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
|
Manyi-Loh CE, Lues R. Listeria monocytogenes and Listeriosis: The Global Enigma. Foods 2025; 14:1266. [PMID: 40238523 PMCID: PMC11989209 DOI: 10.3390/foods14071266] [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: 12/17/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Listeria monocytogenes is an intracellular, Gram-positive, non-spore-forming, non-encapsulated, facultative anaerobic, rod-shaped, and psychrotrophic food-borne pathogen that causes the infection, listeriosis, thus it attracts great attention following listeriosis outbreaks, which are often associated with high mortality rates. The prevalence of listeriosis is quite low globally; however, the most recent and deadliest outbreak occurred in South Africa, during which 216 persons lost their lives. L. monocytogenes is endowed with the potential to multiply through a wide range of harsh environmental conditions, forming biofilms on varying surfaces in the food industry, as well as having persistent and antibiotic-resistant cells, which pose a major threat and burden to the ready-to-eat food industry. A more frustrating characteristic of this bacterium is its strain divergence, alongside an increased level of antibiotic resistance registered among the strains of L. monocytogenes recovered from food, humans, and environmental sources, especially to those antibiotics involved in the treatment of human listeriosis. Antibiotic resistance exerted by and among pathogenic food-borne microbes is an ongoing public health menace that continues to be an issue. Against this background, a thorough search into different databases using various search engines was performed, which led to the gathering of salient information that was organised, chronologically, based on Listeria monocytogenes and listeriosis. Altogether, the findings elaborated in this study present up-to date knowledge on different aspects of this pathogen which will improve our understanding of the mystery associated with it and the ways to prevent and control its dissemination through ready-to-eat foods. In addition, constant monitoring of the antibiotic resistance profiles of strains of L. monocytogenes from varying sources detected changes, giving an update on the trend in antibiotic resistance. Overall, monitoring of bacterial contamination serves as the key aspect in the control of the food safety output in the food industry.
Collapse
Affiliation(s)
- Christy E. Manyi-Loh
- Centre for Applied Food Sustainability and Biotechnology, Central University of Technology, Bloemfontein X9301, South Africa;
| | | |
Collapse
|
3
|
Jiao J, Ma Z, Li N, Duan F, Cai X, Zuo Y, Li J, Meng Q, Qiao J. Listeria monocytogenes Modulates Macrophage Inflammatory Responses to Facilitate Its Intracellular Survival by Manipulating Macrophage-Derived Exosomal ncRNAs. Microorganisms 2025; 13:410. [PMID: 40005775 PMCID: PMC11858176 DOI: 10.3390/microorganisms13020410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Exosomes are nanoscale vesicles secreted by cells that play vital regulatory roles in intercellular communication and immune responses. Listeria monocytogenes (L. Monocytogenes, LM) is a notable Gram-positive intracellular parasitic bacterium that infects humans and diverse animal species. However, the specific biological function of exosomes secreted by macrophages during L. Monocytogenes infection (hereafter EXO-LM) remains elusive. Here, we discovered that EXO-LM stimulated the secretion of inflammation-associated cytokines by macrophages, facilitating the intracellular survival of L. monocytogenes within macrophages. Transcriptomic analysis shows that EXO-LM significantly upregulates immune recognition and inflammation-related signaling pathways in macrophages. Furthermore, a ceRNA regulatory network comprising exosomal ncRNAs and macrophage RNAs was constructed through EXO-LM transcriptome sequencing. Utilizing bioinformatics and dual-luciferase reporter assays, we identified two potential binding sites between lncRNA Rpl13a-213 and miR-132-3p. Cell transfection experiments demonstrated that Rpl13a-213 overexpression augmented pro-inflammatory cytokine expression in macrophages, in contrast to the suppression by miR-132-3p overexpression. The decrease in Rpl13a-213 upon EXO-LM stimulation enhances miR-132-3p expression, dampening the inflammatory response in macrophages and aiding L. monocytogenes intracellular survival. This study unveils the immunomodulatory function of exosomal ncRNAs originating from macrophages, which provides fresh perspectives into the mechanisms underlying macrophage inflammatory response regulation by L. monocytogenes-infected cell-derived exosomes.
Collapse
Affiliation(s)
- Jian Jiao
- College of Animal Science & Technology, Shihezi University, Shihezi 832003, China; (J.J.); (Z.M.); (N.L.)
| | - Zhongmei Ma
- College of Animal Science & Technology, Shihezi University, Shihezi 832003, China; (J.J.); (Z.M.); (N.L.)
| | - Nengxiu Li
- College of Animal Science & Technology, Shihezi University, Shihezi 832003, China; (J.J.); (Z.M.); (N.L.)
| | - Fushuang Duan
- College of Animal Science & Technology, Shihezi University, Shihezi 832003, China; (J.J.); (Z.M.); (N.L.)
| | - Xuepeng Cai
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Yufei Zuo
- College of Animal Science & Technology, Shihezi University, Shihezi 832003, China; (J.J.); (Z.M.); (N.L.)
| | - Jie Li
- College of Animal Science & Technology, Shihezi University, Shihezi 832003, China; (J.J.); (Z.M.); (N.L.)
| | - Qingling Meng
- College of Animal Science & Technology, Shihezi University, Shihezi 832003, China; (J.J.); (Z.M.); (N.L.)
| | - Jun Qiao
- College of Animal Science & Technology, Shihezi University, Shihezi 832003, China; (J.J.); (Z.M.); (N.L.)
| |
Collapse
|
4
|
Nunzi I, D'Achille G, Dhaouadi N, Marcheggiani F, Licini C, Di Vincenzo M, Orciani M, Morroni G, Marchi S. Monitoring cellular dynamics upon infection using a holotomography-based approach. Methods Cell Biol 2025; 194:109-118. [PMID: 40058955 DOI: 10.1016/bs.mcb.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
Many intracellular bacteria interfere with mitochondrial dynamics or target other organelles, thereby inducing a specific cellular response that could emerge as a strategy of the pathogen to ensure its survival, or as a form of defense employed by the host cell to restrict dissemination. In this context, the concomitant monitoring of both pathogen migration and (intra)cellular dynamics in live cells emerges as a pivotal aspect for the comprehension of the infection sequence and to visualize the pathogen-mediated remodeling that could occur to the entire cellular system. Holotomographic microscopy can be used to achieve this goal, allowing the simultaneous analysis of both bacterial movement and intracellular alteration for extended periods of time, with high spatial resolution and avoiding side-effects due to phototoxicity. Here we provide a holotomography-based approach to detect Listeria monocytogenes dynamics and its effects on the entire cellular system at morphological level.
