant in rich medium is suppressed by mutations that osmotically stabilize the c-di-AMP-free strain. We also found that the cdaA mutant has an increased sensitivity against isoleucine. The isoleucine-dependent growth inhibition of the cdaA mutant is suppressed by codY mutations that likely reduce the DNA-binding activity of encoded CodY variants. Moreover, the characterization of the cdaA suppressor mutants revealed that the Opp oligopeptide transport system is involved in the uptake of the antibiotic fosfomycin. In conclusion, the suppressor analysis corroborates a key function of c-di-AMP in controlling osmolyte homeostasis in L. monocytogenes.
Collapse
Affiliation(s)
- Mengyi Wang
- FG Synthetic Microbiology, Institute for Biotechnology, BTU Cottbus-Senftenberg, 01968, Senftenberg, Germany.,Department of General Microbiology, Institute for Microbiology and Genetics, University of Goettingen, 37077, Göttingen, Germany.,FG Molecular Microbiology, Institute of Biology, University of Hohenheim, 70599, Stuttgart, Germany
| | - Sabrina Wamp
- Division of Enteropathogenic Bacteria and Legionella, Robert-Koch-Institute, 38855, Wernigerode, Germany
| | - Johannes Gibhardt
- FG Synthetic Microbiology, Institute for Biotechnology, BTU Cottbus-Senftenberg, 01968, Senftenberg, Germany.,Department of General Microbiology, Institute for Microbiology and Genetics, University of Goettingen, 37077, Göttingen, Germany.,Research Complex NanoBio, Peter the Great Saint Petersburg Polytechnic University, Politekhnicheskaya ulitsa 29A, Saint Petersburg, 195251, Russia
| | - Gudrun Holland
- ZBS4 - Advanced Light and Electron Microscopy, Robert-Koch-Institute, Seestraße 10, 13353, Berlin, Germany
| | - Inge Schwedt
- FG Synthetic Microbiology, Institute for Biotechnology, BTU Cottbus-Senftenberg, 01968, Senftenberg, Germany.,FG Molecular Microbiology, Institute of Biology, University of Hohenheim, 70599, Stuttgart, Germany
| | - Kai-Uwe Schmidtke
- FG Enzyme Technology, Institute for Biotechnology, BTU Cottbus-Senftenberg, 01968, Senftenberg, Germany
| | - Katrin Scheibner
- FG Enzyme Technology, Institute for Biotechnology, BTU Cottbus-Senftenberg, 01968, Senftenberg, Germany
| | - Sven Halbedel
- Division of Enteropathogenic Bacteria and Legionella, Robert-Koch-Institute, 38855, Wernigerode, Germany
| | - Fabian M Commichau
- FG Synthetic Microbiology, Institute for Biotechnology, BTU Cottbus-Senftenberg, 01968, Senftenberg, Germany.,FG Molecular Microbiology, Institute of Biology, University of Hohenheim, 70599, Stuttgart, Germany
| |
Collapse
|
36
|
Wu J, NicAogáin K, McAuliffe O, Jordan K, O’Byrne C. Phylogenetic and Phenotypic Analyses of a Collection of Food and Clinical Listeria monocytogenes Isolates Reveal Loss of Function of Sigma B from Several Clonal Complexes. Appl Environ Microbiol 2022; 88:e0005122. [PMID: 35481758 PMCID: PMC9128516 DOI: 10.1128/aem.00051-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/17/2022] [Indexed: 12/14/2022] Open
Abstract
To understand the molecular mechanisms that contribute to the stress responses of the important foodborne pathogen Listeria monocytogenes, we collected 139 strains (meat, n = 25; dairy, n = 10; vegetable, n = 8; seafood, n = 14; mixed food, n = 4; and food processing environments, n = 78), mostly isolated in Ireland, and subjected them to whole-genome sequencing. These strains were compared to 25 Irish clinical isolates and 4 well-studied reference strains. Core genome and pan-genome analysis confirmed a highly clonal and deeply branched population structure. Multilocus sequence typing showed that this collection contained a diverse range of strains from L. monocytogenes lineages I and II. Several groups of isolates with highly similar genome content were traced to single or multiple food business operators, providing evidence of strain persistence or prevalence, respectively. Phenotypic screening assays for tolerance to salt stress and resistance to acid stress revealed variants within several clonal complexes that were phenotypically distinct. Five of these phenotypic outliers were found to carry mutations in the sigB operon, which encodes the stress-inducible sigma factor sigma B. Transcriptional analysis confirmed that three of the strains that carried mutations in sigB, rsbV, or rsbU had reduced SigB activity, as predicted. These strains exhibited increased tolerance to salt stress and displayed decreased resistance to low pH stress. Overall, this study shows that loss-of-function mutations in the sigB operon are comparatively common in field isolates, probably reflecting the cost of the general stress response to reproductive fitness in this pathogen. IMPORTANCE The bacterial foodborne pathogen Listeria monocytogenes frequently contaminates various categories of food products and is able to cause life-threatening infections when ingested by humans. Thus, it is important to control the growth of this bacterium in food by understanding the mechanisms that allow its proliferation under suboptimal conditions. In this study, intraspecies heterogeneity in stress response was observed across a collection consisting of mainly Irish L. monocytogenes isolates. Through comparisons of genome sequence and phenotypes observed, we identified three strains with impairment of the general stress response regulator SigB. Two of these strains are used widely in food challenge studies for evaluating the growth potential of L. monocytogenes. Given that loss of SigB function is associated with atypical phenotypic properties, the use of these strains in food challenge studies should be re-evaluated.
