1
|
Li Z, Shao Y, Liu X, Wan X, Xiong P, Wang L, Yuan J. Steap3 is a key node in regulating the phagosome escape of Listeria monocytogenes. Mol Immunol 2025; 182:96-106. [PMID: 40252499 DOI: 10.1016/j.molimm.2025.04.006] [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: 02/13/2025] [Revised: 03/26/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Listeria monocytogenes, a foodborne pathogen, poses a significant threat to human health due to its high mortality rate and increasing antibiotic resistance. This study investigates the role of Steap3 in regulating the early phagosomal escape of Listeria monocytogenes. We found that Steap3 expression is downregulated in dendritic and intestinal epithelial cells following infection, and its deficiency exacerbates bacterial proliferation both in vitro and in vivo. Mechanistically, Steap3 interacts with Gm2a and Sting to inhibit Listeria monocytogenes infection. Our results highlight Steap3 as a key regulator in dendritic and intestinal epithelial cells' defense against Listeria monocytogenes infection, suggesting the Steap3-STING/Gm2a axis is a potential therapeutic target for listeriosis. This study provides valuable insights into the molecular mechanisms underlying Listeria monocytogenes pathogenesis and host immune response, offering new directions for developing anti-Listeria monocytogenes therapies.
Collapse
Affiliation(s)
- Zhangfu Li
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Yan Shao
- Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiao Liu
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Xiaoyuan Wan
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Pei Xiong
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China.
| | - Jiangbei Yuan
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China; Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Department of Infectious Diseases, Affiliated Banan Hospital of Chongqing Medical University, Chongqing 400016, China; School of Chemistry and Chemical Engineering, Jinggangshan University, Ji'an, Jiangxi 343009, China.
| |
Collapse
|
2
|
Fayoud H, Belousov MV, Antonets KS, Nizhnikov AA. Pathogenesis-Associated Bacterial Amyloids: The Network of Interactions. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2107-2132. [PMID: 39865026 DOI: 10.1134/s0006297924120022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 01/28/2025]
Abstract
Amyloids are protein fibrils with a characteristic cross-β structure that is responsible for the unusual resistance of amyloids to various physical and chemical factors, as well as numerous pathogenic and functional consequences of amyloidogenesis. The greatest diversity of functional amyloids was identified in bacteria. The majority of bacterial amyloids are involved in virulence and pathogenesis either via facilitating formation of biofilms and adaptation of bacteria to colonization of a host organism or through direct regulation of toxicity. Recent studies have shown that, beside their commonly known activity, amyloids may be involved in the spatial regulation of proteome by modulating aggregation of other amyloidogenic proteins with multiple functional or pathological effects. Although the studies on the role of microbiome-produced amyloids in the development of amyloidoses in humans and animals have only been started, it is clear that humans as holobionts contain amyloids encoded not only by the host genome, but also by microorganisms that constitute the microbiome. Amyloids acquired from external sources (e.g., food) can interact with holobiont amyloids and modulate the effects of bacterial and host amyloids, thus adding another level of complexity to the holobiont-associated amyloid network. In this review, we described bacterial amyloids directly or indirectly involved in disease pathogenesis in humans and discussed the significance of bacterial amyloids in the three-component network of holobiont-associated amyloids.
Collapse
Affiliation(s)
- Haidar Fayoud
- Faculty of Biology, St. Petersburg State University, St. Petersburg, 199034, Russia
- All-Russia Research Institute for Agricultural Microbiology, St. Petersburg, 196608, Russia
| | - Mikhail V Belousov
- Faculty of Biology, St. Petersburg State University, St. Petersburg, 199034, Russia
- All-Russia Research Institute for Agricultural Microbiology, St. Petersburg, 196608, Russia
| | - Kirill S Antonets
- Faculty of Biology, St. Petersburg State University, St. Petersburg, 199034, Russia
- All-Russia Research Institute for Agricultural Microbiology, St. Petersburg, 196608, Russia
| | - Anton A Nizhnikov
- Faculty of Biology, St. Petersburg State University, St. Petersburg, 199034, Russia. ARRAY(0x5ae2b7af6df8)
- All-Russia Research Institute for Agricultural Microbiology, St. Petersburg, 196608, Russia
| |
Collapse
|
3
|
Raab JE, Hamilton DJ, Harju TB, Huynh TN, Russo BC. Pushing boundaries: mechanisms enabling bacterial pathogens to spread between cells. Infect Immun 2024; 92:e0052423. [PMID: 38661369 PMCID: PMC11385730 DOI: 10.1128/iai.00524-23] [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] [Indexed: 04/26/2024] Open
Abstract
For multiple intracellular bacterial pathogens, the ability to spread directly into adjacent epithelial cells is an essential step for disease in humans. For pathogens such as Shigella, Listeria, Rickettsia, and Burkholderia, this intercellular movement frequently requires the pathogens to manipulate the host actin cytoskeleton and deform the plasma membrane into structures known as protrusions, which extend into neighboring cells. The protrusion is then typically resolved into a double-membrane vacuole (DMV) from which the pathogen quickly escapes into the cytosol, where additional rounds of intercellular spread occur. Significant progress over the last few years has begun to define the mechanisms by which intracellular bacterial pathogens spread. This review highlights the interactions of bacterial and host factors that drive mechanisms required for intercellular spread with a focus on how protrusion structures form and resolve.
Collapse
Affiliation(s)
- Julie E. Raab
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Desmond J. Hamilton
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Tucker B. Harju
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Thao N. Huynh
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Brian C. Russo
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| |
Collapse
|
4
|
Prislusky MI, Lam JGT, Contreras VR, Ng M, Chamberlain M, Pathak-Sharma S, Fields M, Zhang X, Amer AO, Seveau S. The septin cytoskeleton is required for plasma membrane repair. EMBO Rep 2024; 25:3870-3895. [PMID: 38969946 PMCID: PMC11387490 DOI: 10.1038/s44319-024-00195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 07/07/2024] Open
Abstract
Plasma membrane repair is a fundamental homeostatic process of eukaryotic cells. Here, we report a new function for the conserved cytoskeletal proteins known as septins in the repair of cells perforated by pore-forming toxins or mechanical disruption. Using a silencing RNA screen, we identified known repair factors (e.g. annexin A2, ANXA2) and novel factors such as septin 7 (SEPT7) that is essential for septin assembly. Upon plasma membrane injury, the septin cytoskeleton is extensively redistributed to form submembranous domains arranged as knob and loop structures containing F-actin, myosin IIA, S100A11, and ANXA2. Formation of these domains is Ca2+-dependent and correlates with plasma membrane repair efficiency. Super-resolution microscopy revealed that septins and F-actin form intertwined filaments associated with ANXA2. Depletion of SEPT7 prevented ANXA2 recruitment and formation of submembranous actomyosin domains. However, ANXA2 depletion had no effect on domain formation. Collectively, our data support a novel septin-based mechanism for resealing damaged cells, in which the septin cytoskeleton plays a key structural role in remodeling the plasma membrane by promoting the formation of SEPT/F-actin/myosin IIA/ANXA2/S100A11 repair domains.
Collapse
Affiliation(s)
- M Isabella Prislusky
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Jonathan G T Lam
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Viviana Ruiz Contreras
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
- Grupo Investigaciones Biomédicas, Universidad de Sucre, Sincelejo, Sucre, Colombia
| | - Marilynn Ng
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Madeline Chamberlain
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Sarika Pathak-Sharma
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Madalyn Fields
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Xiaoli Zhang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Amal O Amer
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Stephanie Seveau
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Guo P, Li Z, Cai T, Guo D, Yang B, Zhang C, Shan Z, Wang X, Peng X, Liu G, Shi C, Alharbi M, Alasmari AF. Inhibitory effect and mechanism of oregano essential oil on Listeria monocytogenes cells, toxins and biofilms. Microb Pathog 2024; 194:106801. [PMID: 39025378 DOI: 10.1016/j.micpath.2024.106801] [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: 04/04/2024] [Revised: 06/12/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Listeria monocytogenes (L. monocytogenes) is a prevalent foodborne pathogen with a remarkable capacity to form biofilms on utensil surfaces. The Listeriolysin O (LLO) exhibits hemolytic activity, which is responsible for causing human infections. In this study, we investigated the inhibitory effect and mechanism of oregano essential oil (OEO) on L. monocytogenes, evaluated the effects on its biofilm removal and hemolytic activity. The minimum inhibitory concentration (MIC) of OEO against L. monocytogenes was 0.03 % (v/v). L. monocytogenes was treated with OEO at 3/2 MIC for 30 min the bacteria was decreased below the detection limit (10 CFU/mL) in PBS and TSB (the initial bacterial load was about 6.5 log CFU/mL). The level of L. monocytogenes in minced pork co-cultured with OEO (15 MIC) about 2.5 log CFU/g lower than that in the untreated group. The inhibitory mechanisms of OEO against planktonic L. monocytogenes encompassed perturbation of cellular morphology, elevation in reactive oxygen species levels, augmentation of lipid oxidation extent, hyperpolarization of membrane potential, and reduction in intracellular ATP concentration. In addition, OEO reduced biofilm coverage on the surface of glass slides by 62.03 % compared with the untreated group. Meanwhile, OEO (1/8 MIC) treatment reduced the hemolytic activity of L. monocytogenes to 24.6 % compared with the positive control. Molecular docking suggested carvacrol and thymol might reduce the hemolytic activity of L. monocytogenes. The results of this study demonstrate that OEO exhibits inhibitory effects against L. monocytogenes, biofilms and LLO, which had potential as natural antimicrobial for the inhibition of L. monocytogenes.
Collapse
Affiliation(s)
- Peng Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhenye Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Ting Cai
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Du Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Baowei Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chunling Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhongguo Shan
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaoli Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Guorong Liu
- China Food Flavor and Nutrition Health Innovation Center, Beijing Technology and Business University, China.
| | - Chao Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China; Northwest A&F University ShenZhen Research Institute, Shenzhen, Guangdong, 518057, China.
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
6
|
Prislusky MI, Lam JG, Contreras VR, Ng M, Chamberlain M, Pathak-Sharma S, Fields M, Zhang X, Amer AO, Seveau S. The Septin Cytoskeleton is Required for Plasma Membrane Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.12.548547. [PMID: 37503091 PMCID: PMC10369955 DOI: 10.1101/2023.07.12.548547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Mammalian cells are frequently exposed to mechanical and biochemical stressors resulting in plasma membrane injuries. Repair mechanisms reseal the plasma membrane to restore homeostasis and prevent cell death. In the present work, a silencing RNA screen was performed to uncover plasma membrane repair mechanisms of cells exposed to a pore-forming toxin (listeriolysin O). This screen identified molecules previously known to repair the injured plasma membrane such as annexin A2 (ANXA2) as well as novel plasma membrane repair candidate proteins. Of the novel candidates, we focused on septin 7 (SEPT7) because the septins are an important family of conserved eukaryotic cytoskeletal proteins. Using diverse experimental approaches, we established for the first time that SEPT7 plays a general role in plasma membrane repair of cells perforated by pore-forming toxins and mechanical wounding. Remarkably, upon cell injury, the septin cytoskeleton is extensively redistributed in a Ca 2+ -dependent fashion, a hallmark of plasma membrane repair machineries. The septins reorganize into subplasmalemmal domains arranged as knob and loop (or ring) structures containing F-actin, myosin II, and annexin A2 (ANXA2) and protrude from the cell surface. Importantly, the formation of these domains correlates with the plasma membrane repair efficiency. Super-resolution microscopy shows that septins and actin are arranged in intertwined filaments associated with ANXA2. Silencing SEPT7 expression prevented the formation of the F-actin/myosin II/ANXA2 domains, however, silencing expression of ANXA2 had no observable effect on their formation. These results highlight the key structural role of the septins in remodeling the plasma membrane and in the recruitment of the repair molecule ANXA2. Collectively, our data support a novel model in which the septin cytoskeleton acts as a scaffold to promote the formation of plasma membrane repair domains containing contractile F-actin and annexin A2.