Collapse
Affiliation(s)
- Ilaria Nunzi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Gloria D'Achille
- Microbiology Unit, Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy
| | - Nada Dhaouadi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Fabio Marcheggiani
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Caterina Licini
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Mariangela Di Vincenzo
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Gianluca Morroni
- Microbiology Unit, Department of Biomedical Sciences and Public Health, Marche Polytechnic University, Ancona, Italy.
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy; Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy.
| |
Collapse
|
5
|
Mantovam VB, Dos Santos DF, Giola Junior LC, Landgraf M, Pinto UM, Todorov SD. Listeria monocytogenes, Salmonella spp., and Staphylococcus aureus: Threats to the Food Industry and Public Health. Foodborne Pathog Dis 2025. [PMID: 39761068 DOI: 10.1089/fpd.2024.0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
Foodborne pathogens have always been of public health concern and represent safety issues for food processors. These pathogens develop new ways to overcome antibiotics, survive in different environmental conditions, and the ability to reproduce in many hostile environments configure them as serious health hazards. Considering the huge number of microorganisms, three bacterial representatives were selected to provide a better knowledge about the question of which one is the worst enemy for humans, from the food industry point of view, taking into consideration their multiplication specificity, virulence, and resistance. As we constantly are exposed to these pathogens in our nutritional habits, this overview aims to summarize the most relevant characteristics associated with the pathogenicity, clinical symptoms and most importantly, how deadly Listeria monocytogenes, Salmonella spp., and Staphylococcus aureus can be in the hospital and the food industry, by comparing among them. Overall, the microbiological knowledge clearly suggests that while all three pathogens are dangerous, L. monocytogenes presents the highest risk of death due to their ability to cause severe complications in vulnerable populations as it presents a range of virulence factors that facilitate evasion of the immune system and cytological effects. Additionally, it shows great resistance to standard food processing and preservation techniques, making it one of the most difficult pathogens to control. Understanding the risks and characteristics of these foodborne pathogens is essential for implementing effective control measures to prevent their occurrence in food products and to promote public health.
Collapse
Affiliation(s)
- Vinicius B Mantovam
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - David F Dos Santos
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Luis C Giola Junior
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Mariza Landgraf
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Uelinton M Pinto
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Svetoslav D Todorov
- Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
- ProBacLab, Laboratório de Microbiologia de Alimentos, Departamento de Alimentos e Nutrição Experimental, FoRC, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Pereira-Dutra FS, Souza EK, Souza TS, Goltara-Gomes TC, Ferraro-Moreira F, Palhinha L, Cunha-Fernandes T, Rajão MA, Silva AR, Bozza PT. Accumulation of lipid droplets induced by Listeria monocytogenes in macrophages: implications for survival and evasion of innate immunity. J Leukoc Biol 2024; 116:1364-1371. [PMID: 38727078 DOI: 10.1093/jleuko/qiae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 11/28/2024] Open
Abstract
Listeriosis, caused by Listeria monocytogenes (L.m.), poses a significant public health concern as one of the most severe foodborne diseases. The pathogenesis of L.m. involves critical steps such as phagosome rupture and escape upon internalization. Throughout infection, L.m. influences various host processes, including lipid metabolism pathways, yet the role of lipid droplets (LDs) remains unclear. Here, we reported a rapid, time-dependent increase in LD formation in macrophages induced by L.m. LD biogenesis was found to be dependent on L.m. viability and virulence genes, particularly on the activity of the pore-forming protein listeriolysin O (LLO). The prevention of LD formation by inhibiting diacylglycerol O-acyltransferase 1 (DGAT1) and cytosolic phospholipase A2 (cPLA2) significantly reduced intracellular bacterial survival, impaired prostaglandin E2 synthesis, and decreased interleukin-10 production. Additionally, inhibiting LD formation led to increased levels of tumor necrosis factor α and interferon β. Collectively, our data suggest a role for LDs in promoting L.m. cell survival and evasion within macrophages.
Collapse
Affiliation(s)
- Filipe S Pereira-Dutra
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Ellen K Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Tamyris S Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Taynná C Goltara-Gomes
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Felipe Ferraro-Moreira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Lohanna Palhinha
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Tamires Cunha-Fernandes
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Matheus A Rajão
- Program of Immunology and Tumor Biology, National Cancer Institute, INCA, André Cavalcanti St, Rio de Janeiro, RJ, 20231-050, Brazil
| | - Adriana R Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| |
Collapse
|
7
|
Xu DZ, Tan QH. Infection with Listeria monocytogenes meningoencephalitis: A case report. World J Clin Cases 2024; 12:6629-6634. [PMID: 39600479 PMCID: PMC11514345 DOI: 10.12998/wjcc.v12.i33.6629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Listeria meningitis is an infectious disease of the central nervous system caused by Listeria monocytogenes. This bacterium is widely present in the natural environment and can be transmitted through channels such as food and water. Patients usually show symptoms such as fever, headache, and neck stiffness. In severe cases, coma, convulsions, or even death may occur. Traditional diagnostic methods, such as cerebrospinal fluid (CSF) culture and serological tests, have certain limitations. Although CSF culture is the "gold standard" for diagnosis, it is time-consuming and has a relatively low positivity rate. Serological detection may also result in false positive or false negative results. The emergence of metagenomic sequencing (mNGS) technology has led to a significant breakthrough in diagnosing Listeria meningitis, allowing quick and accurate detection of various pathogens in samples. CASE SUMMARY Here, we present the case of a previously healthy 64-year-old woman diagnosed with Listeria meningitis using mNGS. She was successfully treated with intravenous ampicillin and meropenem, without any complications. CONCLUSION Listeria meningitis must be considered, especially in patients who fail to show improvement with first-line antibiotic treatments. mNGS significantly reduces the diagnosis time, supporting timely treatment of patients.