Collapse
Affiliation(s)
- Jialun Wu
- Bacterial Stress Response Group, Microbiology, School of Biological and Chemical Sciences, National University of Ireland, Galway, Ireland
| | - Kerrie NicAogáin
- Bacterial Stress Response Group, Microbiology, School of Biological and Chemical Sciences, National University of Ireland, Galway, Ireland
| | | | - Kieran Jordan
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Conor O’Byrne
- Bacterial Stress Response Group, Microbiology, School of Biological and Chemical Sciences, National University of Ireland, Galway, Ireland
| |
Collapse
|
37
|
Wamp S, Rothe P, Stern D, Holland G, Döhling J, Halbedel S. MurA escape mutations uncouple peptidoglycan biosynthesis from PrkA signaling. PLoS Pathog 2022; 18:e1010406. [PMID: 35294506 PMCID: PMC8959180 DOI: 10.1371/journal.ppat.1010406] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 03/28/2022] [Accepted: 02/28/2022] [Indexed: 01/23/2023] Open
Abstract
Gram-positive bacteria are protected by a thick mesh of peptidoglycan (PG) completely engulfing their cells. This PG network is the main component of the bacterial cell wall, it provides rigidity and acts as foundation for the attachment of other surface molecules. Biosynthesis of PG consumes a high amount of cellular resources and therefore requires careful adjustments to environmental conditions. An important switch in the control of PG biosynthesis of Listeria monocytogenes, a Gram-positive pathogen with a high infection fatality rate, is the serine/threonine protein kinase PrkA. A key substrate of this kinase is the small cytosolic protein ReoM. We have shown previously that ReoM phosphorylation regulates PG formation through control of MurA stability. MurA catalyzes the first step in PG biosynthesis and the current model suggests that phosphorylated ReoM prevents MurA degradation by the ClpCP protease. In contrast, conditions leading to ReoM dephosphorylation stimulate MurA degradation. How ReoM controls degradation of MurA and potential other substrates is not understood. Also, the individual contribution of the ~20 other known PrkA targets to PG biosynthesis regulation is unknown. We here present murA mutants which escape proteolytic degradation. The release of MurA from ClpCP-dependent proteolysis was able to activate PG biosynthesis and further enhanced the intrinsic cephalosporin resistance of L. monocytogenes. This latter effect required the RodA3/PBP B3 transglycosylase/transpeptidase pair. One murA escape mutation not only fully rescued an otherwise non-viable prkA mutant during growth in batch culture and inside macrophages but also overcompensated cephalosporin hypersensitivity. Our data collectively indicate that the main purpose of PrkA-mediated signaling in L. monocytogenes is control of MurA stability during standard laboratory growth conditions and intracellular growth in macrophages. These findings have important implications for the understanding of PG biosynthesis regulation and β-lactam resistance of L. monocytogenes and related Gram-positive bacteria. Peptidoglycan (PG) is the main component of the bacterial cell wall and many of the PG synthesizing enzymes are antibiotic targets. We previously have discovered a new signaling route controlling PG production in the human pathogen Listeria monocytogenes. This route also determines the intrinsic resistance of L. monocytogenes against cephalosporins, a group of β-lactam antibiotics. Signaling involves PrkA, a membrane-embedded protein kinase, that is activated during cell wall stress to phosphorylate its target ReoM. Depending on its phosphorylation, ReoM activates or inactivates PG production by controlling the proteolytic stability of MurA, which catalyzes the first step in PG biosynthesis. MurA degradation depends on the ClpCP protease and we here have isolated murA mutations that escape this degradation. Using these mutants, we could show that regulation of PG biosynthesis through control of MurA stability is an important purpose of PrkA-mediated signaling in L. monocytogenes. Further experiments identified the transglycosylase RodA and the transpeptidase PBP B3 as additional downstream factors. Our results suggest that both proteins act together to translate the signals received by PrkA into adjustment of PG biosynthesis. These findings shed new light on the regulation of PG biosynthesis in Gram-positive bacteria with intrinsic β-lactam resistance.
Collapse
Affiliation(s)
- Sabrina Wamp
- FG11 - Division of Enteropathogenic bacteria and Legionella, Robert Koch Institute, Wernigerode, Germany
| | - Patricia Rothe
- FG11 - Division of Enteropathogenic bacteria and Legionella, Robert Koch Institute, Wernigerode, Germany
| | - Daniel Stern
- ZBS3 - Biological Toxins, Robert Koch Institute, Berlin, Germany
| | - Gudrun Holland
- ZBS4 - Advanced Light and Electron Microscopy, Robert Koch Institute, Berlin, Germany
| | - Janina Döhling
- FG11 - Division of Enteropathogenic bacteria and Legionella, Robert Koch Institute, Wernigerode, Germany
| | - Sven Halbedel
- FG11 - Division of Enteropathogenic bacteria and Legionella, Robert Koch Institute, Wernigerode, Germany
- * E-mail:
| |
Collapse
|
38
|
Maudet C, Kheloufi M, Levallois S, Gaillard J, Huang L, Gaultier C, Tsai YH, Disson O, Lecuit M. Bacterial inhibition of Fas-mediated killing promotes neuroinvasion and persistence. Nature 2022; 603:900-906. [PMID: 35296858 DOI: 10.1038/s41586-022-04505-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/03/2022] [Indexed: 12/19/2022]
Abstract
Infections of the central nervous system are among the most serious infections1,2, but the mechanisms by which pathogens access the brain remain poorly understood. The model microorganism Listeria monocytogenes (Lm) is a major foodborne pathogen that causes neurolisteriosis, one of the deadliest infections of the central nervous system3,4. Although immunosuppression is a well-established host risk factor for neurolisteriosis3,5, little is known about the bacterial factors that underlie the neuroinvasion of Lm. Here we develop a clinically relevant experimental model of neurolisteriosis, using hypervirulent neuroinvasive strains6 inoculated in a humanized mouse model of infection7, and we show that the bacterial surface protein InlB protects infected monocytes from Fas-mediated cell death by CD8+ T cells in a manner that depends on c-Met, PI3 kinase and FLIP. This blockade of specific anti-Lm cellular immune killing lengthens the lifespan of infected monocytes, and thereby favours the transfer of Lm from infected monocytes to the brain. The intracellular niche that is created by InlB-mediated cell-autonomous immune resistance also promotes Lm faecal shedding, which accounts for the selection of InlB as a core virulence gene of Lm. We have uncovered a specific mechanism by which a bacterial pathogen confers an increased lifespan to the cells it infects by rendering them resistant to cell-mediated immunity. This promotes the persistence of Lm within the host, its dissemination to the central nervous system and its transmission.