Collapse
|
7
|
Nozawa T, Toh H, Iibushi J, Kogai K, Minowa-Nozawa A, Satoh J, Ito S, Murase K, Nakagawa I. Rab41-mediated ESCRT machinery repairs membrane rupture by a bacterial toxin in xenophagy. Nat Commun 2023; 14:6230. [PMID: 37802980 PMCID: PMC10558455 DOI: 10.1038/s41467-023-42039-2] [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: 11/21/2022] [Accepted: 09/26/2023] [Indexed: 10/08/2023] Open
Abstract
Xenophagy, a type of selective autophagy, is a bactericidal membrane trafficking that targets cytosolic bacterial pathogens, but the membrane homeostatic system to cope with bacterial infection in xenophagy is not known. Here, we show that the endosomal sorting complexes required for transport (ESCRT) machinery is needed to maintain homeostasis of xenophagolysosomes damaged by a bacterial toxin, which is regulated through the TOM1L2-Rab41 pathway that recruits AAA-ATPase VPS4. We screened Rab GTPases and identified Rab41 as critical for maintaining the acidification of xenophagolysosomes. Confocal microscopy revealed that ESCRT components were recruited to the entire xenophagolysosome, and this recruitment was inhibited by intrabody expression against bacterial cytolysin, indicating that ESCRT targets xenophagolysosomes in response to a bacterial toxin. Rab41 translocates to damaged autophagic membranes via adaptor protein TOM1L2 and recruits VPS4 to complete ESCRT-mediated membrane repair in a unique GTPase-independent manner. Finally, we demonstrate that the TOM1L2-Rab41 pathway-mediated ESCRT is critical for the efficient clearance of bacteria through xenophagy.
Collapse
Affiliation(s)
- Takashi Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hirotaka Toh
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Junpei Iibushi
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Kohei Kogai
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Atsuko Minowa-Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Junko Satoh
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Kazunori Murase
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
8
|
Kolypetri S, Kostoglou D, Nikolaou A, Kourkoutas Y, Giaouris E. Chemical Composition, Antibacterial and Antibiofilm Actions of Oregano ( Origanum vulgare subsp. hirtum) Essential Oil against Salmonella Typhimurium and Listeria monocytogenes. Foods 2023; 12:2893. [PMID: 37569162 PMCID: PMC10418746 DOI: 10.3390/foods12152893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/29/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Essential oils (EOs) are plant mixtures that are known to present strong bioactivities, including a wide antimicrobial action. Biofilms are microbial sessile structures that represent the default mode of growth of microorganisms in most environments. This study focused on the antimicrobial action of the EO extracted from one of the most representative oregano species, that is, Origanum vulgare (subsp. hirtum), against two important foodborne pathogens, Salmonella enterica (serovar Typhimurium) and Listeria monocytogenes. For this, the minimum inhibitory concentrations of the EO against the planktonic and biofilm growth of each bacterium were determined (MICs, MBICs), together with the minimum bactericidal and biofilm eradication concentrations (MBCs, MBECs). The EO was also analyzed for its chemical composition by gas chromatography-mass spectrometry analysis (GC-MS). The influence of EO exposure on the expression of some important virulence genes (hly, inlA, inlB and prfA) was also studied in L. monocytogenes. Results revealed a strong antibacterial and antibiofilm action with MICs and MBICs ranging from 0.03% to 0.06% (v/v) and from 0.06% to 0.13% (v/v), respectively. The application of the EO at 6.25% (v/v) for 15 min resulted in the total eradication of the biofilm cells of both pathogens. The EO was mainly composed of thymol, p-cymene, γ-terpinene and carvacrol. The 3 h exposure of L. monocytogenes planktonic cells to the EO at its MBIC (0.06% v/v) resulted in the significant downregulation of all the studied genes (p < 0.05). To sum, the results obtained advocate for the further exploitation of the antimicrobial action of oregano EO in food and health applications.
Collapse
Affiliation(s)
- Sonia Kolypetri
- Laboratory of Food Microbiology and Hygiene, Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Myrina, Lemnos, Greece
| | - Dimitra Kostoglou
- Laboratory of Food Microbiology and Hygiene, Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Myrina, Lemnos, Greece
| | - Anastasios Nikolaou
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Yiannis Kourkoutas
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Efstathios Giaouris
- Laboratory of Food Microbiology and Hygiene, Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Myrina, Lemnos, Greece
| |
Collapse
|
9
|
Wiktorczyk-Kapischke N, Skowron K, Wałecka-Zacharska E. Genomic and pathogenicity islands of Listeria monocytogenes-overview of selected aspects. Front Mol Biosci 2023; 10:1161486. [PMID: 37388250 PMCID: PMC10300472 DOI: 10.3389/fmolb.2023.1161486] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023] Open
Abstract
Listeria monocytogenes causes listeriosis, a disease characterized by a high mortality rate (up to 30%). Since the pathogen is highly tolerant to changing conditions (high and low temperature, wide pH range, low availability of nutrients), it is widespread in the environment, e.g., water, soil, or food. L. monocytogenes possess a number of genes that determine its high virulence potential, i.e., genes involved in the intracellular cycle (e.g., prfA, hly, plcA, plcB, inlA, inlB), response to stress conditions (e.g., sigB, gadA, caspD, clpB, lmo1138), biofilm formation (e.g., agr, luxS), or resistance to disinfectants (e.g., emrELm, bcrABC, mdrL). Some genes are organized into genomic and pathogenicity islands. The islands LIPI-1 and LIPI-3 contain genes related to the infectious life cycle and survival in the food processing environment, while LGI-1 and LGI-2 potentially ensure survival and durability in the production environment. Researchers constantly have been searching for new genes determining the virulence of L. monocytogenes. Understanding the virulence potential of L. monocytogenes is an important element of public health protection, as highly pathogenic strains may be associated with outbreaks and the severity of listeriosis. This review summarizes the selected aspects of L. monocytogenes genomic and pathogenicity islands, and the importance of whole genome sequencing for epidemiological purposes.
Collapse
Affiliation(s)
- Natalia Wiktorczyk-Kapischke
- Department of Microbiology, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Krzysztof Skowron
- Department of Microbiology, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Ewa Wałecka-Zacharska
- Department of Food Hygiene and Consumer Health, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| |
Collapse
|
10
|
Marked inter-strain heterogeneity in the differential expression of some key stress response and virulence-related genes between planktonic and biofilm cells in Listeria monocytogenes. Int J Food Microbiol 2023; 390:110136. [PMID: 36807004 DOI: 10.1016/j.ijfoodmicro.2023.110136] [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: 11/14/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
Listeria monocytogenes is a facultatively intracellular pathogenic bacterium that can provoke invasive listeriosis, a severe foodborne infection in humans. Outside the host, this is capable to survive for long periods in soil, and water, as well as on plants, while, like many other microorganisms, this can also attach to abiotic surfaces, such as food contact ones, forming biofilms on them. It has been suggested that inside those sessile communities, L. monocytogenes cells not only display an increased stress tolerance but may also boost their pathogenicity. In this work, the expression of ten key stress response and/or virulence-related genes (i.e., groEL, hly, iap, inlA, inlB, lisK, mdrD, mdrL, prfA, and sigB) was studied in three different L. monocytogenes strains (AAL20066, AAL20107, and PL24), all isolated from foods and each belonging to a different listeriosis-associated serovar (1/2a, 1/2b, and 1/2c, respectively). For this, each strain was initially left to develop a mature biofilm on a model polystyrene surface (Petri dish) by incubating for 144 h (6 days) at 20 °C in tryptone soya broth (with medium renewal every 48 h). Following incubation, both biofilm and the surrounding free-swimming (planktonic) cells were recovered, and their gene expressions were comparatively evaluated through targeted reverse transcription-quantitative polymerase chain reactions (RT-qPCR). Results revealed a strain-dependent differential gene expression between the two cell types. Thus, for instance, in strain AAL20107 (ser. 1/2b) biofilm growth worryingly resulted in a significant overexpression of all the studied genes (P < 0.05), whereas in strain PL24 (ser. 1/2c), the expression of most genes (8/10) did not change upon biofilm growth, with only two of them (groEL and hly) being again significantly upregulated. Such transcriptomic strain variability in stress adaptation and/or virulence induction should be generally considered in the physiological studies of pathogenic biofilms and preferably upon designing and implementing novel and more efficient eradication methods.
Collapse
|
11
|
Mora DSO, Cox M, Magunda F, Williams AB, Linke L. An optimized live bacterial delivery vehicle safely and efficaciously delivers bacterially transcribed therapeutic nucleic acids. Eng Life Sci 2023; 23:e2200037. [PMID: 36874611 PMCID: PMC9978928 DOI: 10.1002/elsc.202200037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/03/2022] [Accepted: 12/09/2022] [Indexed: 02/07/2023] Open
Abstract
There is an unmet need for delivery platforms that realize the full potential of next-generation nucleic acid therapeutics. The in vivo usefulness of current delivery systems is limited by numerous weaknesses, including poor targeting specificity, inefficient access to target cell cytoplasm, immune activation, off-target effects, small therapeutic windows, limited genetic encoding and cargo capacity, and manufacturing challenges. Here we characterize the safety and efficacy of a delivery platform comprising engineered live, tissue-targeting, non-pathogenic bacteria (Escherichia coli SVC1) for intracellular cargo delivery. SVC1 bacteria are engineered to specifically bind to epithelial cells via a surface-expressed targeting ligand, to allow escape of their cargo from the phagosome, and to have minimal immunogenicity. We describe SVC1's ability to deliver short hairpin RNA (shRNA), localized SVC1 administration to various tissues, and its minimal immunogenicity. To validate the therapeutic potential of SVC1, we used it to deliver influenza-targeting antiviral shRNAs to respiratory tissues in vivo. These data are the first to establish the safety and efficacy of this bacteria-based delivery platform for use in multiple tissue types and as an antiviral in the mammalian respiratory tract. We expect that this optimized delivery platform will enable a variety of advanced therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Forgivemore Magunda
- Department of Microbiology, Immunology and PathologyColorado State UniversityFort CollinsColoradoUSA
| | | | | |
Collapse
|
12
|
Liu M, Lv Q, Xu J, Liu B, Zhou Y, Zhang S, Shen X, Wang L. Isoflavone glucoside genistin, an inhibitor targeting Sortase A and Listeriolysin O, attenuates the virulence of Listeria monocytogenes in vivo and in vitro. Biochem Pharmacol 2023; 209:115447. [PMID: 36746262 DOI: 10.1016/j.bcp.2023.115447] [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: 08/24/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
As a common intracellular facultative anaerobic Gram-positive bacterium, Listeria monocytogenes (L. monocytogenes) exhibits strong resistance to extreme environments, such as low temperature and a wide range of pH values, causing contamination in food production and processing. Sortase A (SrtA) and listeriolysin O (LLO), two crucial virulence factors of L. monocytogenes, are widely recognized as potential targets for the development of anti-L. monocytogenes infection drugs. In this study, we found that genistin simultaneously inhibits the peptidase activity of SrtA and the hemolytic activity of LLO without affecting the growth of L. monocytogenes, alleviating concerns about developing resistance. Furthermore, we demonstrated that genistin reduces L. monocytogenes biofilm formation and invasion of human colorectal cancer (Caco-2) cells. Subsequent mechanistic studies revealed that genistin inhibited LLO-mediated Caco-2 cell damage by blocking LLO oligomerization. Fluorescence quenching assay revealed the potential binding mode of SrtA and LLO to genistin. Genistin might bind to the active pocket of SrtA through residues Leu33, Asn29, and Met40, interacting with D1 domain of LLO involved in oligomerization and pore formation through residues Asn259. Studies in infection models revealed that genistin reduces mortality and pathological damage in mice infected with L. monocytogenes. These results indicate that genistin is a promising anti-virulence agent that could be considered an alternative candidate for the treatment of L. monocytogenes infection.