Collapse
Affiliation(s)
- Da-Zhen Xu
- Department of Nursing, Shanghai Sixth People’s Hospital, Shanghai 200233, China
| | - Quan-Hui Tan
- Department of Infectious Disease, Shanghai Sixth People’s Hospital, Shanghai 200233, China
| |
Collapse
|
8
|
Andriyanov P, Zhurilov P, Menshikova A, Tutrina A, Yashin I, Kashina D. Large-scale genomic analysis of Elizabethkingia anophelis. BMC Genomics 2024; 25:1015. [PMID: 39472795 PMCID: PMC11523902 DOI: 10.1186/s12864-024-10921-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
The recent emergence of Elizabethkingia anophelis as a human pathogen is a major concern for global public health. This organism has the potential to cause severe infections and has inherent antimicrobial resistance. The potential for widespread outbreaks and rapid global spread highlights the critical importance of understanding the biology and transmission dynamics of this infectious agent. We performed a large-scale analysis of available 540 E. anophelis, including one novel strain isolated from raw milk and sequenced in this study. Pan-genome analysis revealed an open and diverse pan-genome in this species, characterized by the presence of many accessory genes. This suggests that the species has a high level of adaptability and can thrive in a variety of environments. Phylogenetic analysis has also revealed a complex population structure, with limited source-lineage correlation. We identified diverse antimicrobial resistance factors, including core-genome and accessory ones often associated with mobile genetic elements within specific lineages. Mobilome analysis revealed a dynamic landscape primarily composed of genetic islands, integrative and conjugative elements, prophage elements, and small portion of plasmids emphasizing a complex mechanism of horizontal gene transfer. Our study underscores the adaptability of E. anophelis, characterized by a diverse range of antimicrobial resistance genes, putative virulence factors, and genes enhancing fitness. This adaptability is also supported by the organism's ability to acquire genetic material through horizontal gene transfer, primarily facilitated by mobile genetic elements such as integrative and conjugative elements (ICEs). The potential for rapid evolution of this emerging pathogen poses a significant challenge to public health efforts.
Collapse
Affiliation(s)
- Pavel Andriyanov
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia.
| | - Pavel Zhurilov
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Alena Menshikova
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Anastasia Tutrina
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Ivan Yashin
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Daria Kashina
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
9
|
Kwon H, Lim DJ, Choi C. Prevention of foodborne viruses and pathogens in fresh produce and root vegetables. ADVANCES IN FOOD AND NUTRITION RESEARCH 2024; 113:219-285. [PMID: 40023562 DOI: 10.1016/bs.afnr.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Every year, 1 in 10 people suffers from food poisoning, and in recent years, the highest number of foodborne outbreaks has been attributed to roots/underground vegetables and fresh produce. Major pathogens include as Escherichia coli, Salmonella enterica, Listeria monocytogenes, Human Norovirus, Hepatitis A virus and Cyclospora. The primary sources of contamination for agriculture products stem from uncontrolled exposure to soil, water, and animal waste. Contamination can occur in various ways during food cultivation, harvesting, processing, and distribution. Mechanical washing and disinfection are primarily employed as practices to control biological contaminants such as bacteria, viruses, and parasites. Current practices may encounter challenges such as microbial resistance to disinfectants or antibiotics, and the cleaning effectiveness could be compromised due to the internalization of bacteria and viruses into some plants. High-pressure processing, pulse electric fields, and cold plasma are environmentally friendly technologies, albeit with associated costs. Low-temperature sterilization technologies capable of controlling biological contaminants, such as bacteria and viruses, play a crucial role in preventing food safety issues. Compared to conventional cleaning methods, these technologies are effective in controlling microorganisms that are strongly attached to the food surface or internalized due to damage. Periodic surveillance is essential to ensure the overall microbiological safety of fresh produce and root vegetables.
Collapse
Affiliation(s)
- Hyojin Kwon
- Department of Food Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do, Republic of Korea
| | - Dong Jae Lim
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do, Republic of Korea
| | - Changsun Choi
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
10
|
Qi Y, Cao Q, Zhao X, Tian C, Li T, Shi W, Wei H, Song C, Xue H, Gou H. Comparative genomic analysis of pathogenic factors of Listeria spp. using whole-genome sequencing. BMC Genomics 2024; 25:935. [PMID: 39375592 PMCID: PMC11457443 DOI: 10.1186/s12864-024-10849-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
Listeria monocytogenes is an important foodborne pathogen known for causing listeriosis. To gain insights into the pathogenicity, genetic characterization, and evolution of various Listeria species, in vitro cell adhesion and invasion ability assays and whole-genome sequencing were performed using four Listeria strains isolated from livestock and poultry slaughterhouses. The four Listeria strains exhibited adhesion and invasion abilities in Caco-2 and RAW264.7 cells. Pathogenic Liv1-1 and Lm2-20 had higher adhesion ability, but non-pathogenic Lin4-99 was more invasive than Lm2-20 (p < 0.05). Genetic characterization revealed the presence of a single chromosome without plasmid across four strains with similar whole-genome sizes and G + C% content. Analysis of key pathogenic genes underscored the presence of multiple virulence genes among the four Listeria strains. In contrast, non-pathogenic Listeria lacked LIPI-1, LIPI-2, and LIPI-3 genes, which could possibly be the cause of their non-pathogenicity despite their in vitro cell adhesion and invasion abilities. Thus, genetic determinants of Listeria do not necessarily predict cell adhesion and/or invasive ability in vitro. This study presents a comprehensive comparative genome-wide analysis of four Listeria strains, offering invaluable insights into the pathogenesis of the Listeria genus.