Collapse
Affiliation(s)
- Claire Maudet
- Institut Pasteur, Université de Paris, Inserm U1117, Biology of Infection Unit, Paris, France
| | - Marouane Kheloufi
- Institut Pasteur, Université de Paris, Inserm U1117, Biology of Infection Unit, Paris, France
| | - Sylvain Levallois
- Institut Pasteur, Université de Paris, Inserm U1117, Biology of Infection Unit, Paris, France
| | - Julien Gaillard
- Institut Pasteur, Université de Paris, Inserm U1117, Biology of Infection Unit, Paris, France
| | - Lei Huang
- Institut Pasteur, Université de Paris, Inserm U1117, Biology of Infection Unit, Paris, France
| | - Charlotte Gaultier
- Institut Pasteur, Université de Paris, Inserm U1117, Biology of Infection Unit, Paris, France
| | - Yu-Huan Tsai
- Institut Pasteur, Université de Paris, Inserm U1117, Biology of Infection Unit, Paris, France.,Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Olivier Disson
- Institut Pasteur, Université de Paris, Inserm U1117, Biology of Infection Unit, Paris, France
| | - Marc Lecuit
- Institut Pasteur, Université de Paris, Inserm U1117, Biology of Infection Unit, Paris, France. .,Institut Pasteur, National Reference Center and WHO Collaborating Center Listeria, Paris, France. .,Necker-Enfants Malades University Hospital, Division of Infectious Diseases and Tropical Medicine, APHP, Institut Imagine, Paris, France.
| |
Collapse
|
39
|
Guerreiro DN, Pucciarelli MG, Tiensuu T, Gudynaite D, Boyd A, Johansson J, García-del Portillo F, O’Byrne CP. Acid stress signals are integrated into the σB-dependent general stress response pathway via the stressosome in the food-borne pathogen Listeria monocytogenes. PLoS Pathog 2022; 18:e1010213. [PMID: 35275969 PMCID: PMC8942246 DOI: 10.1371/journal.ppat.1010213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/23/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
The general stress response (GSR) in Listeria monocytogenes plays a critical role in the survival of this pathogen in the host gastrointestinal tract. The GSR is regulated by the alternative sigma factor B (σB), whose role in protection against acid stress is well established. Here, we investigated the involvement of the stressosome, a sensory hub, in transducing low pH signals to induce the GSR. Mild acid shock (15 min at pH 5.0) activated σB and conferred protection against a subsequent lethal pH challenge. A mutant strain where the stressosome subunit RsbR1 was solely present retained the ability to induce σB activity at pH 5.0. The role of stressosome phosphorylation in signal transduction was investigated by mutating the putative phosphorylation sites in the core stressosome proteins RsbR1 (rsbR1-T175A, -T209A, -T241A) and RsbS (rsbS-S56A), or the stressosome kinase RsbT (rsbT-N49A). The rsbS S56A and rsbT N49A mutations abolished the response to low pH. The rsbR1-T209A and rsbR1-T241A mutants displayed constitutive σB activity. Mild acid shock upregulates invasion genes inlAB and stimulates epithelial cell invasion, effects that were abolished in mutants with an inactive or overactive stressosome. Overall, the results show that the stressosome is required for acid-induced activation of σB in L. monocytogenes. Furthermore, they show that RsbR1 can function independently of its paralogues and signal transduction requires RsbT-mediated phosphorylation of RsbS on S56 and RsbR1 on T209 but not T175. These insights shed light on the mechanisms of signal transduction that activate the GSR in L. monocytogenes in response to acidic environments, and highlight the role this sensory process in the early stages of the infectious cycle. The stress sensing hub known as the stressosome, found in many bacterial and archaeal lineages, plays a crucial role in both stress tolerance and virulence in the food-borne pathogen Listeria monocytogenes. However, the mechanisms that lead to its activation and the subsequent activation of the general stress response have remained elusive. In this study, we examined the signal transduction mechanisms that operate in the stressosome in response to acid stress. We found that only one of the five putative sensory proteins present in L. monocytogenes, RsbR1, was required for effective transduction of acid tress signals. We further found that phosphorylation of RsbS and RsbR1, mediated by the RsbT kinase, is essential for signal transduction. Failure to phosphorylate RsbS on Serine 56 completely abolished acid sensing by the stressosome, which prevented the development of adaptive acid tolerance. The acid-induced activation of internalin gene expression was also abolished in mutants with defective stressosome signalling, suggesting a role for the stressosome in the invasion of host cells. Together the data provide new insights into the mechanisms that activate the stressosome in response to acid stress and highlight the role this sensory hub plays in virulence.