Collapse
Affiliation(s)
- Minda Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, China
| | - Qianghua Lv
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, P.R.China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, P.R.China
| | - Jingwen Xu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Baichen Liu
- The Second Bethune Clinical Medical College of Jilin University, Changchun 130012, Jilin, People's Republic of China
| | - Yonglin Zhou
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, China
| | - Siqi Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, China
| | - Xue Shen
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Department of Food Science, College of Food Science and Engineering, Jilin University, Changchun, China.
| | - Lin Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
13
|
The Application of Cinnamon Twig Extract as an Inhibitor of Listeriolysin O against Listeria monocytogenes Infection. Molecules 2023; 28:molecules28041625. [PMID: 36838612 PMCID: PMC9962927 DOI: 10.3390/molecules28041625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 02/11/2023] Open
Abstract
As a major virulence factor of Listeria monocytogenes (L. monocytogenes), listeriolysin O (LLO) can assist in the immune escape of L. monocytogenes, which is critical for the pathogen to evade host immune recognition, leading to various infectious diseases. Cinnamon twig (CT), as a traditional medicine, has been widely used in clinics for multiple functions and it has exhibited excellent safety, efficacy and stability. There are few reports on the effects of the extracts of traditional medicine on bacterial virulence factors. CT has not been reported to be effective in the treatment of L. monocytogenes infection. Therefore, this study aims to explore the preventive effect of CT against L. monocytogenes infection in vivo and in vitro by targeting LLO. Firstly, a hemolysis assay and a cell viability determination are used to detect the effect of CT extract on the inhibition of the cytolytic activity of LLO. The potential mechanism through which CT extract inhibits LLO activity is predicted through network pharmacology, molecular docking assay, real-time polymerase chain reaction (RT-PCR), Western blotting and circular dichroism (CD) analysis. The experimental therapeutic effect of CT extract is examined in a mouse model infected with L. monocytogenes. Then, the ingredients are identified through a high-performance liquid chromatography (HPLC) and thin layer chromatography (TLC) analysis. Here we find that CT extract, containing mainly cinnamic acid, cinnamaldehyde, β-sitosterol, taxifolin, catechin and epicatechin, shows a potential inhibition of LLO-mediated hemolysis without any antimicrobial activity. The results of the mechanism research show that CT extract treatment can simultaneously inhibit LLO expression and oligomerization. Furthermore, the addition of CT extract led to a remarkable alleviation of LLO-induced cytotoxicity. After treatment with CT extract, the mortality, bacterial load, pathological damage and inflammatory responses of infected mice are significantly reduced when compared with the untreated group. This study suggests that CT extract can be a novel and multicomponent inhibitor of LLO with multiple strategies against L. monocytogenes infection, which could be further developed into a novel treatment for infections caused by L. monocytogenes.
Collapse
|
14
|
Johnstone BA, Joseph R, Christie MP, Morton CJ, McGuiness C, Walsh JC, Böcking T, Tweten RK, Parker MW. Cholesterol-dependent cytolysins: The outstanding questions. IUBMB Life 2022; 74:1169-1179. [PMID: 35836358 PMCID: PMC9712165 DOI: 10.1002/iub.2661] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/23/2022] [Indexed: 11/06/2022]
Abstract
The cholesterol-dependent cytolysins (CDCs) are a major family of bacterial pore-forming proteins secreted as virulence factors by Gram-positive bacterial species. CDCs are produced as soluble, monomeric proteins that bind specifically to cholesterol-rich membranes, where they oligomerize into ring-shaped pores of more than 30 monomers. Understanding the details of the steps the toxin undergoes in converting from monomer to a membrane-spanning pore is a continuing challenge. In this review we summarize what we know about CDCs and highlight the remaining outstanding questions that require answers to obtain a complete picture of how these toxins kill cells.
Collapse
Affiliation(s)
- Bronte A Johnstone
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Riya Joseph
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Michelle P Christie
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Craig J Morton
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Conall McGuiness
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - James C Walsh
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Till Böcking
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Rodney K Tweten
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Michael W Parker
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- Australian Cancer Research Foundation Rational Drug Discovery Centre, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| |
Collapse
|
15
|
Xu L, Zhou Y, Xu J, Xu X, Lu G, Lv Q, Wei L, Deng X, Shen X, Feng H, Wang J. Anti-inflammatory, antioxidant and anti-virulence roles of atractylodin in attenuating Listeria monocytogenes infection. Front Immunol 2022; 13:977051. [PMID: 36389842 PMCID: PMC9651212 DOI: 10.3389/fimmu.2022.977051] [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: 06/24/2022] [Accepted: 08/18/2022] [Indexed: 11/26/2022] Open
Abstract
Background Listeria monocytogenes (L. monocytogenes), as a pandemic foodborne pathogen, severely threatens food security and public health care worldwide, which evolves multiple bacterial virulence factors (such as listeriolysin O, LLO) for manipulating the immune response of L. monocytogenes-host interactions. Methods Hemolysis assay was employed to screen a potential LLO inhibitor and the underlying mechanisms were investigated using molecular dynamics (MD) simulation and oligomerization assay. The effects of candidates on immune response were examined by qRT-PCR and immunoblotting analysis. Histological analysis, ELISA assay and biochemistry detection were conducted to assess in vivo efficacy of candidates. Results In the present study, natural terpenoid atractylodin was characterized as an alternative drug candidate for the treatment of L. monocytogenes by the regulation of LLO function and host Nrf2/NLRP3 signaling pathway. Notably, in vivo infection model by L. monocytogenes also highlighted that atractylodin treatment provided effective therapeutic benefits, as evidenced by decreased bacterial burden and diminished inflammation. Congruently, the survival rate of L. monocytogenes-infection mice increased significantly from 10.0% to 40.0% by atractylodin treatment. Conclusion Collectively, our study showed for the first time that atractylodin has tremendous potential to attenuate L. monocytogenes pathogenicity by blocking LLO pore formation and mediating the suppression of inflammation and oxidative stress, providing a promising therapeutic strategy and broadening the applications of atractylodin against L. monocytogenes infection.
Collapse
Affiliation(s)
- Lei Xu
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yonglin Zhou
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jingwen Xu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiangzhu Xu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Gejin Lu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Qianghua Lv
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lijuan Wei
- Hebei Veterinary Medicine Technology Innovation Center, Shijiazhuang, China
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xue Shen
- Department of Food Science, College of Food Science and Engineering, Jilin University, Changchun, China
| | - Haihua Feng
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- *Correspondence: Jianfeng Wang, ; Haihua Feng,
| | - Jianfeng Wang
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- *Correspondence: Jianfeng Wang, ; Haihua Feng,
| |
Collapse
|
16
|
Pyz-Łukasik R, Paszkiewicz W, Kiełbus M, Ziomek M, Gondek M, Domaradzki P, Michalak K, Pietras-Ożga D. Genetic Diversity and Potential Virulence of Listeria monocytogenes Isolates Originating from Polish Artisanal Cheeses. Foods 2022; 11:2805. [PMID: 36140933 PMCID: PMC9497517 DOI: 10.3390/foods11182805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Artisanal cheeses can be sources of Listeria monocytogenes and cause disease in humans. This bacterial pathogen is a species of diverse genotypic and phenotypic characteristics. The aim of the study was to characterize 32 isolates of L. monocytogenes isolated in 2014-2018 from artisanal cheeses. The isolates were characterized using whole genome sequencing and bioinformatics analysis. The artisanal cheese isolates resolved to four molecular groups: 46.9% of them to IIa (1/2a-3a), 31.2% to IVb (4ab-4b-4d-4e), 12.5% to IIc (1/2c-3c), and 9.4% to IIb (1/2b-3b-7). Two evolutionary lineages emerged: lineage II having 59.4% of the isolates and lineage I having 40.6%. The sequence types (ST) totaled 18: ST6 (15.6% of the isolates), ST2, ST20, ST26, and ST199 (each 9.4%), ST7 and ST9 (each 6.3%), and ST1, ST3, ST8, ST16, ST87, ST91, ST121, ST122, ST195, ST217, and ST580 (each 3.1%). There were 15 detected clonal complexes (CC): CC6 (15.6% of isolates), CC9 (12.5%), CC2, CC20, CC26, and CC199 (each 9.4%), CC7 and CC8 (each 6.3%), and CC1, CC3, CC14, CC87, CC121, CC195, and CC217 (each 3.1%). The isolates were varied in their virulence genes and the differences concerned: inl, actA, LIPI-3, ami, gtcA, aut, vip, and lntA.
Collapse
Affiliation(s)
- Renata Pyz-Łukasik
- Department of Food Hygiene of Animal Origin, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-033 Lublin, Poland
| | - Waldemar Paszkiewicz
- Department of Food Hygiene of Animal Origin, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-033 Lublin, Poland
| | - Michał Kiełbus
- Department of Experimental Hematooncology, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland
| | - Monika Ziomek
- Department of Food Hygiene of Animal Origin, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-033 Lublin, Poland
| | - Michał Gondek
- Department of Food Hygiene of Animal Origin, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-033 Lublin, Poland
| | - Piotr Domaradzki
- Department of Quality Assessment and Processing of Animal Products, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland
| | - Katarzyna Michalak
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612 Lublin, Poland
| | - Dorota Pietras-Ożga
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612 Lublin, Poland
| |
Collapse
|
17
|
Kaempferol-Driven Inhibition of Listeriolysin O Pore Formation and Inflammation Suppresses Listeria monocytogenes Infection. Microbiol Spectr 2022; 10:e0181022. [PMID: 35856678 PMCID: PMC9431489 DOI: 10.1128/spectrum.01810-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Listeria monocytogenes remains a nonnegligible cause of foodborne infection, posing a critical threat to public health. Under the global antibiotic crisis, novel alternative approaches are urgently needed. The indispensable role of listeriolysin O (LLO) in the intracellular life cycle, barrier penetration, colonization, and systemic dissemination of L. monocytogenes renders it a potent drug target, which means curbing L. monocytogenes via interfering with LLO-associated pathogenic mechanisms. Here, we identified kaempferol, a natural small molecule compound, as an effective LLO inhibitor that engaged the residues Glu437, Ile468, and Tyr469 of LLO, thereby suppressing LLO-mediated membrane perforation and barrier disruption. Moreover, we found that kaempferol also suppressed host-derived inflammation in a distinct way independent of LLO inhibition. The in vivo study revealed that kaempferol treatment significantly reduced bacterial burden and cytokine burst in target organs, thereby effectively protecting mice from systemic L. monocytogenes infection. Our findings present kaempferol as a potential therapeutic application for L. monocytogenes infection, which is less likely to induce drug resistance than antibiotics because of its superiority of interfering with the pathogenesis process rather than exerting pressure on bacterial viability. IMPORTANCE Currently, we are facing a global crisis of antibiotic resistance, and novel alternative approaches are urgently needed to curb L. monocytogenes infection. Our study demonstrated that kaempferol alleviated L. monocytogenes infection via suppressing LLO pore formation and inflammation response, which might represent a novel antimicrobial-independent strategy to curb listeriosis.
Collapse
|
18
|
Ju S, Chen H, Wang S, Lin J, Ma Y, Aroian RV, Peng D, Sun M. C. elegans monitor energy status via the AMPK pathway to trigger innate immune responses against bacterial pathogens. Commun Biol 2022; 5:643. [PMID: 35773333 PMCID: PMC9246835 DOI: 10.1038/s42003-022-03589-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Pathogen recognition and the triggering of host innate immune system are critical to understanding pathogen-host interaction. Cellular surveillance systems have been identified as an important strategy for the identification of microbial infection. In the present study, using Bacillus thuringiensis-Caenorhabditis elegans as a model, we found an approach for surveillance systems to sense pathogens. We report that Bacillus thuringiensis Cry5Ba, a typical pore-forming toxin, caused mitochondrial damage and energy imbalance by triggering potassium ion leakage, instead of directly targeting mitochondria. Interestingly, we find C. elegans can monitor intracellular energy status to trigger innate immune responses via AMP-activated protein kinase (AMPK), secreting multiple effectors to defend against pathogenic attacks. Our study indicates that the imbalance of energy status is a prevalent side effect of pathogen infection. Furthermore, the AMPK-dependent surveillance system may serve as a practicable strategy for the host to recognize and defense against pathogens. Bacillus thuringiensis toxin Cry5Ba triggers potassium ion leakage, causing mitochondrial damage and energy imbalance. C. elegans can monitor this intracellular energy imbalance via AMP-activated protein kinase to trigger innate immune responses.