Collapse
Affiliation(s)
- Yumei Qi
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Qing Cao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xuehui Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Changqing Tian
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Tianhao Li
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Wenjing Shi
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Huilin Wei
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Chen Song
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Huiwen Xue
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Huitian Gou
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
| |
Collapse
|
11
|
Sembada AA, Theda Y, Faizal A. Duckweeds as edible vaccines in the animal farming industry. 3 Biotech 2024; 14:222. [PMID: 39247453 PMCID: PMC11379843 DOI: 10.1007/s13205-024-04074-8] [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: 06/03/2024] [Accepted: 08/29/2024] [Indexed: 09/10/2024] Open
Abstract
Animal diseases are among the most debilitating issues in the animal farming industry, resulting in decreased productivity and product quality worldwide. An emerging alternative to conventional injectable vaccines is edible vaccines, which promise increased delivery efficiency while maintaining vaccine effectiveness. One of the most promising platforms for edible vaccines is duckweeds, due to their high growth rate, ease of transformation, and excellent nutritional content. This review explores the potential, feasibility, and advantages of using duckweeds as platforms for edible vaccines. Duckweeds have proven to be superb feed sources, as evidenced by numerous improvements in both quantity (e.g., weight gain) and quality (e.g., yolk pigmentation). In terms of heterologous protein production, duckweeds, being plants, are capable of expressing proteins with complex structures and post-translational modifications. Research efforts have focused on the development of duckweed-based edible vaccines, including those against avian influenza, tuberculosis, Newcastle disease, and mastitis, among others. As with any emerging technology, the development of duckweeds as a platform for edible vaccines is still in its early stages compared to well-established injectable vaccines. It is evident that more proof-of-concept studies are required to bring edible vaccines closer to the current standards of conventional vaccines. Specifically, the duckweed expression system needs further development in areas such as yield and growth rate, especially when compared to bacterial and mammalian expression systems. Continued efforts in this field could lead to breakthroughs that significantly improve the resilience of the animal farming industry against disease threats.
Collapse
Affiliation(s)
- Anca Awal Sembada
- Research Center for New and Renewable Energy, Bandung Institute of Technology, Bandung, 40132 Indonesia
- Forestry Technology Research Group, School of Life Sciences and Technology, Bandung Institute of Technology, Bandung, 40132 Indonesia
| | - Yohanes Theda
- Department of Biochemical Engineering, University College London, London, WC1E 6BT UK
| | - Ahmad Faizal
- Plant Science and Biotechnology Research Group, School of Life Sciences and Technology, Bandung Institute of Technology, Bandung, 40132 Indonesia
| |
Collapse
|
12
|
Gul A, Pewe LL, Willems P, Mayer R, Thery F, Asselman C, Aernout I, Verbeke R, Eggermont D, Van Moortel L, Upton E, Zhang Y, Boucher K, Miret-Casals L, Demol H, De Smedt SC, Lentacker I, Radoshevich L, Harty JT, Impens F. Immunopeptidomics Mapping of Listeria monocytogenes T Cell Epitopes in Mice. Mol Cell Proteomics 2024; 23:100829. [PMID: 39147027 PMCID: PMC11414675 DOI: 10.1016/j.mcpro.2024.100829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/21/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024] Open
Abstract
Listeria monocytogenes is a foodborne intracellular bacterial model pathogen. Protective immunity against Listeria depends on an effective CD8+ T cell response, but very few T cell epitopes are known in mice as a common animal infection model for listeriosis. To identify epitopes, we screened for Listeria immunopeptides presented in the spleen of infected mice by mass spectrometry-based immunopeptidomics. We mapped more than 6000 mouse self-peptides presented on MHC class I molecules, including 12 high confident Listeria peptides from 12 different bacterial proteins. Bacterial immunopeptides with confirmed fragmentation spectra were further tested for their potential to activate CD8+ T cells, revealing VTYNYINI from the putative cell wall surface anchor family protein LMON_0576 as a novel bona fide peptide epitope. The epitope showed high biological potency in a prime boost model and can be used as a research tool to probe CD8+ T cell responses in the mouse models of Listeria infection. Together, our results demonstrate the power of immunopeptidomics for bacterial antigen identification.
Collapse
Affiliation(s)
- Adillah Gul
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lecia L Pewe
- Department of Pathology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA
| | - Patrick Willems
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Center for Plant Systems Biology, VIB, Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Rupert Mayer
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Fabien Thery
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Caroline Asselman
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Ilke Aernout
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Rein Verbeke
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Denzel Eggermont
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Laura Van Moortel
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Ellen Upton
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Yifeng Zhang
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA
| | - Katie Boucher
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Laia Miret-Casals
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Hans Demol
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA; Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA.
| | - John T Harty
- Department of Pathology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium.