Collapse
Affiliation(s)
- Duarte N. Guerreiro
- Bacterial Stress Response Group, Microbiology, School of Biological and Chemical Sciences, National University of Ireland, Galway, Ireland
| | - M. Graciela Pucciarelli
- Laboratory of Intracellular Bacterial Pathogens, National Centre for Biotechnology (CNB)-CSIC, Madrid, Spain
- Department of Molecular Biology, Universidad Autónoma de Madrid, Centre of Molecular Biology ‘Severo Ochoa’ (CBMSO CSIC-UAM), Madrid, Spain
| | - Teresa Tiensuu
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre of Microbial Research, Umeå, Sweden
| | - Diana Gudynaite
- Bacterial Stress Response Group, Microbiology, School of Biological and Chemical Sciences, National University of Ireland, Galway, Ireland
| | - Aoife Boyd
- Pathogenic Mechanisms Research Group, Microbiology, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - Jörgen Johansson
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre of Microbial Research, Umeå, Sweden
| | | | - Conor P. O’Byrne
- Bacterial Stress Response Group, Microbiology, School of Biological and Chemical Sciences, National University of Ireland, Galway, Ireland
- * E-mail:
| |
Collapse
|
40
|
Cheng C, Han X, Xu J, Sun J, Li K, Han Y, Chen M, Song H. YjbH mediates the oxidative stress response and infection by regulating SpxA1 and the phosphoenolpyruvate-carbohydrate phosphotransferase system (PTS) in Listeria monocytogenes. Gut Microbes 2022; 13:1-19. [PMID: 33573432 PMCID: PMC7889195 DOI: 10.1080/19490976.2021.1884517] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The foodborne pathogen Listeria monocytogenes relies on its ability to fine-tune the expression of virulence factors and stress regulators in response to rapidly changing environments. Here, we reveal that YjbH, a putative thioredoxin family oxidoreductase, plays a pivotal role in bacterial adaption to oxidative stress and host infection. YjbH directly interacts with SpxA1, an ArsC family oxidative stress response regulator, and the deletion of YjbH compromised the oxidative stress tolerance of L. monocytogenes. Also, YjbH is required for the bacterial spread in host cells and proliferation in mouse organs, thereby contributing to virulence. Transcriptomic analysis of strains treated with Cd2+ revealed that most virulence genes and phosphoenolpyruvate-carbohydrate phosphotransferase system (PTS) genes were significantly downregulated in the absence of YjbH. However, YjbH inhibits PrfA expression when bacteria were grown in the media, suggesting that YjbH participates in regulating the virulence genes via a complicated regulatory network involving PrfA and PTS. Collectively, these findings provide a valuable model for clarifying the roles of thioredoxins from foodborne pathogens regarding improving survival in the external environment and, more importantly, successfully establishing infection within the host.
Collapse
Affiliation(s)
- Changyong Cheng
- College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Hangzhou, Zhejiang, P. R. China
| | - Xiao Han
- College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Hangzhou, Zhejiang, P. R. China
| | - Jiali Xu
- College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Hangzhou, Zhejiang, P. R. China
| | - Jing Sun
- College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Hangzhou, Zhejiang, P. R. China
| | - Kang Li
- College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Hangzhou, Zhejiang, P. R. China
| | - Yue Han
- College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Hangzhou, Zhejiang, P. R. China
| | - Mianmian Chen
- College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Hangzhou, Zhejiang, P. R. China
| | - Houhui Song
- College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Hangzhou, Zhejiang, P. R. China,CONTACT Houhui Song College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, China-Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Hangzhou, Zhejiang311300, P. R. China
| |
Collapse
|
41
|
Braverman J, Monk IR, Ge C, Westall GP, Stinear TP, Wakim LM. Staphylococcus aureus specific lung resident memory CD4 + Th1 cells attenuate the severity of influenza virus induced secondary bacterial pneumonia. Mucosal Immunol 2022; 15:783-796. [PMID: 35637249 PMCID: PMC9148937 DOI: 10.1038/s41385-022-00529-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/25/2022] [Accepted: 05/03/2022] [Indexed: 02/04/2023]
Abstract
Staphylococcus aureus is a major cause of severe pulmonary infections. The evolution of multi-drug resistant strains limits antibiotic treatment options. To date, all candidate vaccines tested have failed, highlighting the need for an increased understanding of the immunological requirements for effective S. aureus immunity. Using an S. aureus strain engineered to express a trackable CD4+ T cell epitope and a murine model of S. aureus pneumonia, we show strategies that lodge Th1 polarised bacterium specific CD4+ tissue resident memory T cells (Trm) in the lung can significantly attenuate the severity of S. aureus pneumonia. This contrasts natural infection of mice that fails to lodge CD4+ Trm cells along the respiratory tract or provide protection against re-infection, despite initially generating Th17 bacterium specific CD4+ T cell responses. Interestingly, lack of CD4+ Trm formation after natural infection in mice appears to be reflected in humans, where the frequency of S. aureus reactive CD4+ Trm cells in lung tissue is also low. Our findings reveal the protective capacity of S. aureus specific respiratory tract CD4+ Th1 polarised Trm cells and highlight the potential for targeting these cells in vaccines that aim to prevent the development of S. aureus pneumonia.