Collapse
Affiliation(s)
- Shouyong Ju
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hanqiao Chen
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shaoying Wang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jian Lin
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yanli Ma
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Raffi V Aroian
- Program in Molecular Medicine, University of Massachusetts Chan Medical School Worcester, Worcester, MA, 01605-2377, USA
| | - Donghai Peng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Ming Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
19
|
Rosenberg G, Riquelme S, Prince A, Avraham R. Immunometabolic crosstalk during bacterial infection. Nat Microbiol 2022; 7:497-507. [PMID: 35365784 DOI: 10.1038/s41564-022-01080-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 02/03/2022] [Indexed: 01/22/2023]
Abstract
Following detection of bacteria, macrophages switch their metabolism from oxidative respiration through the tricarboxylic acid cycle to high-rate aerobic glycolysis. This immunometabolic shift enables pro-inflammatory and antimicrobial responses and is facilitated by the accumulation of fatty acids, tricarboxylic acid-derived metabolites and catabolism of amino acids. Recent studies have shown that these immunometabolites are co-opted by pathogens as environmental cues for expression of virulence genes. We review mechanisms by which host immunometabolites regulate bacterial pathogenicity and discuss opportunities for the development of therapeutics targeting metabolic host-pathogen crosstalk.
Collapse
Affiliation(s)
- Gili Rosenberg
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | - Alice Prince
- Columbia University Medical Center, New York, NY, USA.
| | - Roi Avraham
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
20
|
Larsen SE, Williams BD, Rais M, Coler RN, Baldwin SL. It Takes a Village: The Multifaceted Immune Response to Mycobacterium tuberculosis Infection and Vaccine-Induced Immunity. Front Immunol 2022; 13:840225. [PMID: 35359957 PMCID: PMC8960931 DOI: 10.3389/fimmu.2022.840225] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Despite co-evolving with humans for centuries and being intensely studied for decades, the immune correlates of protection against Mycobacterium tuberculosis (Mtb) have yet to be fully defined. This lapse in understanding is a major lag in the pipeline for evaluating and advancing efficacious vaccine candidates. While CD4+ T helper 1 (TH1) pro-inflammatory responses have a significant role in controlling Mtb infection, the historically narrow focus on this cell population may have eclipsed the characterization of other requisite arms of the immune system. Over the last decade, the tuberculosis (TB) research community has intentionally and intensely increased the breadth of investigation of other immune players. Here, we review mechanistic preclinical studies as well as clinical anecdotes that suggest the degree to which different cell types, such as NK cells, CD8+ T cells, γ δ T cells, and B cells, influence infection or disease prevention. Additionally, we categorically outline the observed role each major cell type plays in vaccine-induced immunity, including Mycobacterium bovis bacillus Calmette-Guérin (BCG). Novel vaccine candidates advancing through either the preclinical or clinical pipeline leverage different platforms (e.g., protein + adjuvant, vector-based, nucleic acid-based) to purposefully elicit complex immune responses, and we review those design rationales and results to date. The better we as a community understand the essential composition, magnitude, timing, and trafficking of immune responses against Mtb, the closer we are to reducing the severe disease burden and toll on human health inflicted by TB globally.
Collapse
Affiliation(s)
- Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Brittany D. Williams
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,*Correspondence: Susan L. Baldwin,
| |
Collapse
|
21
|
Ulhuq FR, Mariano G. Bacterial pore-forming toxins. MICROBIOLOGY (READING, ENGLAND) 2022; 168:001154. [PMID: 35333704 PMCID: PMC9558359 DOI: 10.1099/mic.0.001154] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/03/2022] [Indexed: 12/11/2022]
Abstract
Pore-forming toxins (PFTs) are widely distributed in both Gram-negative and Gram-positive bacteria. PFTs can act as virulence factors that bacteria utilise in dissemination and host colonisation or, alternatively, they can be employed to compete with rival microbes in polymicrobial niches. PFTs transition from a soluble form to become membrane-embedded by undergoing large conformational changes. Once inserted, they perforate the membrane, causing uncontrolled efflux of ions and/or nutrients and dissipating the protonmotive force (PMF). In some instances, target cells intoxicated by PFTs display additional effects as part of the cellular response to pore formation. Significant progress has been made in the mechanistic description of pore formation for the different PFTs families, but in several cases a complete understanding of pore structure remains lacking. PFTs have evolved recognition mechanisms to bind specific receptors that define their host tropism, although this can be remarkably diverse even within the same family. Here we summarise the salient features of PFTs and highlight where additional research is necessary to fully understand the mechanism of pore formation by members of this diverse group of protein toxins.
Collapse
Affiliation(s)
- Fatima R. Ulhuq
- Microbes in Health and Disease Theme, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Giuseppina Mariano
- Microbes in Health and Disease Theme, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
22
|
Grijmans BJM, van der Kooij SB, Varela M, Meijer AH. LAPped in Proof: LC3-Associated Phagocytosis and the Arms Race Against Bacterial Pathogens. Front Cell Infect Microbiol 2022; 11:809121. [PMID: 35047422 PMCID: PMC8762105 DOI: 10.3389/fcimb.2021.809121] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/10/2021] [Indexed: 01/05/2023] Open
Abstract
Cells of the innate immune system continuously patrol the extracellular environment for potential microbial threats that are to be neutralized by phagocytosis and delivery to lysosomes. In addition, phagocytes employ autophagy as an innate immune mechanism against pathogens that succeed to escape the phagolysosomal pathway and invade the cytosol. In recent years, LC3-associated phagocytosis (LAP) has emerged as an intermediate between phagocytosis and autophagy. During LAP, phagocytes target extracellular microbes while using parts of the autophagic machinery to label the cargo-containing phagosomes for lysosomal degradation. LAP contributes greatly to host immunity against a multitude of bacterial pathogens. In the pursuit of survival, bacteria have developed elaborate strategies to disarm or circumvent the LAP process. In this review, we will outline the nature of the LAP mechanism and discuss recent insights into its interplay with bacterial pathogens.
Collapse
Affiliation(s)
| | | | - Monica Varela
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | | |
Collapse
|
23
|
Bagatella S, Tavares-Gomes L, Oevermann A. Listeria monocytogenes at the interface between ruminants and humans: A comparative pathology and pathogenesis review. Vet Pathol 2021; 59:186-210. [PMID: 34856818 DOI: 10.1177/03009858211052659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The bacterium Listeria monocytogenes (Lm) is widely distributed in the environment as a saprophyte, but may turn into a lethal intracellular pathogen upon ingestion. Invasive infections occur in numerous species worldwide, but most commonly in humans and farmed ruminants, and manifest as distinct forms. Of those, neuroinfection is remarkably threatening due to its high mortality. Lm is widely studied not only as a pathogen but also as an essential model for intracellular infections and host-pathogen interactions. Many aspects of its ecology and pathogenesis, however, remain unclear and are rarely addressed in its natural hosts. This review highlights the heterogeneity and adaptability of Lm by summarizing its association with the environment, farm animals, and disease. It also provides current knowledge on key features of the pathology and (molecular) pathogenesis of various listeriosis forms in naturally susceptible species with a special focus on ruminants and on the neuroinvasive form of the disease. Moreover, knowledge gaps on pathomechanisms of listerial infections and relevant unexplored topics in Lm pathogenesis research are highlighted.
Collapse
Affiliation(s)
- Stefano Bagatella
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Leticia Tavares-Gomes
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Anna Oevermann
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
24
|
Kokkoni EA, Andritsos N, Sakarikou C, Michailidou S, Argiriou A, Giaouris E. Investigating Transcriptomic Induction of Resistance and/or Virulence in Listeria monocytogenes Cells Surviving Sublethal Antimicrobial Exposure. Foods 2021; 10:foods10102382. [PMID: 34681431 PMCID: PMC8535302 DOI: 10.3390/foods10102382] [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: 08/29/2021] [Revised: 09/26/2021] [Accepted: 10/05/2021] [Indexed: 01/06/2023] Open
Abstract
The potential transcriptomic induction of resistance and/or virulence in two L. monocytogenes strains belonging to the most frequent listeriosis-associated serovars (i.e., 1/2a and 4b), following their sublethal antimicrobial exposure, was studied through qPCR determination of the relative expression of 10 selected related genes (i.e., groEL, hly, iap, inlA, inlB, lisK, mdrD, mdrL, prfA, and sigB). To induce sublethal stress, three common antimicrobials (i.e., benzalkonium chloride, thymol, and ampicillin) were individually applied for 2 h at 37 °C against stationary phase cells of each strain, each at a sublethal concentration. In general, the expression of most of the studied genes remained either stable or was significantly downregulated following the antimicrobial exposure, with some strain-specific differences to be yet recorded. Thymol provoked downregulation of most of the studied genes, significantly limiting the expression of 6/10 and 4/10 genes in the strains of ser. 1/2a and ser. 4b, respectively, including those coding for the master regulators of stress response and virulence (SigB and PrfA, respectively), in both strains. At the same time, the two genes coding for the invasion internalin proteins (InlA and InlB), with crucial role in the onset of L. monocytogenes pathogenesis, were both importantly upregulated in ser. 4b strain. The results obtained increase our knowledge of the stress physiology of L. monocytogenes under certain sublethal antimicrobial conditions that could be encountered within the food chain and in clinical settings, and may assist in better and more effective mitigation strategies.
Collapse
Affiliation(s)
- Eleni-Anna Kokkoni
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, Ierou Lochou 10 & Makrygianni, 81400 Myrina, Greece; (E.-A.K.); (N.A.); (C.S.); (S.M.); (A.A.)
| | - Nikolaos Andritsos
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, Ierou Lochou 10 & Makrygianni, 81400 Myrina, Greece; (E.-A.K.); (N.A.); (C.S.); (S.M.); (A.A.)
- Athens Analysis Laboratories S.A., Microbiology Laboratory, Nafpliou 29, 14452 Metamorfosi, Greece
| | - Christina Sakarikou
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, Ierou Lochou 10 & Makrygianni, 81400 Myrina, Greece; (E.-A.K.); (N.A.); (C.S.); (S.M.); (A.A.)
| | - Sofia Michailidou
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, Ierou Lochou 10 & Makrygianni, 81400 Myrina, Greece; (E.-A.K.); (N.A.); (C.S.); (S.M.); (A.A.)
- Centre for Research and Technology Hellas (CERTH), Institute of Applied Biosciences, 57001 Thessaloniki, Greece
| | - Anagnostis Argiriou
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, Ierou Lochou 10 & Makrygianni, 81400 Myrina, Greece; (E.-A.K.); (N.A.); (C.S.); (S.M.); (A.A.)
- Centre for Research and Technology Hellas (CERTH), Institute of Applied Biosciences, 57001 Thessaloniki, Greece
| | - Efstathios Giaouris
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, Ierou Lochou 10 & Makrygianni, 81400 Myrina, Greece; (E.-A.K.); (N.A.); (C.S.); (S.M.); (A.A.)