| |
Collapse
|
13
|
Cho AJ, Han S, Nahar S, Her E, Kang JG, Ha SD. Synergistic effects of ε-poly-l-lysine and lysozyme against Pseudomonas aeruginosa and Listeria monocytogenes biofilms on beef and food contact surfaces. Meat Sci 2024; 214:109534. [PMID: 38749270 DOI: 10.1016/j.meatsci.2024.109534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/01/2024] [Accepted: 05/05/2024] [Indexed: 05/28/2024]
Abstract
This study investigated the synergistic effects of ε-poly- L -lysine (ε-PL) and lysozyme against P. aeruginosa and L. monocytogenes biofilms. Single-culture biofilms of two bacteria were formed on silicone rubber (SR), stainless steel (SS), and beef surfaces and then treated with lysozyme (0.05-5 mg/mL) and ε-PL at minimum inhibitory concentrations (MICs) of 1 to 4 separately or in combination. On the SR surface, P. aeruginosa biofilm was reduced by 1.4 and 1.9 log CFU/cm2 within 2 h when treated with lysozyme (5 mg/mL) and ε-PL (4 MIC), respectively, but this reduction increased significantly to 4.1 log CFU/cm2 (P < 0.05) with the combined treatment. On beef surface, P. aeruginosa and L. monocytogenes biofilm was reduced by 4.2-5.0, and 3.3-4.2 log CFU/g when lysozyme was combined with 1, 2, and 4 MIC of ε-PL at 25 °C, respectively. Compared to 5 mg/mL lysozyme alone, the combined treatment with 1, 2, and 4 MIC of ε-PL on beef surface achieved additional reduction against P. aeruginosa biofilm of 0.5, 0.8, and 0.7 log CFU/g, respectively, at 25 °C. In addition, 0.25 mg/mL lysozyme and 0.5 MIC of ε-PL significantly (P < 0.05) suppressed the quorum-sensing (agrA) and virulence-associated (hlyA and prfA) genes of L. monocytogenes.
Collapse
Affiliation(s)
- Ah Jin Cho
- Department of Food Safety and Regulatory Science, Advanced Food Safety Research Group, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Sangha Han
- Department of Food Safety and Regulatory Science, Advanced Food Safety Research Group, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Shamsun Nahar
- NextGen Precision Health, University of Missouri, Columbia, USA
| | - Eun Her
- Department of Food Safety and Regulatory Science, Advanced Food Safety Research Group, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - June Gu Kang
- Department of Food Safety and Regulatory Science, Advanced Food Safety Research Group, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Sang-Do Ha
- Department of Food Safety and Regulatory Science, Advanced Food Safety Research Group, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea.
| |
Collapse
|
14
|
Feodorova VA, Zaitsev SS, Khizhnyakova MA, Lavrukhin MS, Saltykov YV, Zaberezhny AD, Larionova OS. Complete genome of the Listeria monocytogenes strain AUF, used as a live listeriosis veterinary vaccine. Sci Data 2024; 11:643. [PMID: 38886393 PMCID: PMC11183264 DOI: 10.1038/s41597-024-03440-8] [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: 08/08/2023] [Accepted: 05/28/2024] [Indexed: 06/20/2024] Open
Abstract
Listeria monocytogenes (Lm) is a highly pathogenic bacterium that can cause listeriosis, a relatively rare food-borne infectious disease that affects farm, domestic, wild animals and humans as well. The infected livestock is the frequent sources of Lm. Vaccination is one of the methods of controlling listeriosis in target farm animals to prevent Lm-associated food contamination. Here we report the complete sequence of the Lm strain AUF attenuated from a fully-virulent Lm strain by ultraviolet irradiation, successfully used since the 1960s as a live whole-cell veterinary vaccine. The de novo assembled genome consists of a circular chromosome of 2,942,932 bp length, including more than 2,800 CDSs, 17 pseudogenes, 5 antibiotic resistance genes, and 56/92 virulence genes. Two wild Lm strains, the EGD and the 10403S that is also used in cancer Immunotherapy, were the closest homologs for the Lm strain AUF. Although all three strains belonged to different sequence types (ST), namely ST12, ST85, and ST1538, they were placed in the same genetic lineage II, CC7.
Collapse
Affiliation(s)
- Valentina A Feodorova
- Laboratory for Fundamental and Applied Research, Saratov State University of Genetics, Biotechnology and Engineering named after N.I. Vavilov, Saratov, Russia.
- Department for Microbiology and Biotechnology, Saratov State University of Genetics, Biotechnology and Engineering named after N.I. Vavilov, Saratov, Russia.
| | - Sergey S Zaitsev
- Laboratory for Fundamental and Applied Research, Saratov State University of Genetics, Biotechnology and Engineering named after N.I. Vavilov, Saratov, Russia
| | - Mariya A Khizhnyakova
- Laboratory for Fundamental and Applied Research, Saratov State University of Genetics, Biotechnology and Engineering named after N.I. Vavilov, Saratov, Russia
| | - Maxim S Lavrukhin
- Laboratory for Fundamental and Applied Research, Saratov State University of Genetics, Biotechnology and Engineering named after N.I. Vavilov, Saratov, Russia
| | - Yury V Saltykov
- Laboratory for Fundamental and Applied Research, Saratov State University of Genetics, Biotechnology and Engineering named after N.I. Vavilov, Saratov, Russia
| | - Alexey D Zaberezhny
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, Moscow, Russia
| | - Olga S Larionova
- Laboratory for Fundamental and Applied Research, Saratov State University of Genetics, Biotechnology and Engineering named after N.I. Vavilov, Saratov, Russia
- Department for Microbiology and Biotechnology, Saratov State University of Genetics, Biotechnology and Engineering named after N.I. Vavilov, Saratov, Russia
| |
Collapse
|
15
|
Viera Herrera C, O'Connor PM, Ratrey P, Paul Ross R, Hill C, Hudson SP. Anionic liposome formulation for oral delivery of thuricin CD, a potential antimicrobial peptide therapeutic. Int J Pharm 2024; 654:123918. [PMID: 38401875 PMCID: PMC7615751 DOI: 10.1016/j.ijpharm.2024.123918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/30/2024] [Accepted: 02/16/2024] [Indexed: 02/26/2024]
Abstract
Thuricin CD is a two-peptide antimicrobial produced by Bacillus thuringiensis. Unlike previous antibiotics, it has shown narrow spectrum activity against Clostridioides difficile, a bacterium capable of causing infectious disease in the colon. However, peptide antibiotics have stability, solubility, and permeability problems that can affect their performance in vivo. This work focuses on the bioactivity and bioavailability of thuricin CD with a view to developing a formulation for delivery of active thuricin CD peptides through the gastrointestinal tract (GIT) for local delivery in the colon. The results indicate that thuricin CD is active at low concentrations only when both peptides are present. While thuricin CD was degraded by proteases and was unstable and poorly soluble in gastric fluid, it showed increased solubility in intestinal fluid, probably due to micelle encapsulation. Based on this, thuricin CD was encapsulated in anionic liposomes, which showed increased activity compared to the free peptide, maintained activity after exposure to pepsin in gastric fluid and intestinal fluid, was stable in suspension for over 21 days at room temperature and for 60 days at 4 °C, and exhibited no toxicity to epithelial intestinal cells. These findings suggest that an anionic lipid-based nano formulation may be a promising approach for local oral delivery of thuricin CD.