Collapse
Affiliation(s)
- Jessica Braverman
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Ian R. Monk
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Chenghao Ge
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia ,grid.12527.330000 0001 0662 3178School of Medicine, Tsinghua University, Beijing, China
| | - Glen P. Westall
- grid.1002.30000 0004 1936 7857Lung Transplant Service, Alfred Hospital, Melbourne, Victoria, Australia; Department of Medicine, Monash University, Melbourne, VIC Australia
| | - Timothy P. Stinear
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Linda M. Wakim
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| |
Collapse
|
42
|
Cross-genus Boot-up of Synthetic Bacteriophage in Staphylococcus aureus Using a New and Efficient DNA Transformation Method. Appl Environ Microbiol 2021; 88:e0148621. [PMID: 34818102 DOI: 10.1128/aem.01486-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen causing a wide range of infections and food poisoning in humans with antibiotic resistance, specifically to methicillin, compounding the problem. Bacteriophages (phages) provide an alternative treatment strategy, but only infect a limited number of circulating strains and may quickly become ineffective due to bacterial resistance. To overcome these obstacles, engineered phages have been proposed, but methods are needed for efficient transformation of large DNA molecules into S. aureus to boot-up (i.e., rescue) infectious phages. We present a new, efficient and reproducible DNA transformation method, NEST (Non-Electroporation Staphylococcus Transformation), for S. aureus to boot-up of purified phage genomic DNA (at least 150 kb in length tested) and whole yeast-assembled synthetic phage genomes. This method is a powerful new tool for transformation of DNA in S. aureus and will enable the rapid development of engineered therapeutic phages and phage cocktails against Gram-positive pathogens. Importance The continued emergence of antibiotic resistant bacterial pathogens has heightened the urgency for alternative antibacterial strategies. Phages provide an alternative treatment strategy, but are difficult to optimize. Synthetic biology approaches have been successfully used to construct and rescue genomes of model phages, but only in a limited number of highly transformable host species. In this study, we used a new, reproducible, and efficient transformation method to reconstitute a functional non-model Siphophage from a constructed synthetic genome. This method will facilitate not only the engineering of Staphylococcus and Enterococcus phages for therapeutic applications but also the engineering of Staphylococcus strains by enabling transformation of higher molecular weight DNA to introduce more complex modifications.
Collapse
|
43
|
Sun L, Rogiers G, Courtin P, Chapot-Chartier MP, Bierne H, Michiels CW. AsnB Mediates Amidation of Meso-Diaminopimelic Acid Residues in the Peptidoglycan of Listeria monocytogenes and Affects Bacterial Surface Properties and Host Cell Invasion. Front Microbiol 2021; 12:760253. [PMID: 34721369 PMCID: PMC8554201 DOI: 10.3389/fmicb.2021.760253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/24/2021] [Indexed: 11/30/2022] Open
Abstract
A mutant of Listeria monocytogenes ScottA with a transposon in the 5' untranslated region of the asnB gene was identified to be hypersensitive to the antimicrobial t-cinnamaldehyde. Here, we report the functional characterization of AsnB in peptidoglycan (PG) modification and intracellular infection. While AsnB of Listeria is annotated as a glutamine-dependent asparagine synthase, sequence alignment showed that this protein is closely related to a subset of homologs that catalyze the amidation of meso-diaminopimelic acid (mDAP) residues in the peptidoglycan of other bacterial species. Structural analysis of peptidoglycan from an asnB mutant, compared to that of isogenic wild-type (WT) and complemented mutant strains, confirmed that AsnB mediates mDAP amidation in L. monocytogenes. Deficiency in mDAP amidation caused several peptidoglycan- and cell surface-related phenotypes in the asnB mutant, including formation of shorter but thicker cells, susceptibility to lysozyme, loss of flagellation and motility, and a strong reduction in biofilm formation. In addition, the mutant showed reduced invasion of human epithelial JEG-3 and Caco-2 cells. Analysis by immunofluorescence microscopy revealed that asnB inactivation abrogated the proper display at the listerial surface of the invasion protein InlA, which normally gets cross-linked to mDAP via its LPXTG motif. Together, this work shows that AsnB of L. monocytogenes, like several of its homologs in related Gram-positive bacteria, mediates the amidation of mDAP residues in the peptidoglycan and, in this way, affects several cell wall and cell surface-related properties. It also for the first time implicates the amidation of peptidoglycan mDAP residues in cell wall anchoring of InlA and in bacterial virulence.
Collapse
Affiliation(s)
- Lei Sun
- Laboratory of Food Microbiology, Department of Microbial and Molecular Systems (M2S) and Leuven Food Science and Nutrition Research Center (LFoRCe), KU Leuven, Leuven, Belgium
| | - Gil Rogiers
- Laboratory of Food Microbiology, Department of Microbial and Molecular Systems (M2S) and Leuven Food Science and Nutrition Research Center (LFoRCe), KU Leuven, Leuven, Belgium
| | - Pascal Courtin
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | | | - Hélène Bierne
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Chris W Michiels
- Laboratory of Food Microbiology, Department of Microbial and Molecular Systems (M2S) and Leuven Food Science and Nutrition Research Center (LFoRCe), KU Leuven, Leuven, Belgium
| |
Collapse
|
44
|
Listeria monocytogenes requires the RsbX protein to prevent SigB-activation under non-stressed conditions. J Bacteriol 2021; 204:e0048621. [PMID: 34694900 PMCID: PMC8765406 DOI: 10.1128/jb.00486-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The survival of microbial cells under changing environmental conditions requires an efficient reprogramming of transcription, often mediated by alternative sigma factors. The Gram-positive human pathogen Listeria monocytogenes senses and responds to environmental stress mainly through the alternative sigma factor σB (SigB), which controls expression of the general stress response regulon. SigB activation is achieved through a complex series of phosphorylation/dephosphorylation events culminating in the release of SigB from its anti-sigma factor RsbW. At the top of the signal transduction pathway lies a large multiprotein complex known as the stressosome that is believed to act as a sensory hub for stresses. Following signal detection, stressosome proteins become phosphorylated. Resetting of the stressosome is hypothesized to be exerted by a putative phosphatase, RsbX, which presumably removes phosphate groups from stressosome proteins poststress. We addressed the role of the RsbX protein in modulating the activity of the stressosome and consequently regulating SigB activity in L. monocytogenes. We show that RsbX is required to reduce SigB activation levels under nonstress conditions and that it is required for appropriate SigB-mediated stress adaptation. A strain lacking RsbX displayed impaired motility and biofilm formation and also an increased survival at low pH. Our results could suggest that absence of RsbX alters the multiprotein composition of the stressosome without dramatically affecting its phosphorylation status. Overall, the data show that RsbX plays a critical role in modulating the signal transduction pathway by blocking SigB activation under nonstressed conditions. IMPORTANCE Pathogenic bacteria need to sense and respond to stresses to survive harsh environments and also to turn off the response when no longer facing stress. Activity of the stress sigma factor SigB in the human pathogen Listeria monocytogenes is controlled by a hierarchic system having a large stress-sensing multiprotein complex known as the stressosome at the top. Following stress exposure, proteins in the stressosome become phosphorylated, leading to SigB activation. We have studied the role of a putative phosphatase, RsbX, which is hypothesized to dephosphorylate stressosome proteins. RsbX is critical not only to switch off the stress response poststress but also to keep the activity of SigB low at nonstressed conditions to prevent unnecessary gene expression and save energy.