- Correspondence: ; Tel.: +30-22540-83115
| |
Collapse
|
25
|
Bai X, Nakatsu CH, Bhunia AK. Bacterial Biofilms and Their Implications in Pathogenesis and Food Safety. Foods 2021; 10:2117. [PMID: 34574227 PMCID: PMC8472614 DOI: 10.3390/foods10092117] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/21/2021] [Accepted: 08/30/2021] [Indexed: 12/28/2022] Open
Abstract
Biofilm formation is an integral part of the microbial life cycle in nature. In food processing environments, bacterial transmissions occur primarily through raw or undercooked foods and by cross-contamination during unsanitary food preparation practices. Foodborne pathogens form biofilms as a survival strategy in various unfavorable environments, which also become a frequent source of recurrent contamination and outbreaks of foodborne illness. Instead of focusing on bacterial biofilm formation and their pathogenicity individually, this review discusses on a molecular level how these two physiological processes are connected in several common foodborne pathogens such as Listeria monocytogenes, Staphylococcus aureus, Salmonella enterica and Escherichia coli. In addition, biofilm formation by Pseudomonas aeruginosa is discussed because it aids the persistence of many foodborne pathogens forming polymicrobial biofilms on food contact surfaces, thus significantly elevating food safety and public health concerns. Furthermore, in-depth analyses of several bacterial molecules with dual functions in biofilm formation and pathogenicity are highlighted.
Collapse
Affiliation(s)
- Xingjian Bai
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA;
| | - Cindy H. Nakatsu
- Department of Agronomy, Purdue University, West Lafayette, IN 47907, USA;
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| | - Arun K. Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA;
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
26
|
Gluschko A, Farid A, Herb M, Grumme D, Krönke M, Schramm M. Macrophages target Listeria monocytogenes by two discrete non-canonical autophagy pathways. Autophagy 2021; 18:1090-1107. [PMID: 34482812 DOI: 10.1080/15548627.2021.1969765] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Non-canonical autophagy pathways decorate single-membrane vesicles with Atg8-family proteins such as MAP1LC3/LC3 (microtubule-associated protein 1 light chain 3). Phagosomes containing the bacterial pathogen Listeria monocytogenes (L.m.) can be targeted by a non-canonical autophagy pathway called LC3-associated phagocytosis (LAP), which substantially contributes to the anti-listerial activity of macrophages and immunity. We here characterized a second non-canonical autophagy pathway targeting L.m.-containing phagosomes, which is induced by damage caused to the phagosomal membrane by the pore-forming toxin of L.m., listeriolysin O. This pore-forming toxin-induced non-canonical autophagy pathway (PINCA) was the only autophagic pathway evoked in tissue macrophages deficient for the NADPH oxidase CYBB/NOX2 that produces the reactive oxygen species (ROS) that are required for LAP induction. Similarly, also bone marrow-derived macrophages (BMDM) exclusively targeted L.m. by PINCA as they completely failed to induce LAP because of insufficient production of ROS through CYBB, in part, due to low expression of some CYBB complex subunits. Priming of BMDM with proinflammatory cytokines such as TNF and IFNG/IFNγ increased ROS production by CYBB and endowed them with the ability to target L.m. by LAP. Targeting of L.m. by LAP remained relatively rare, though, preventing LAP from substantially contributing to the anti-listerial activity of BMDM. Similar to LAP, the targeting of L.m.-containing phagosomes by PINCA promoted their fusion with lysosomes. Surprisingly, however, this did not substantially contribute to anti-listerial activity of BMDM. Thus, in contrast to LAP, PINCA does not have clear anti-listerial function suggesting that the two different non-canonical autophagy pathways targeting L.m. may have discrete functions.Abbreviations: actA/ActA: actin assembly-inducing protein A; ATG: autophagy-related; BMDM: Bone marrow-derived macrophages; CALCOCO2/NDP52: calcium-binding and coiled-coil domain-containing protein 2; CYBA/p22phox: cytochrome b-245 light chain; CYBB/NOX2: cytochrome b(558) subunit beta; E. coli: Escherichia coli; IFNG/IFNγ: interferon gamma; L.m.: Listeria monocytogenes; LAP: LC3-associated phagocytosis; LGALS: galectin; LLO: listeriolysin O; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; NCF1/p47phox: neutrophil cytosol factor 1; NCF2/p67phox: neutrophil cytosol factor 2; NCF4/p67phox: neutrophil cytosol factor 4; Peritoneal macrophages: PM; PINCA: pore-forming toxin-induced non-canonical autophagy; plc/PLC: 1-phosphatidylinositol phosphodiesterase; PMA: phorbol 12-myristate 13-acetate; RB1CC1/FIP200: RB1-inducible coiled-coil protein 1; ROS: reactive oxygen species; S. aureus: Staphylococcus aureus; S. flexneri: Shigella flexneri; SQSTM1/p62: sequestosome 1; S. typhimurium: Salmonella typhimurium; T3SS: type III secretion system; TNF: tumor necrosis factor; ULK: unc-51 like autophagy activating kinase; PM: peritoneal macrophages; WT: wild type.
Collapse
Affiliation(s)
- Alexander Gluschko
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Alina Farid
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Daniela Grumme
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Martin Krönke
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany.,Center of Molecular Medicine Cologne, Cologne, Germany.,Cologne Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases Cecad, Cologne, Germany.,German Center for Infection Research Dzif, Cologne, Germany
| | - Michael Schramm
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| |
Collapse
|
27
|
Ilangumaran Ponmalar I, Ayappa KG, Basu JK. Bacterial protein listeriolysin O induces nonmonotonic dynamics because of lipid ejection and crowding. Biophys J 2021; 120:3040-3049. [PMID: 34214525 DOI: 10.1016/j.bpj.2021.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 01/15/2023] Open
Abstract
Membrane-bound protein complexes involving pore forming toxins (PFTs) released by virulent bacteria are known to form transmembrane pores leading to host cell lysis. Developing alternative strategies against PFT mediated bacterial virulence factors requires an understanding of the cellular membrane response. However, membrane disruption and related lipid reorganization events during attack by PFTs remain largely unexplored. We report counterintuitive and nonmonotonic variations in lipid diffusion, measured using confocal fluorescence correlation spectroscopy, due to interplay of lipid ejection and crowding by membrane-bound oligomers of a prototypical cholesterol-dependent cytolysin, listeriolysin O (LLO). The observed dynamical crossover is correlated with concentration dependent transitions of LLO oligomeric state populations from rings to arc-like pore complexes, predicted using a proposed two-state free area-based diffusion model. At low PFT concentrations, a hitherto unexplored regime of increased lipid diffusivity is attributed to lipid ejection events because of a preponderance of ring-like pore states. At higher protein concentrations in which membrane-inserted arc-like pores dominate, lipid ejection is less efficient and the ensuing crowding results in a lowering of lipid diffusion. These variations in lipid dynamics are corroborated by macroscopic rheological response measurements of PFT bound vesicles. Our study correlates PFT oligomeric state transitions, membrane remodeling, and mechanical property variations, providing unique insights into the pore forming mechanisms of cholesterol-dependent cytolysins.
Collapse
Affiliation(s)
| | - K Ganapathy Ayappa
- Center for BioSystems Science and Engineering Bengaluru, India; Department of Chemical Engineering Bengaluru, India.
| | - Jaydeep K Basu
- Department of Physics, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
28
|
Impens F, Dussurget O. Three decades of listeriology through the prism of technological advances. Cell Microbiol 2021; 22:e13183. [PMID: 32185895 DOI: 10.1111/cmi.13183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022]
Abstract
Decades of breakthroughs resulting from cross feeding of microbiological research and technological innovation have promoted Listeria monocytogenes to the rank of model microorganism to study host-pathogen interactions. The extraordinary capacity of this bacterium to interfere with a vast array of host cellular processes uncovered new concepts in microbiology, cell biology and infection biology. Here, we review technological advances that revealed how bacteria and host interact in space and time at the molecular, cellular, tissue and whole body scales, ultimately revolutionising our understanding of Listeria pathogenesis. With the current bloom of multidisciplinary integrative approaches, Listeria entered a new microbiology era.
Collapse
Affiliation(s)
- Francis Impens
- Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department for Biomedical Medicine, Ghent University, Ghent, Belgium.,VIB Proteomics Core, VIB, Ghent, Belgium
| | - Olivier Dussurget
- Institut Pasteur, Unité de Recherche Yersinia, Paris, France.,Université de Paris, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
29
|
Bierne H, Hamon M. Targeting host epigenetic machinery: The Listeria paradigm. Cell Microbiol 2021; 22:e13169. [PMID: 32185898 DOI: 10.1111/cmi.13169] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 12/11/2022]
Abstract
By modifying the host cell transcription programme, pathogenic bacteria disrupt a wide range of cellular processes and take control of the host's immune system. Conversely, by mobilising a network of defence genes, the host cells trigger various responses that allow them to tolerate or eliminate invaders. The study of the molecular basis of this crosstalk is crucial to the understanding of infectious diseases. Although research has long focused on the targeting of eukaryotic DNA-binding transcription factors, more recently, another powerful way by which bacteria modify the expression of host genes has emerged: chromatin modifications in the cell nucleus. One of the most prolific bacterial models in this area has been Listeria monocytogenes, a facultative intracellular bacterium responsible for serious food-borne infections. Here, we aim to highlight the contribution of this model to the field of bacteria-mediated chromatin modifications. We will first recall the general principles of epigenetic regulation and then illustrate five mechanisms that mobilise the epigenetic machinery in response to Listeria factors, either through bacterial molecular patterns, a toxin, an invasion protein, or nucleomodulins. Strategies used by Listeria to control the expression of host genes at the chromatin level, by activation of cytosolic signalling pathways or direct targeting of epifactors in the nucleus, have contributed to the emergence of a new discipline combining cellular microbiology and epigenetics: "patho-epigenetics."
Collapse
Affiliation(s)
- Hélène Bierne
- Université Paris Saclay, INRAE, AgroParisTech, Micalis Institute, Epigenetics and Cellular Microbiology Team, Jouy-en-Josas, France
| | - Mélanie Hamon
- G5 Chromatin and Infection, Institut Pasteur, Paris, France
| |
Collapse
|
30
|
Li T, Kong L, Li X, Wu S, Attri KS, Li Y, Gong W, Zhao B, Li L, Herring LE, Asara JM, Xu L, Luo X, Lei YL, Ma Q, Seveau S, Gunn JS, Cheng X, Singh PK, Green DR, Wang H, Wen H. Listeria monocytogenes upregulates mitochondrial calcium signalling to inhibit LC3-associated phagocytosis as a survival strategy. Nat Microbiol 2021; 6:366-379. [PMID: 33462436 PMCID: PMC8323152 DOI: 10.1038/s41564-020-00843-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/27/2020] [Indexed: 01/29/2023]
Abstract
Mitochondria are believed to have originated ~2.5 billion years ago. As well as energy generation in cells, mitochondria have a role in defence against bacterial pathogens. Despite profound changes in mitochondrial morphology and functions following bacterial challenge, whether intracellular bacteria can hijack mitochondria to promote their survival remains elusive. We report that Listeria monocytogenes-an intracellular bacterial pathogen-suppresses LC3-associated phagocytosis (LAP) by modulation of mitochondrial Ca2+ (mtCa2+) signalling in order to survive inside cells. Invasion of macrophages by L. monocytogenes induced mtCa2+ uptake through the mtCa2+ uniporter (MCU), which in turn increased acetyl-coenzyme A (acetyl-CoA) production by pyruvate dehydrogenase. Acetylation of the LAP effector Rubicon with acetyl-CoA decreased LAP formation. Genetic ablation of MCU attenuated intracellular bacterial growth due to increased LAP formation. Our data show that modulation of mtCa2+ signalling can increase bacterial survival inside cells, and highlight the importance of mitochondrial metabolism in host-microbial interactions.