Collapse
Affiliation(s)
- Camila Viera Herrera
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland
| | - Paula M O'Connor
- Food Biosciences, Teagasc, Moorepark Food Research Centre, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland Cork, Cork, Ireland
| | - Poonam Ratrey
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland
| | - R Paul Ross
- APC Microbiome Ireland Cork, Cork, Ireland; School of Microbiology, University College Cork, College Road, Cork, Ireland
| | - Colin Hill
- APC Microbiome Ireland Cork, Cork, Ireland; School of Microbiology, University College Cork, College Road, Cork, Ireland
| | - Sarah P Hudson
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland.
| |
Collapse
|
16
|
Budvytyte L, Schroeder M, Graf E, Vaillant JJ. Rapid Progression of Invasive Listeria monocytogenes Infection in a Patient With Cirrhosis and Primary Sclerosing Cholangitis on Ustekinumab. Cureus 2024; 16:e58116. [PMID: 38738136 PMCID: PMC11088815 DOI: 10.7759/cureus.58116] [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] [Accepted: 03/31/2024] [Indexed: 05/14/2024] Open
Abstract
We present the case of a 62-year-old immunocompromised man with ulcerative colitis, primary sclerosing cholangitis, and cirrhosis treated with azathioprine and ustekinumab who quickly developed invasive Listeria monocytogenes infection after incidental identification on routine paracentesis. The infection rapidly progressed from bacterial peritonitis to bacteremia and meningitis within three days. Treatment with ampicillin and trimethoprim/sulfamethoxazole was successful. We highlight the increased risk of invasive listeriosis in immunocompromised individuals, including those on biologic therapies, and the importance of considering Listeria as a pathogen from sterile sites even in asymptomatic patients.
Collapse
Affiliation(s)
- Laura Budvytyte
- Department of Laboratory Medicine and Pathology, Mayo Clinic Alix School of Medicine, Scottsdale, USA
| | - Mariah Schroeder
- Department of Laboratory Medicine and Pathology, Mayo Clinic Alix School of Medicine, Scottsdale, USA
| | - Erin Graf
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, USA
| | - James J Vaillant
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, USA
| |
Collapse
|
17
|
Roodsant TJ, van der Ark KC, Schultsz C. Translocation across a human enteroid monolayer by zoonotic Streptococcus suis correlates with the presence of Gb3-positive cells. iScience 2024; 27:109178. [PMID: 38439959 PMCID: PMC10909756 DOI: 10.1016/j.isci.2024.109178] [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: 04/26/2023] [Revised: 11/14/2023] [Accepted: 02/06/2024] [Indexed: 03/06/2024] Open
Abstract
Streptococcus suis is a zoonotic pathogen that can cause meningitis and septicaemia. The consumption of undercooked pig products is an important risk factor for zoonotic infections, suggesting an oral route of infection. In a human enteroid model, we show that the zoonotic CC1 genotype has a 40% higher translocation frequency than the non-zoonotic CC16 genotype. Translocation occurred without increasing the permeability or disrupting the adherens junctions and tight junctions of the epithelial monolayer. The translocation of zoonotic S. suis was correlated with the presence of Gb3-positive cells, a human glycolipid receptor found on Paneth cells and targeted by multiple enteric pathogens. The virulence factors Streptococcal adhesin Protein and suilysin, known to interact with Gb3, were not essential for translocation in our epithelial model. Thus, the ability to translocate across an enteroid monolayer correlates with S. suis core genome composition and the presence of Gb3-positive cells in the intestinal epithelium.
Collapse
Affiliation(s)
- Thomas J. Roodsant
- Amsterdam UMC, Location University of Amsterdam, Department of Global Health, Amsterdam Institute for Global Health and Development, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Meibergdreef 9, Amsterdam, the Netherlands
| | - Kees C.H. van der Ark
- Amsterdam UMC, Location University of Amsterdam, Department of Global Health, Amsterdam Institute for Global Health and Development, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Meibergdreef 9, Amsterdam, the Netherlands
| | - Constance Schultsz
- Amsterdam UMC, Location University of Amsterdam, Department of Global Health, Amsterdam Institute for Global Health and Development, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Meibergdreef 9, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Tavares LS, Oliveira-Silva RL, Moura MT, da Silva JB, Benko-Iseppon AM, Lima-Filho JV. Reference genes for gene expression profiling in mouse models of Listeria monocytogenes infection. Biotechniques 2024; 76:104-113. [PMID: 38112054 DOI: 10.2144/btn-2023-0063] [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: 12/20/2023] Open
Abstract
RT-qPCR dissects transcription-based processes but requires reference genes (RGs) for data normalization. This study prospected RGs for mouse macrophages (pMØ) and spleen infected with Listeria monocytogenes. The pMØ were infected in vitro with L. monocytogenes or vehicle for 4 h. Mice were injected with L. monocytogenes (or vehicle) and euthanized 24 h post-injection. The RGs came from a multispecies primer set, from the literature or designed here. The RG ranking relied on GeNorm, NormFinder, BestKeeper, Delta-CT and RefFinder. B2m-H3f3a-Ppia were the most stable RGs for pMØ, albeit RG indexes fine-tuned estimations of cytokine relative expression. Actβ-Ubc-Ppia were the best RGs for spleen but modestly impacted the cytokine relative expression. Hence, mouse models of L. monocytogenes require context-specific RGs for RT-qPCR, thus reinforcing its paramount contribution to accurate gene expression profiling.