Collapse
|
45
|
Kelliher JL, Grunenwald CM, Abrahams RR, Daanen ME, Lew CI, Rose WE, Sauer JD. PASTA kinase-dependent control of peptidoglycan synthesis via ReoM is required for cell wall stress responses, cytosolic survival, and virulence in Listeria monocytogenes. PLoS Pathog 2021; 17:e1009881. [PMID: 34624065 PMCID: PMC8528326 DOI: 10.1371/journal.ppat.1009881] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/20/2021] [Accepted: 09/27/2021] [Indexed: 02/01/2023] Open
Abstract
Pathogenic bacteria rely on protein phosphorylation to adapt quickly to stress, including that imposed by the host during infection. Penicillin-binding protein and serine/threonine-associated (PASTA) kinases are signal transduction systems that sense cell wall integrity and modulate multiple facets of bacterial physiology in response to cell envelope stress. The PASTA kinase in the cytosolic pathogen Listeria monocytogenes, PrkA, is required for cell wall stress responses, cytosolic survival, and virulence, yet its substrates and downstream signaling pathways remain incompletely defined. We combined orthogonal phosphoproteomic and genetic analyses in the presence of a β-lactam antibiotic to define PrkA phosphotargets and pathways modulated by PrkA. These analyses synergistically highlighted ReoM, which was recently identified as a PrkA target that influences peptidoglycan (PG) synthesis, as an important phosphosubstrate during cell wall stress. We find that deletion of reoM restores cell wall stress sensitivities and cytosolic survival defects of a ΔprkA mutant to nearly wild-type levels. While a ΔprkA mutant is defective for PG synthesis during cell wall stress, a double ΔreoM ΔprkA mutant synthesizes PG at rates similar to wild type. In a mouse model of systemic listeriosis, deletion of reoM in a ΔprkA background almost fully restored virulence to wild-type levels. However, loss of reoM alone also resulted in attenuated virulence, suggesting ReoM is critical at some points during pathogenesis. Finally, we demonstrate that the PASTA kinase/ReoM cell wall stress response pathway is conserved in a related pathogen, methicillin-resistant Staphylococcus aureus. Taken together, our phosphoproteomic analysis provides a comprehensive overview of the PASTA kinase targets of an important model pathogen and suggests that a critical role of PrkA in vivo is modulating PG synthesis through regulation of ReoM to facilitate cytosolic survival and virulence. Many antibiotics target bacterial cell wall biosynthesis, justifying continued study of this process and the ways bacteria respond to cell wall insults during infection. Penicillin-binding protein and serine/threonine-associated (PASTA) kinases are master regulators of cell wall stress responses in bacteria and are conserved in several major pathogens, including Listeria monocytogenes, Staphylococcus aureus, and Mycobacterium tuberculosis. We previously showed that the PASTA kinase in L. monocytogenes, PrkA, is essential for the response to cell wall stress and for virulence. In this work, we combined proteomic and genetic approaches to identify PrkA substrates in L. monocytogenes. We show that regulation of one candidate from both screens, ReoM, increases synthesis of the cell wall component peptidoglycan and that this regulation is required for pathogenesis. We also demonstrate that the PASTA kinase-ReoM pathway regulates cell wall stress responses in another significant pathogen, methicillin-resistant S. aureus. Additionally, we uncover a PrkA-independent role for ReoM in vivo in L. monocytogenes, suggesting a need for nuanced modulation of peptidoglycan synthesis during infection. Cumulatively, this study provides new insight into how bacterial pathogens control cell wall synthesis during infection.
Collapse
Affiliation(s)
- Jessica L. Kelliher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Caroline M. Grunenwald
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Rhiannon R. Abrahams
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - McKenzie E. Daanen
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Cassandra I. Lew
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Warren E. Rose
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 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
|
46
|
Establishing recombinant production of pediocin PA-1 in Corynebacterium glutamicum. Metab Eng 2021; 68:34-45. [PMID: 34492380 PMCID: PMC8593837 DOI: 10.1016/j.ymben.2021.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/30/2021] [Accepted: 09/01/2021] [Indexed: 11/21/2022]
Abstract
Bacteriocins are antimicrobial peptides produced by bacteria to inhibit competitors in their natural environments. Some of these peptides have emerged as commercial food preservatives and, due to the rapid increase in antibiotic resistant bacteria, are also discussed as interesting alternatives to antibiotics for therapeutic purposes. Currently, commercial bacteriocins are produced exclusively with natural producer organisms on complex substrates and are sold as semi-purified preparations or crude fermentates. To allow clinical application, efficacy of production and purity of the product need to be improved. This can be achieved by shifting production to recombinant microorganisms. Here, we identify Corynebacterium glutamicum as a suitable production host for the bacteriocin pediocin PA-1. C. glutamicum CR099 shows resistance to high concentrations of pediocin PA-1 and the bacteriocin was not inactivated when spiked into growing cultures of this bacterium. Recombinant C. glutamicum expressing a synthetic pedACDCgl operon releases a compound that has potent antimicrobial activity against Listeria monocytogenes and Listeria innocua and matches size and mass:charge ratio of commercial pediocin PA-1. Fermentations in shake flasks and bioreactors suggest that low levels of dissolved oxygen are favorable for production of pediocin. Under these conditions, however, reduced activity of the TCA cycle resulted in decreased availability of the important pediocin precursor l-asparagine suggesting options for further improvement. Overall, we demonstrate that C. glutamicum is a suitable host for recombinant production of bacteriocins of the pediocin family. C. glutamicum CR099 is resistant to high concentrations of pediocin PA-1. Recombinant C. glutamicum CR099/pEKEx-pedACDCg produces of a compound with antimicrobial activity against Listeria sp. The purified compound matches size and mass:charge ratio of commercial pediocin PA-1. Low oxygen levels are favorable for production of active pediocin by C. glutamicum in batch fermentations.