Collapse
Affiliation(s)
- Tianliang Li
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Ligang Kong
- Shandong Institute of Otolaryngology, Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Xinghui Li
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Sijin Wu
- College of Pharmacy, Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, OH, USA
| | - Kuldeep S Attri
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yan Li
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Weipeng Gong
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Bao Zhao
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Lupeng Li
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura E Herring
- Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Lei Xu
- Shandong Institute of Otolaryngology, Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Xiaobo Luo
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Yu L Lei
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Stephanie Seveau
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - John S Gunn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Xiaolin Cheng
- College of Pharmacy, Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, OH, USA
| | - Pankaj K Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Haibo Wang
- Shandong Institute of Otolaryngology, Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China.
| | - Haitao Wen
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
31
|
Pickering RJ, Booty LM. NLR in eXile: Emerging roles of NLRX1 in immunity and human disease. Immunology 2020; 162:268-280. [PMID: 33314068 DOI: 10.1111/imm.13291] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
NLRX1 is a member of the NOD-like receptor family, a set of pattern recognition receptors associated with innate immunity. Interestingly, NLRX1 exists in somewhat of an exile from its NLR counterparts with unique features that mediate atypical functions compared with traditional NOD-like receptors (NLRs). Aside from a mitochondrial targeting sequence, the N-terminal region is yet to be characterized. Mitochondrially located, NLRX1 sits within a subgroup of regulatory NLRs responsible for negatively regulating cellular inflammatory signalling. As well as modulating pathogen response, emerging evidence is implicating NLRX1 as a central homeostatic gatekeeper between mitochondrial biology and immunological response. More recently, NLRX1 has been implicated in a wide range of disease, both pathogen-driven and otherwise. Emerging links of NLRX1 in cancer biology, autoimmunity and other inflammatory conditions are raising the potential of targeting NLRX1 therapeutically, with recent studies in inflammatory bowel disease showing great promise. Within this review, we address the unique features of NLRX1, its roles in innate immune signalling and its involvement in a range of inflammatory, metabolic and oncology disease indications with a focus on areas that could benefit from therapeutic targeting of NLRX1.
Collapse
Affiliation(s)
- Robert J Pickering
- Immunology Network, Adaptive Immunity Research Unit, GlaxoSmithKline, Stevenage, UK.,Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, UK
| | - Lee M Booty
- Immunology Network, Adaptive Immunity Research Unit, GlaxoSmithKline, Stevenage, UK
| |
Collapse
|
32
|
Multifunctional Amyloids in the Biology of Gram-Positive Bacteria. Microorganisms 2020; 8:microorganisms8122020. [PMID: 33348645 PMCID: PMC7766987 DOI: 10.3390/microorganisms8122020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 01/18/2023] Open
Abstract
Since they were discovered, amyloids have proven to be versatile proteins able to participate in a variety of cellular functions across all kingdoms of life. This multitask trait seems to reside in their ability to coexist as monomers, aggregates or fibrillar entities, with morphological and biochemical peculiarities. It is precisely this common molecular behaviour that allows amyloids to cross react with one another, triggering heterologous aggregation. In bacteria, many of these functional amyloids are devoted to the assembly of biofilms by organizing the matrix scaffold that keeps cells together. However, consistent with their notion of multifunctional proteins, functional amyloids participate in other biological roles within the same organisms, and emerging unprecedented functions are being discovered. In this review, we focus on functional amyloids reported in gram-positive bacteria, which are diverse in their assembly mechanisms and remarkably specific in their biological functions that they perform. Finally, we consider cross-seeding between functional amyloids as an emerging theme in interspecies interactions that contributes to the diversification of bacterial biology.
Collapse
|
33
|
Anast JM, Bobik TA, Schmitz-Esser S. The Cobalamin-Dependent Gene Cluster of Listeria monocytogenes: Implications for Virulence, Stress Response, and Food Safety. Front Microbiol 2020; 11:601816. [PMID: 33240255 PMCID: PMC7677406 DOI: 10.3389/fmicb.2020.601816] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
Several genes of the eut, pdu, and cob/cbi operons are responsible for the metabolism of ethanolamine (EA) and 1,2-propanediol (PD) and are essential during the pathogenic lifecycles of various enteric pathogens. Studies concerning EA and PD metabolism have primarily focused on bacterial genera from the family Enterobacteriaceae, especially the genus Salmonella. Listeria monocytogenes is a member of the Firmicutes phylum and is the causative agent of the rare but highly fatal foodborne disease listeriosis. The eut, pdu, and cob/cbi operons are organized as a single large locus collectively referred to as the cobalamin-dependent gene cluster (CDGC). The CDGC is well conserved in L. monocytogenes; however, functional characterization of the genes in this cluster and how they may contribute to Listeria virulence and stress tolerance in food production environments is highly limited. Previous work suggests that the degradation pathway of PD is essential for L. monocytogenes establishment in the gastrointestinal tract. In contrast, EA metabolism may be more important during intracellular replication. Other studies indicate that the CDGC is utilized when L. monocytogenes is exposed to food and food production relevant stress conditions. Perhaps most noteworthy, L. monocytogenes exhibits attenuated growth at cold temperatures when a key EA utilization pathway gene was deleted. This review aims to summarize the current knowledge of these pathways in L. monocytogenes and their significance in virulence and stress tolerance, especially considering recent developments.
Collapse
Affiliation(s)
- Justin M Anast
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States.,Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Thomas A Bobik
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States.,Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, United States
| | - Stephan Schmitz-Esser
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States.,Department of Animal Science, Iowa State University, Ames, IA, United States
| |
Collapse
|
34
|
Cytotoxic property of Streptococcus mitis strain producing two different types of cholesterol-dependent cytolysins. INFECTION GENETICS AND EVOLUTION 2020; 85:104483. [PMID: 32731044 DOI: 10.1016/j.meegid.2020.104483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 01/08/2023]
Abstract
Streptococcus mitis strain Nm-65 secretes an atypical 5-domain-type cholesterol-dependent cytolysin (CDC) called S. mitis-derived human platelet aggregation factor (Sm-hPAF) originally described as a platelet aggregation factor. Sm-hPAF belongs to Group III CDC that recognize both membrane cholesterol and human CD59 as the receptors, and shows preferential activity towards human cells. Draft genome analyses have shown that the Nm-65 strain also harbors a gene encoding another CDC called mitilysin (MLY). This CDC belongs to Group I CDC that recognizes only membrane cholesterol as a receptor, and it is a homolog of the pneumococcal CDC, pneumolysin. The genes encoding each CDC are located about 20 kb apart on the Nm-65 genome. Analysis of the genomic locus of these CDC-encoding genes in silico showed that the gene encoding Sm-hPAF and the region including the gene encoding MLY were both inserted into a specific locus of the S. mitis genome. The results obtained using deletion mutants of the gene(s) encoding CDC in Nm-65 indicated that each CDC contributes to both hemolysis and cytotoxicity, and that MLY is the major hemolysin/cytolysin in Nm-65. The present study aimed to determine the potential pathogenicity of an S. mitis strain that produces two CDC with different receptor recognition properties and secretion modes.
Collapse
|
35
|
Phelps CC, Vadia S, Boyaka PN, Varikuti S, Attia Z, Dubey P, Satoskar AR, Tweten R, Seveau S. A listeriolysin O subunit vaccine is protective against Listeria monocytogenes. Vaccine 2020; 38:5803-5813. [PMID: 32684498 DOI: 10.1016/j.vaccine.2020.06.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 05/12/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022]
Abstract
Listeria monocytogenes is a facultative intracellular pathogen responsible for the life-threatening disease listeriosis. The pore-forming toxin listeriolysin O (LLO) is a critical virulence factor that plays a major role in the L. monocytogenes intracellular lifecycle and is indispensable for pathogenesis. LLO is also a dominant antigen for T cells involved in sterilizing immunity and it was proposed that LLO acts as a T cell adjuvant. In this work, we generated a novel full-length LLO toxoid (LLOT) in which the cholesterol-recognition motif, a threonine-leucine pair located at the tip of the LLO C-terminal domain, was substituted with two glycine residues. We showed that LLOT lost its ability to bind cholesterol and to form pores. Importantly, LLOT retained binding to the surface of epithelial cells and macrophages, suggesting that it could efficiently be captured by antigen-presenting cells. We then determined if LLOT can be used as an antigen and adjuvant to protect mice from L. monocytogenes infection. Mice were immunized with LLOT alone or together with cholera toxin or Alum as adjuvants. We found that mice immunized with LLOT alone or in combination with the Th2-inducing adjuvant Alum were not protected against L. monocytogenes. On the other hand, mice immunized with LLOT along with the experimental adjuvant cholera toxin, were protected against L. monocytogenes, as evidenced by a significant decrease in bacterial burden in the liver and spleen three days post-infection. This immunization regimen elicited mixed Th1, Th2, and Th17 responses, as well as the generation of LLO-neutralizing antibodies. Further, we identified T cells as being required for immunization-induced reductions in bacterial burden, whereas B cells were dispensable in our model of non-pregnant young mice. Overall, this work establishes that LLOT is a promising vaccine antigen for the induction of protective immunity against L. monocytogenes by subunit vaccines containing Th1-driving adjuvants.
Collapse
Affiliation(s)
- Christopher C Phelps
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA; Department of Microbiology, The Ohio State University, The Ohio State University, Columbus, OH, USA
| | - Stephen Vadia
- Department of Microbiology, The Ohio State University, The Ohio State University, Columbus, OH, USA; Department of Biology, Washington University in St. Louis, MO 63130, USA
| | - Prosper N Boyaka
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA; Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Sanjay Varikuti
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Zayed Attia
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Rodney Tweten
- Department of Microbial & Immunology, University of Oklahoma, Oklahoma City, OK, USA
| | - Stephanie Seveau
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA; Department of Microbiology, The Ohio State University, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
36
|
Matereke LT, Okoh AI. Listeria monocytogenes Virulence, Antimicrobial Resistance and Environmental Persistence: A Review. Pathogens 2020; 9:E528. [PMID: 32629911 PMCID: PMC7400505 DOI: 10.3390/pathogens9070528] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/06/2020] [Accepted: 06/20/2020] [Indexed: 12/23/2022] Open
Abstract
Listeria monocytogenes is a ubiquitous opportunistic pathogen responsible for the well-known listeriosis disease. This bacterium has become a common contaminant of food, threatening the food processing industry. Once consumed, the pathogen is capable of traversing epithelial barriers, cellular invasion, and intracellular replication through the modulation of virulence factors such as internalins and haemolysins. Mobile genetic elements (plasmids and transposons) and other sophisticated mechanisms are thought to contribute to the increasing antimicrobial resistance of L. monocytogenes. The environmental persistence of the pathogen is aided by its ability to withstand environmental stresses such as acidity, cold stress, osmotic stress, and oxidative stress. This review seeks to give an insight into L. monocytogenes biology, with emphasis on its virulence factors, antimicrobial resistance, and adaptations to environmental stresses.
Collapse
Affiliation(s)
- Lavious Tapiwa Matereke
- SAMRC Microbial Water Quality Monitoring Centre, University of Fort Hare, Alice 5700, South Africa
- Applied and Environmental Microbiology Research Group, Department of Biochemistry and Microbiology, University of Fort Hare, Alice 5700, South Africa
| | - Anthony Ifeanyi Okoh
- SAMRC Microbial Water Quality Monitoring Centre, University of Fort Hare, Alice 5700, South Africa
- Applied and Environmental Microbiology Research Group, Department of Biochemistry and Microbiology, University of Fort Hare, Alice 5700, South Africa
| |
Collapse
|
37
|
Li G, Wang G, Li M, Li L, Liu H, Sun M, Wen Z. Morin inhibits Listeria monocytogenes virulence in vivo and in vitro by targeting listeriolysin O and inflammation. BMC Microbiol 2020; 20:112. [PMID: 32398085 PMCID: PMC7216731 DOI: 10.1186/s12866-020-01807-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/29/2020] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Listeria monocytogenes (L. monocytogenes) is a global opportunistic intracellular pathogen that can cause many infections, including meningitis and abortion in humans and animals; thus, L. monocytogenes poses a great threat to public safety and the development of the aquaculture industry. The isolation rate of Listeria monocytogenes in fishery products has always been high. And the pore-forming toxin listeriolysin O (LLO) is one of the most important virulence factors of L. monocytogenes. LLO can promote cytosolic bacterial proliferation and help the pathogen evade attacks from the host immune system. In addition, L. monocytogenes infection can trigger a series of severe inflammatory reactions. RESULTS Here, we further confirmed that morin lacking anti-Listeria activity could inhibit LLO oligomerization. We also found that morin can effectively alleviate the inflammation induced by Listeria in vivo and in vitro and exerted an obvious protective effect on infected cells and mice. CONCLUSIONS Morin does not possess anti-Listeria activity, neither does it interfere with secretion of LLO. However, morin inhibits oligomerisation of LLO and morin does reduce the inflammation caused during Listeria infection.