Collapse
Affiliation(s)
| | | | - Marcelo Tigre Moura
- Departamento de Biologia Celular e Molecular, Centro de Biotecnologia, Campus I, Universidade Federal da Paraíba, João Pessoa, PB, Brasil
| | | | | | - José Vitor Lima-Filho
- Departamento de Biologia, Universidade Federal Rural de Pernambuco, Recife, PE, Brasil
| |
Collapse
|
19
|
Kaavya R, Rajasekaran B, Shah K, Nickhil C, Palanisamy S, Palamae S, Chandra Khanashyam A, Pandiselvam R, Benjakul S, Thorakattu P, Ramesh B, Aurum FS, Babu KS, Rustagi S, Ramniwas S. Radical species generating technologies for decontamination of Listeria species in food: a recent review report. Crit Rev Food Sci Nutr 2024; 65:1974-1998. [PMID: 38380625 DOI: 10.1080/10408398.2024.2316295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Foodborne illnesses occur due to the contamination of fresh, frozen, or processed food products by some pathogens. Among several pathogens responsible for the illnesses, Listeria monocytogenes is one of the lethal bacteria that endangers public health. Several preexisting and novel technologies, especially non-thermal technologies are being studied for their antimicrobial effects, particularly toward L. monocytogenes. Some noteworthy emerging technologies include ultraviolet (UV) or light-emitting diode (LED), pulsed light, cold plasma, and ozonation. These technologies are gaining popularity since no heat is employed and undesirable deterioration of food quality, especially texture, and taste is devoided. This review aims to summarize the most recent advances in non-thermal processing technologies and their effect on inactivating L. monocytogenes in food products and on sanitizing packaging materials. These technologies use varying mechanisms, such as photoinactivation, photosensitization, disruption of bacterial membrane and cytoplasm, etc. This review can help food processing industries select the appropriate processing techniques for optimal benefits, in which the structural integrity of food can be preserved while simultaneously destroying L. monocytogenes present in foods. To eliminate Listeria spp., different technologies possess varying mechanisms such as rupturing the cell wall, formation of pyrimidine dimers in the DNA through photochemical effect, excitation of endogenous porphyrins by photosensitizers, generating reactive species, causing leakage of cellular contents and oxidizing proteins and lipids. These technologies provide an alternative to heat-based sterilization technologies and further development is still required to minimize the drawbacks associated with some technologies.
Collapse
Affiliation(s)
| | - Bharathipriya Rajasekaran
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | | | - C Nickhil
- Department of Food Engineering and Technology, Tezpur University, Assam, India
| | - Suguna Palanisamy
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Suriya Palamae
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | | | - R Pandiselvam
- Physiology, Biochemistry, and Post-Harvest Technology Division, ICAR - Central Plantation Crops Research Institute, Kasaragod, Kerala, India
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Priyamavada Thorakattu
- Department of Animal Sciences and Industry/Food Science Institute, Kansas State University, Manhattan, KS, USA
| | - Bharathi Ramesh
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE, USA
| | - Fawzan Sigma Aurum
- Research Center for Food Technology and Processing, National Research and Innovation Agency, Yogyakarta, Indonesia
| | | | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Seema Ramniwas
- University Centre for Research and Development, University of Biotechnology, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
20
|
Cong Z, Xiong Y, Lyu L, Fu B, Guo D, Sha Z, Yang B, Wu H. The relationship between Listeria infections and host immune responses: Listeriolysin O as a potential target. Biomed Pharmacother 2024; 171:116129. [PMID: 38194738 DOI: 10.1016/j.biopha.2024.116129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
Listeria monocytogenes (Lm), a foodborne bacterium, can infect people and has a high fatality rate in immunocompromised individuals. Listeriolysin O (LLO), the primary virulence factor of Lm, is critical in regulating the pathogenicity of Lm. This review concludes that LLO may either directly or indirectly activate a number of host cell viral pathophysiology processes, such as apoptosis, pyroptosis, autophagy, necrosis and necroptosis. We describe the invasion of host cells by Lm and the subsequent removal of Lm by CD8 T cells and CD4 T cells upon receipt of the LLO epitopes from major histocompatibility complex class I (MHC-I) and major histocompatibility complex class II (MHC-II). The development of several LLO-based vaccines that make use of the pore-forming capabilities of LLO and the immune response of the host cells is then described. Finally, we conclude by outlining the several natural substances that have been shown to alter the three-dimensional conformation of LLO by binding to particular amino acid residues of LLO, which reduces LLO pathogenicity and may be a possible pharmacological treatment for Lm.