Collapse
|
47
|
Sun L, Rogiers G, Michiels CW. The Natural Antimicrobial trans-Cinnamaldehyde Interferes with UDP-N-Acetylglucosamine Biosynthesis and Cell Wall Homeostasis in Listeria monocytogenes. Foods 2021; 10:foods10071666. [PMID: 34359536 PMCID: PMC8307235 DOI: 10.3390/foods10071666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 01/07/2023] Open
Abstract
Trans-cinnamaldehyde (t-CIN), an antimicrobial compound from cinnamon essential oil, is of interest because it inhibits various foodborne pathogens. In the present work, we investigated the antimicrobial mechanisms of t-CIN in Listeria monocytogenes using a previously isolated yvcK::Himar1 transposon mutant which shows hypersensitivity to t-CIN. Time-lapse microscopy revealed that t-CIN induces a bulging cell shape followed by lysis in the mutant. Complementation with wild-type yvcK gene completely restored the tolerance of yvcK::Himar1 strain to t-CIN and the cell morphology. Suppressor mutants which partially reversed the t-CIN sensitivity of the yvcK::Himar1 mutant were isolated from evolutionary experiments. Three out of five suppression mutations were in the glmU-prs operon and in nagR, which are linked to the biosynthesis of the peptidoglycan precursor uridine-diphosphate-N-acetylglucosamine (UDP-GlcNAc). GlmU catalyzes the last two steps of UDP-GlcNAc biosynthesis and NagR represses the uptake and utilization of N-acetylglucosamine. Feeding N-acetylglucosamine or increasing the production of UDP-GlcNAc synthetic enzymes fully or partially restored the t-CIN tolerance of the yvcK mutant. Together, these results suggest that YvcK plays a pivotal role in diverting substrates to UDP-GlcNAc biosynthesis in L. monocytogenes and that t-CIN interferes with this pathway, leading to a peptidoglycan synthesis defect.
Collapse
|
48
|
Nowak J, Visnovsky SB, Pitman AR, Cruz CD, Palmer J, Fletcher GC, Flint S. Biofilm Formation by Listeria monocytogenes 15G01, a Persistent Isolate from a Seafood-Processing Plant, Is Influenced by Inactivation of Multiple Genes Belonging to Different Functional Groups. Appl Environ Microbiol 2021; 87:e02349-20. [PMID: 33741610 PMCID: PMC8117777 DOI: 10.1128/aem.02349-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/25/2021] [Indexed: 01/13/2023] Open
Abstract
Listeria monocytogenes is a ubiquitous foodborne pathogen that results in a high rate of mortality in sensitive and immunocompromised people. Contamination of food with L. monocytogenes is thought to occur during food processing, most often as a result of the pathogen producing a biofilm that persists in the environment and acting as the source for subsequent dispersal of cells onto food. A survey of seafood-processing plants in New Zealand identified the persistent strain 15G01, which has a high capacity to form biofilms. In this study, a transposon library of L. monocytogenes 15G01 was screened for mutants with altered biofilm formation, assessed by a crystal violet assay, to identify genes involved in biofilm formation. This screen identified 36 transposants that showed a significant change in biofilm formation compared to the wild type. The insertion sites were in 27 genes, 20 of which led to decreased biofilm formation and seven to an increase. Two insertions were in intergenic regions. Annotation of the genes suggested that they are involved in diverse cellular processes, including stress response, autolysis, transporter systems, and cell wall/membrane synthesis. Analysis of the biofilms produced by the transposants using scanning electron microscopy and fluorescence microscopy showed notable differences in the structure of the biofilms compared to the wild type. In particular, inactivation of uvrB and mltD produced coccoid-shaped cells and elongated cells in long chains, respectively, and the mgtB mutant produced a unique biofilm with a sandwich structure which was reversed to the wild-type level upon magnesium addition. The mltD transposant was successfully complemented with the wild-type gene, whereas the phenotypes were not or only partially restored for the remaining mutants.IMPORTANCE The major source of contamination of food with Listeria monocytogenes is thought to be due to biofilm formation and/or persistence in food-processing plants. By establishing as a biofilm, L. monocytogenes cells become harder to eradicate due to their increased resistance to environmental threats. Understanding the genes involved in biofilm formation and their influence on biofilm structure will help identify new ways to eliminate harmful biofilms in food processing environments. To date, multiple genes have been identified as being involved in biofilm formation by L. monocytogenes; however, the exact mechanism remains unclear. This study identified four genes associated with biofilm formation by a persistent strain. Extensive microscopic analysis illustrated the effect of the disruption of mgtB, clsA, uvrB, and mltD and the influence of magnesium on the biofilm structure. The results strongly suggest an involvement in biofilm formation for the four genes and provide a basis for further studies to analyze gene regulation to assess the specific role of these biofilm-associated genes.