Collapse
Affiliation(s)
- Gen Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, China.,Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Guizhen Wang
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China.,College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Meng Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, China.,Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Li Li
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Hongtao Liu
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Meiyang Sun
- Department of Breast Surgery, Jilin Provincial Cancer Hospital, Changchun, China
| | - Zhongmei Wen
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
38
|
Potential Roles and Functions of Listerial Virulence Factors during Brain Entry. Toxins (Basel) 2020; 12:toxins12050297. [PMID: 32380697 PMCID: PMC7291126 DOI: 10.3390/toxins12050297] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
Although it rarely induces disease in humans, Listeria monocytogenes (Lm) is important due to the frequency of serious pathological conditions—such as sepsis and meningitis—it causes in those few people that do get infected. Virulence factors (VF) of Lm—especially those involved in the passage through multiple cellular barriers of the body, including internalin (Inl) family members and listeriolysin O (LLO)—have been investigated both in vitro and in vivo, but the majority of work was focused on the mechanisms utilized during penetration of the gut and fetoplacental barriers. The role of listerial VF during entry into other organs remain as only partially solved puzzles. Here, we review the current knowledge on the entry of Lm into one of its more significant destinations, the brain, with a specific focus on the role of various VF in cellular adhesion and invasion.
Collapse
|
39
|
La Pietra L, Hudel M, Pillich H, Abu Mraheil M, Berisha B, Aden S, Hodnik V, Lochnit G, Rafiq A, Perniss A, Anderluh G, Chakraborty T. Phosphocholine Antagonizes Listeriolysin O-Induced Host Cell Responses of Listeria monocytogenes. J Infect Dis 2020; 222:1505-1516. [DOI: 10.1093/infdis/jiaa022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/20/2020] [Indexed: 12/20/2022] Open
Abstract
Abstract
Background
Bacterial toxins disrupt plasma membrane integrity with multitudinous effects on host cells. The secreted pore-forming toxin listeriolysin O (LLO) of the intracellular pathogen Listeria monocytogenes promotes egress of the bacteria from vacuolar compartments into the host cytosol often without overt destruction of the infected cell. Intracellular LLO activity is tightly controlled by host factors including compartmental pH, redox, proteolytic, and proteostatic factors, and inhibited by cholesterol.
Methods
Combining infection studies of L. monocytogenes wild type and isogenic mutants together with biochemical studies with purified phospholipases, we investigate the effect of their enzymatic activities on LLO.
Results
Here, we show that phosphocholine (ChoP), a reaction product of the phosphatidylcholine-specific phospholipase C (PC-PLC) of L. monocytogenes, is a potent inhibitor of intra- and extracellular LLO activities. Binding of ChoP to LLO is redox-independent and leads to the inhibition of LLO-dependent induction of calcium flux, mitochondrial damage, and apoptosis. ChoP also inhibits the hemolytic activities of the related cholesterol-dependent cytolysins (CDC), pneumolysin and streptolysin.
Conclusions
Our study uncovers a strategy used by L. monocytogenes to modulate cytotoxic LLO activity through the enzymatic activity of its PC-PLC. This mechanism appears to be widespread and also used by other CDC pore-forming toxin-producing bacteria.
Collapse
Affiliation(s)
- Luigi La Pietra
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Martina Hudel
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Helena Pillich
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Mobarak Abu Mraheil
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Besim Berisha
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Saša Aden
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Vesna Hodnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Günter Lochnit
- Protein Analytics, Institute of Biochemistry, Justus-Liebig University Giessen, Giessen, Germany
| | - Amir Rafiq
- Institute of Anatomy and Cell Biology, Cardiopulmonary Institute, German Center for Lung Research, Justus-Liebig University Giessen, Giessen, Germany
| | - Alexander Perniss
- Institute of Anatomy and Cell Biology, Cardiopulmonary Institute, German Center for Lung Research, Justus-Liebig University Giessen, Giessen, Germany
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Trinad Chakraborty
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| |
Collapse
|
40
|
Pleckaityte M. Cholesterol-Dependent Cytolysins Produced by Vaginal Bacteria: Certainties and Controversies. Front Cell Infect Microbiol 2020; 9:452. [PMID: 31998661 PMCID: PMC6966277 DOI: 10.3389/fcimb.2019.00452] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/12/2019] [Indexed: 01/16/2023] Open
Abstract
Bacterial vaginosis (BV) is a vaginal anaerobic dysbiosis that affects women of reproductive age worldwide. BV is microbiologically characterized by the depletion of vaginal lactobacilli and the overgrowth of anaerobic bacterial species. Accumulated evidence suggests that Gardnerella spp. have a pivotal role among BV-associated bacteria in the initiation and development of BV. However, Gardnerella spp. often colonize healthy women. Lactobacillus iners is considered as a prevalent constituent of healthy vaginal microbiota, and is abundant in BV. Gardnerella spp. and L. iners secrete the toxins vaginolysin (VLY) and inerolysin (INY), which have structural and activity features attributed to cholesterol-dependent cytolysins (CDCs). CDCs are produced by many pathogenic bacteria as virulence factors that participate in various stages of disease progression by forming lytic and non-lytic pores in cell membranes or via pore-independent pathways. VLY is expressed in the majority of Gardnerella spp. isolates; less is known about the prevalence of the gene that encodes INY. INY is a classical CDC; membrane cholesterol acts a receptor for INY. VLY uses human CD59 as its receptor, although cholesterol remains indispensable for VLY pore-forming activity. INY-induced damage of artificial membranes is directly dependent on cholesterol concentration in the bilayer, whereas VLY-induced damage occurs with high levels of membrane cholesterol (>40 mol%). VLY primarily forms membrane-embedded complete rings in the synthetic bilayer, whereas INY forms arciform structures with smaller pore sizes. VLY activity is high at elevated pH, which is characteristic of BV, whereas INY activity is high at more acidic pH, which is specific for a healthy vagina. Increased VLY levels in vaginal mucosa in vivo were associated with clinical indicators of BV. However, experimental evidence is lacking for the specific roles of VLY and INY in BV. The interplay between vaginal bacterial species affects the expression of the gene encoding VLY, thereby modulating the virulence of Gardnerella spp. This review discusses the current evidence for VLY and INY cytolysins, including their structures and activities, factors affecting their expression, and their potential impacts on the progression of anaerobic dysbiosis.
Collapse
Affiliation(s)
- Milda Pleckaityte
- Laboratory of Immunology and Cell Biology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
41
|
Nguyen BN, Portnoy DA. An Inducible Cre- lox System to Analyze the Role of LLO in Listeria monocytogenes Pathogenesis. Toxins (Basel) 2020; 12:E38. [PMID: 31936068 PMCID: PMC7020405 DOI: 10.3390/toxins12010038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 12/14/2019] [Accepted: 12/31/2019] [Indexed: 11/16/2022] Open
Abstract
Listeriolysin O (LLO) is a pore-forming cytolysin that allows Listeria monocytogenes to escape from phagocytic vacuoles and enter the host cell cytosol. LLO is expressed continuously during infection, but it has been a challenge to evaluate the importance of LLO secreted in the host cell cytosol because deletion of the gene encoding LLO (hly) prevents localization of L. monocytogenes to the cytosol. Here, we describe a L. monocytogenes strain (hlyfl) in which hly is flanked by loxP sites and Cre recombinase is under the transcriptional control of the L. monocytogenesactA promoter, which is highly induced in the host cell cytosol. In less than 2 h after infection of bone marrow-derived macrophages (BMMs), bacteria were 100% non-hemolytic. hlyfl grew intracellularly to levels 10-fold greater than wildtype L. monocytogenes and was less cytotoxic. In an intravenous mouse model, 90% of bacteria were non-hemolytic within three hours in the spleen and eight hours in the liver. The loss of LLO led to a 2-log virulence defect in the spleen and a 4-log virulence defect in the liver compared to WT L. monocytogenes. Thus, the production of LLO in the cytosol has significant impact on the pathogenicity of L. monocytogenes.
Collapse
Affiliation(s)
- Brittney N. Nguyen
- Graduate Group in Microbiology, University of California, Berkeley, CA 94720, USA;
| | - Daniel A. Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
42
|
Morton CJ, Sani MA, Parker MW, Separovic F. Cholesterol-Dependent Cytolysins: Membrane and Protein Structural Requirements for Pore Formation. Chem Rev 2019; 119:7721-7736. [DOI: 10.1021/acs.chemrev.9b00090] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Craig J. Morton
- Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Marc-Antoine Sani
- School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Michael W. Parker
- Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Parkville, Victoria 3010, Australia
- St. Vincent’s Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Frances Separovic
- School of Chemistry, Bio21 Institute, University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
43
|
Fotoohi-Ardakani G, Kheirollahi M, Zarei Jaliani H, Noorian M, Ansariniyia H. Targeting MCF-7 Cell Line by Listeriolysin O Pore Forming Toxin Fusion with AHNP Targeted Peptide. Adv Biomed Res 2019; 8:33. [PMID: 31259162 PMCID: PMC6543864 DOI: 10.4103/abr.abr_18_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background Tumor-targeting peptides are attracting subjects in cancer therapy. These peptides, which are widely studied, deliver therapeutic agents to the specific sites of tumors. In this study, we produced a new form of recombinant listeriolysin O (LLO) with genetically fused Anti-HER2/neu peptide (AHNP) sequence adding to its C-terminal end. The aim of the study was to engineer this pore-forming toxin to make it much more specific to tumor cells. Materials and Method and Results Two forms of the toxin (with and without peptide) were subcloned into a bacterial expression plasmid. Subcloning was performed using a polymerase chain reaction (PCR) product as a megaprimer in a quick-change PCR to introduce the whole insert gene into the expression plasmid. After expression of two recombinant forms of LLO in BL21 DE3 cells, purification was performed using Ni-NTA affinity column. MDA-MB-231 and MCF-7 cell lines (as negative and positive controls, respectively) were treated with both LLO toxins to evaluate their cytotoxicity and specificity. The IC50 of LLO on MDA-MB-231 and MCF-7 cells was 21 and 5 ng/ml, respectively. In addition, IC50 for the fusion AHNP-LLO toxin was 140 and 60 ng/ml, respectively. It was found that the cytotoxicity of the new engineered AHNP-LLO toxin has decreased by about 9x compared to the wild-type toxin and the specificity of the AHNP-LLO toxin has been also reduced. Conclusions Results show that the C-terminal of the LLO should not be modified and it seems that N-terminal of the toxin should be preferred for engineering and adding peptide modules.
Collapse
Affiliation(s)
- Gholamreza Fotoohi-Ardakani
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease and Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Medical Genetics, School of Medicine, Protein Engineering Laboratory, Shahidsadoughi University of Medical Sciences, Yazd, Iran
| | - Majid Kheirollahi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease and Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Zarei Jaliani
- Department of Medical Genetics, School of Medicine, Protein Engineering Laboratory, Shahidsadoughi University of Medical Sciences, Yazd, Iran
| | - Mohadese Noorian
- Department of Medical Genetics, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Hossein Ansariniyia
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Science, Yazd, Iran
| |
Collapse
|
44
|
Lu G, Xu L, Zhang T, Deng X, Wang J. A potential bio-control agent from baical skullcap root against listeriosis via the inhibition of sortase A and listeriolysin O. J Cell Mol Med 2019; 23:2042-2051. [PMID: 30585434 PMCID: PMC6378236 DOI: 10.1111/jcmm.14110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 12/01/2018] [Indexed: 01/29/2023] Open
Abstract
Listeria monocytogenes (LM) is a classical model intracellular pathogen and the leading cause of listeriosis, which has long been a global public health issue. The successful infection of LM is related to a series of virulence factors, such as the transpeptidase enzyme sortase A (SrtA) and listeriolysin O (LLO), which are crucial for bacterial internalization and escape from phagosomes respectively. It is speculated that targeting multiple virulence factors may be due to a synergistic effect on listeriosis therapy. In this study, an active flavonoids component of Scutellaria baicalensis Georgi, baicalein, was found to potently block both listerial SrtA catalyzed activity and LLO hemolytic activity within 16 μg/mL. After pretreatment with baicalein, 86.30 (±11.35) % of LM failed to associate with Caco-2 cells compared to the LM without preincubation (regarded as 100% internalization). Furthermore, baicalein addition may aid in bacterial degradation and clearance in macrophagocytes. During a 5 h observation, LM in cells incubated with baicalein showed significantly decreased vacuole escapes and sluggish endocellular growth. In addition, baicalein directly prevented LM-induced cells injury and mice fatality (survival rate from 10.00% to 54.55% in 4 days post-intraperitoneal injection). Taken together, as an antagonist against SrtA and LLO, baicalein can be further developed into a biotherapeutic agent for listeriosis.
Collapse
Affiliation(s)
- Gejin Lu
- Center of Infection and Immunity, First HospitalJilin UniversityChangchunJilin, 130021China
- Key Laboratory of Zoonosis, Ministry of EducationInstitute of Zoonosis, College of Veterinary Medicine, Jilin UniversityChangchunJilin, 130062China
| | - Lei Xu
- Center of Infection and Immunity, First HospitalJilin UniversityChangchunJilin, 130021China
- Key Laboratory of Zoonosis, Ministry of EducationInstitute of Zoonosis, College of Veterinary Medicine, Jilin UniversityChangchunJilin, 130062China
| | - Tong Zhang
- Center of Infection and Immunity, First HospitalJilin UniversityChangchunJilin, 130021China
- Key Laboratory of Zoonosis, Ministry of EducationInstitute of Zoonosis, College of Veterinary Medicine, Jilin UniversityChangchunJilin, 130062China
| | - Xuming Deng
- Center of Infection and Immunity, First HospitalJilin UniversityChangchunJilin, 130021China
- Key Laboratory of Zoonosis, Ministry of EducationInstitute of Zoonosis, College of Veterinary Medicine, Jilin UniversityChangchunJilin, 130062China
| | - Jianfeng Wang
- Center of Infection and Immunity, First HospitalJilin UniversityChangchunJilin, 130021China
- Key Laboratory of Zoonosis, Ministry of EducationInstitute of Zoonosis, College of Veterinary Medicine, Jilin UniversityChangchunJilin, 130062China
| |
Collapse
|
45
|
Nguyen BN, Peterson BN, Portnoy DA. Listeriolysin O: A phagosome-specific cytolysin revisited. Cell Microbiol 2019; 21:e12988. [PMID: 30511471 DOI: 10.1111/cmi.12988] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
Abstract
Listeriolysin O (LLO) is an essential determinant of Listeria monocytogenes pathogenesis that mediates the escape of L. monocytogenes from host cell vacuoles, thereby allowing replication in the cytosol without causing appreciable cell death. As a member of the cholesterol-dependent cytolysin (CDC) family of pore-forming toxins, LLO is unique in that it is secreted by a facultative intracellular pathogen, whereas all other CDCs are produced by pathogens that are largely extracellular. Replacement of LLO with other CDCs results in strains that are extremely cytotoxic and 10,000-fold less virulent in mice. LLO has structural and regulatory features that allow it to function intracellularly without causing cell death, most of which map to a unique N-terminal region of LLO referred to as the proline, glutamic acid, serine, threonine (PEST)-like sequence. Yet, while LLO has unique properties required for its intracellular site of action, extracellular LLO, like other CDCs, affects cells in a myriad of ways. Because all CDCs form pores in cholesterol-containing membranes that lead to rapid Ca2+ influx and K+ efflux, they consequently trigger a wide range of host cell responses, including mitogen-activated protein kinase activation, histone modification, and caspase-1 activation. There is no debate that extracellular LLO, like all other CDCs, can stimulate multiple cellular activities, but the primary question we wish to address in this perspective is whether these activities contribute to L. monocytogenes pathogenesis.
Collapse
Affiliation(s)
- Brittney N Nguyen
- Graduate Group in Microbiology, University of California, Berkeley, Berkeley, California
| | - Bret N Peterson
- Graduate Group in Microbiology, University of California, Berkeley, Berkeley, California
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California.,Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, California
| |
Collapse
|
46
|
Lam JGT, Song C, Seveau S. High-throughput Measurement of Plasma Membrane Resealing Efficiency in Mammalian Cells. J Vis Exp 2019. [PMID: 30663635 DOI: 10.3791/58351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
In their physiological environment, mammalian cells are often subjected to mechanical and biochemical stresses that result in plasma membrane damage. In response to these damages, complex molecular machineries rapidly reseal the plasma membrane to restore its barrier function and maintain cell survival. Despite 60 years of research in this field, we still lack a thorough understanding of the cell resealing machinery. With the goal of identifying cellular components that control plasma membrane resealing or drugs that can improve resealing, we have developed a fluorescence-based high-throughput assay that measures the plasma membrane resealing efficiency in mammalian cells cultured in microplates. As a model system for plasma membrane damage, cells are exposed to the bacterial pore-forming toxin listeriolysin O (LLO), which forms large 30-50 nm diameter proteinaceous pores in cholesterol-containing membranes. The use of a temperature-controlled multi-mode microplate reader allows for rapid and sensitive spectrofluorometric measurements in combination with brightfield and fluorescence microscopy imaging of living cells. Kinetic analysis of the fluorescence intensity emitted by a membrane impermeant nucleic acid-binding fluorochrome reflects the extent of membrane wounding and resealing at the cell population level, allowing for the calculation of the cell resealing efficiency. Fluorescence microscopy imaging allows for the enumeration of cells, which constitutively express a fluorescent chimera of the nuclear protein histone 2B, in each well of the microplate to account for potential variations in their number and allows for eventual identification of distinct cell populations. This high-throughput assay is a powerful tool expected to expand our understanding of membrane repair mechanisms via screening for host genes or exogenously added compounds that control plasma membrane resealing.
Collapse
Affiliation(s)
- Jonathan G T Lam
- Department of Microbial Infection and Immunity, The Ohio State University; Department of Microbiology, The Ohio State University; Infectious Diseases Institute, The Ohio State University
| | - Chi Song
- Division of Biostatistics, College of Public Health, The Ohio State University
| | - Stephanie Seveau
- Department of Microbial Infection and Immunity, The Ohio State University; Department of Microbiology, The Ohio State University; Infectious Diseases Institute, The Ohio State University;
| |
Collapse
|
47
|
An ATG16L1-dependent pathway promotes plasma membrane repair and limits Listeria monocytogenes cell-to-cell spread. Nat Microbiol 2018; 3:1472-1485. [PMID: 30478389 DOI: 10.1038/s41564-018-0293-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022]
Abstract
Plasma membrane integrity is essential for the viability of eukaryotic cells. In response to bacterial pore-forming toxins, disrupted regions of the membrane are rapidly repaired. However, the pathways that mediate plasma membrane repair are unclear. Here we show that autophagy-related (ATG) protein ATG16L1 and its binding partners ATG5 and ATG12 are required for plasma membrane repair through a pathway independent of macroautophagy. ATG16L1 is required for lysosome fusion with the plasma membrane and blebbing responses that promote membrane repair. ATG16L1 deficiency causes accumulation of cholesterol in lysosomes that contributes to defective membrane repair. Cell-to-cell spread by Listeria monocytogenes requires membrane damage by the bacterial toxin listeriolysin O, which is restricted by ATG16L1-dependent membrane repair. Cells harbouring the ATG16L1 T300A allele associated with inflammatory bowel disease were also found to accumulate cholesterol and be defective in repair, linking a common inflammatory disease to plasma membrane integrity. Thus, plasma membrane repair could be an important therapeutic target for the treatment of bacterial infections and inflammatory disorders.
Collapse
|
48
|
Christie MP, Johnstone BA, Tweten RK, Parker MW, Morton CJ. Cholesterol-dependent cytolysins: from water-soluble state to membrane pore. Biophys Rev 2018; 10:1337-1348. [PMID: 30117093 DOI: 10.1007/s12551-018-0448-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/07/2018] [Indexed: 12/22/2022] Open
Abstract
The cholesterol-dependent cytolysins (CDCs) are a family of bacterial toxins that are important virulence factors for a number of pathogenic Gram-positive bacterial species. CDCs are secreted as soluble, stable monomeric proteins that bind specifically to cholesterol-rich cell membranes, where they assemble into well-defined ring-shaped complexes of around 40 monomers. The complex then undergoes a concerted structural change, driving a large pore through the membrane, potentially lysing the target cell. Understanding the details of this process as the protein transitions from a discrete monomer to a complex, membrane-spanning protein machine is an ongoing challenge. While many of the details have been revealed, there are still questions that remain unanswered. In this review, we present an overview of some of the key features of the structure and function of the CDCs, including the structure of the secreted monomers, the process of interaction with target membranes, and the transition from bound monomers to complete pores. Future directions in CDC research and the potential of CDCs as research tools will also be discussed.
Collapse
Affiliation(s)
- Michelle P Christie
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Bronte A Johnstone
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Rodney K Tweten
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Michael W Parker
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
- Australian Cancer Research Foundation Rational Drug Discovery Centre, St Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia.
| | - Craig J Morton
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
49
|
Gilbert RJC, Bayley H, Anderluh G. Membrane pores: from structure and assembly, to medicine and technology. Philos Trans R Soc Lond B Biol Sci 2018. [PMID: 28630148 DOI: 10.1098/rstb.2016.0208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Robert J C Gilbert
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Hagan Bayley
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| |
Collapse
|
50
|
The Ethanolamine Permease EutH Promotes Vacuole Adaptation of Salmonella enterica and Listeria monocytogenes during Macrophage Infection. Infect Immun 2018. [PMID: 29531136 DOI: 10.1128/iai.00172-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ethanolamine is a ubiquitous and essential molecule within a host. Significantly, bacterial pathogens exploit ethanolamine during infection to promote growth and regulate virulence. The ethanolamine permease EutH is dispensable for growth in vitro under standard conditions, whereas EutH is required for ethanolamine utilization at low pH. These findings suggested a model in which EutH facilitates diffusion of ethanolamine into the bacterial cell in acidic environments. To date, the ecological significance of this model has not been thoroughly investigated, and the importance of EutH to bacterial growth under physiologically relevant conditions is not known. During infection, immune cells internalize invading bacteria within an acidic, nutrient-depleted vacuole called the phagosome. Here, we investigated the hypothesis that EutH promotes bacterial survival following phagocytosis. Our findings indicate that EutH is important for survival and replication of the facultative intracellular pathogens Salmonella enterica serovar Typhimurium and Listeria monocytogenes during prolonged or transient exposure to the phagosome, respectively. Furthermore, in agreement with EutH being important in the acidic environment, neutralization of the vacuole abolished the requirement for EutH. Significantly, consistent with a role for EutH in promoting intramacrophage survival, EutH was not required during S Typhimurium local intestinal infection but specifically conferred an advantage upon dissemination to peripheral organs. These findings reveal a physiologically relevant and conserved role for EutH in spatiotemporal niche adaptation during infection.
Collapse
|