Collapse
Affiliation(s)
- Jessika Nowak
- The New Zealand Institute for Plant and Food Research Limited, Auckland, New Zealand
- Institute of Food, Nutrition and Human Health, Massey University, Palmerston North, New Zealand
| | - Sandra B Visnovsky
- The New Zealand Institute for Plant and Food Research Limited, Lincoln, New Zealand
| | - Andrew R Pitman
- The Foundation for Arable Research, Christchurch, New Zealand
| | - Cristina D Cruz
- The New Zealand Institute for Plant and Food Research Limited, Auckland, New Zealand
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jon Palmer
- Institute of Food, Nutrition and Human Health, Massey University, Palmerston North, New Zealand
| | - Graham C Fletcher
- The New Zealand Institute for Plant and Food Research Limited, Auckland, New Zealand
| | - Steve Flint
- Institute of Food, Nutrition and Human Health, Massey University, Palmerston North, New Zealand
| |
Collapse
|
49
|
Elements in the LftR Repressor Operator Interface Contributing to Regulation of Aurantimycin Resistance in Listeria monocytogenes. J Bacteriol 2021; 203:JB.00503-20. [PMID: 33649145 DOI: 10.1128/jb.00503-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/18/2021] [Indexed: 12/12/2022] Open
Abstract
The bacterium Listeria monocytogenes ubiquitously occurs in the environment but can cause severe invasive disease in susceptible individuals when ingested. We recently identified the L. monocytogenes genes lieAB and lftRS, encoding a multidrug resistance ABC transporter and a regulatory module, respectively. These genes jointly mediate resistance against aurantimycin, an antibiotic produced by the soil-dwelling species Streptomyces aurantiacus, and thus contribute to the survival of L. monocytogenes in its natural habitat, the soil. Repression of lieAB and lftRS is exceptionally tight but strongly induced in the presence of aurantimycin. Repression depends on LftR, which belongs to subfamily 2 of the PadR-like transcriptional repressors. To better understand this interesting class of transcriptional repressors, we here deduce the LftR operator sequence from a systematic truncation and mutation analysis of the P lieAB promoter. The sequence identified is also present in the P lftRS promoter but not found elsewhere in the chromosome. Mutational analysis of the putative operator in the P lftRS promoter confirmed its relevance for LftR-dependent repression. The proposed operator sequence was sufficient for DNA binding by LftR in vitro, and a mutation in this sequence affected aurantimycin resistance. Our results provide further insights into the transcriptional adaptation of an important human pathogen to survive the conditions in its natural reservoir.IMPORTANCE Listeria monocytogenes is an environmental bacterium that lives in the soil but can infect humans upon ingestion, and this can lead to severe invasive disease. Adaptation to these entirely different habitats involves massive reprogramming of transcription. Among the differentially expressed genes is the lieAB operon, which encodes a transporter for the detoxification of aurantimycin, an antimicrobial compound produced by soil-dwelling competitors. While lieAB is important for survival in the environment, its expression is detrimental during infection. We here identify critical elements in the lieAB promoter and its transcriptional regulator LftR that contribute to habitat-specific expression of the lieAB genes. These results further clarify the molecular mechanisms underlying the aurantimycin resistance of L. monocytogenes.
Collapse
|
50
|
Listeria monocytogenes MenI Encodes a DHNA-CoA Thioesterase Necessary for Menaquinone Biosynthesis, Cytosolic Survival, and Virulence. Infect Immun 2021; 89:IAI.00792-20. [PMID: 33619030 DOI: 10.1128/iai.00792-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/13/2021] [Indexed: 12/14/2022] Open
Abstract
Listeria monocytogenes is a Gram-positive, intracellular pathogen that is highly adapted to invade and replicate in the cytosol of eukaryotic cells. Intermediate metabolites in the menaquinone biosynthesis pathway are essential for the cytosolic survival and virulence of L. monocytogenes, independent of the production of menaquinone (MK) and aerobic respiration. Determining which specific intermediate metabolite(s) are essential for cytosolic survival and virulence has been hindered by the lack of an identified 1,4-dihydroxy-2-naphthoyl-coenzyme A (DHNA-CoA) thioesterase essential for converting DHNA-CoA to DHNA in the MK synthesis pathway. Using the recently identified Escherichia coli DHNA-CoA thioesterase as a query, homology sequence analysis revealed a single homolog in L. monocytogenes, LMRG_02730 Genetic deletion of LMRG_02730 resulted in an ablated membrane potential, indicative of a nonfunctional electron transport chain (ETC) and an inability to aerobically respire. Biochemical kinetic analysis of LMRG_02730 revealed strong activity toward DHNA-CoA, similar to its E. coli homolog, further demonstrating that LMRG_02730 is a DHNA-CoA thioesterase. Functional analyses in vitro, ex vivo, and in vivo using mutants directly downstream and upstream of LMRG_02730 revealed that DHNA-CoA is sufficient to facilitate in vitro growth in minimal medium, intracellular replication, and plaque formation in fibroblasts. In contrast, protection against bacteriolysis in the cytosol of macrophages and tissue-specific virulence in vivo requires the production of 1,4-dihydroxy-2-naphthoate (DHNA). Taken together, these data implicate LMRG_02730 (renamed MenI) as a DHNA-CoA thioesterase and suggest that while DHNA, or an unknown downstream product of DHNA, protects the bacteria from killing in the macrophage cytosol, DHNA-CoA is necessary for intracellular bacterial replication.
Collapse
|