Collapse
Affiliation(s)
- Zixuan Cong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Lyu Lyu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Beibei Fu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Zhou Sha
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Bo Yang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
21
|
Mohapatra RK, Mishra S, Tuglo LS, Sarangi AK, Kandi V, AL Ibrahim AA, Alsaif HA, Rabaan AA, Zahan MK. Recurring food source-based Listeria outbreaks in the United States: An unsolved puzzle of concern? Health Sci Rep 2024; 7:e1863. [PMID: 38317674 PMCID: PMC10839161 DOI: 10.1002/hsr2.1863] [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: 07/15/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024] Open
Abstract
Recurring Listeria outbreaks in the United States is a growing public healthcare concern. Although no associated reported death, 17 were hospitalized out of the 18 reported illnesses in the recent outbreak in 15 US states. The United States has experienced about 30 Listeria outbreaks in the last decade with 524 Listeriosis cases and 80 deaths. The identified origin were ice cream, leafy greens, mushroom, meat slice, dairy products like cheese, packaged salads, cooked chicken, hard-boiled egg, pork product, frozen vegetable, raw milk, packaged caramel apple, bean sprout and soya products. Although rare, Listeria may lead to serious illness (invasive listeriosis) or death. Listeriosis is critically harmful and medically complicated, especially in the pregnant, the old above 65 years and in the immunocompromised. It could cause premature birth, miscarriage or even neonatal death. Hospitalization is often necessary in the geriatric, being fatal at times. Among Listeria sp., Listeria monocytogenes is often human infection-associated. It is a gram-positive, non-sporulating, motile bacillus opportunistic pathogen. Food-borne listeriosis is often associated with frozen foods due to its ability to thrive at low temperatures. Hypervirulent strains of L. monocytogenes with an ability to infect the respiratory system (the lungs) was recently reported in the coronavirus disease-19 patients during the pandemic. L. monocytogenes seemed to have developed antimicrobial resistance to ciprofloxacin and meropenem, possibly acquired through the food chain. An early onset of listeriosis in the newborn is evident in the first 7 days postparturition. As the bacteria colonize the genitourinary tract, majority of such cases result from teratogenic transfer during vaginal delivery. Premature newborns, neonates born outside healthcare facilities and low-birth-weight babies were increasingly predisposed to an early onset of listeriosis. Listeria outbreaks were earlier reported in South Africa, Australia and Europe, with an unclear origin of the outbreaks. Social media updates about such outbreaks, the most likely food source, and measures to self-protect are suggested as preventive measures. The article deals on various such aspects related to listeriosis primarily originating from food, to ensure better public healthcare and human wellness.
Collapse
Affiliation(s)
| | - Snehasish Mishra
- School of BiotechnologyKIIT Deemed UniversityBhubaneswarOdishaIndia
| | - Lawrence Sena Tuglo
- Department of Nutrition and Dietetics, School of Allied Health SciencesUniversity of Health and Allied SciencesHoGhana
| | - Ashish K. Sarangi
- Department of Chemistry, School of Applied SciencesCenturion University of Technology and ManagementBalangirOdishaIndia
| | - Venkataramana Kandi
- Department of MicrobiologyPrathima Institute of Medical SciencesKarimnagarTelanganaIndia
| | | | | | - Ali A. Rabaan
- Molecular Diagnostic LaboratoryJohns Hopkins Aramco HealthcareDhahranSaudi Arabia
- College of MedicineAlfaisal UniversityRiyadhSaudi Arabia
- Department of Public Health and NutritionThe University of HaripurHaripurPakistan
| | | |
Collapse
|
22
|
Richardson IM, Calo CJ, Ginter EL, Niehaus E, Pacheco KA, Hind LE. Diverse bacteria elicit distinct neutrophil responses in a physiologically relevant model of infection. iScience 2024; 27:108627. [PMID: 38188520 PMCID: PMC10770534 DOI: 10.1016/j.isci.2023.108627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/24/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
An efficient neutrophil response is critical for fighting bacterial infections, which remain a significant global health concern; therefore, modulating neutrophil function could be an effective therapeutic approach. While we have a general understanding of how neutrophils respond to bacteria, how neutrophil function differs in response to diverse bacterial infections remains unclear. Here, we use a microfluidic infection-on-a-chip device to investigate the neutrophil response to four bacterial species: Pseudomonas aeruginosa, Salmonella enterica, Listeria monocytogenes, and Staphylococcus aureus. We find enhanced neutrophil extravasation to L. monocytogenes, a limited overall response to S. aureus, and identify IL-6 as universally important for neutrophil extravasation. Furthermore, we demonstrate a higher percentage of neutrophils generate reactive oxygen species (ROS) when combating gram-negative bacteria versus gram-positive bacteria. For all bacterial species, we found the percentage of neutrophils producing ROS increased following extravasation through an endothelium, underscoring the importance of studying neutrophil function in physiologically relevant models.
Collapse
Affiliation(s)
- Isaac M. Richardson
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| | - Christopher J. Calo
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| | - Eric L. Ginter
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| | - Elise Niehaus
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| | - Kayla A. Pacheco
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| | - Laurel E. Hind
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303, USA
| |
Collapse
|
23
|
Deng C, Chiu KHY, Lou N, Xing F. Case report: Occult Listeria monocytogenes invasion leading to prosthetic hip joint infection in a patient with rheumatoid arthritis taking tofacitinib. Front Med (Lausanne) 2024; 10:1322993. [PMID: 38264038 PMCID: PMC10803402 DOI: 10.3389/fmed.2023.1322993] [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: 10/17/2023] [Accepted: 12/28/2023] [Indexed: 01/25/2024] Open
Abstract
It has been suggested that targeted therapy may potentially increase the risk of listeriosis. However, no reported cases of Listeria monocytogenes prosthetic joint infection have been documented during Janus Kinase (JAK) pathway inhibitor use. Herein, we present a 70-year-old female with rheumatoid arthritis who had undergone bilateral hip joint replacement and subsequently developed Listeria monocytogenes prosthetic joint infection following tofacitinib therapy. We suggest that the use of tofacitinib may potentially heighten susceptibility to listeriosis in patients afflicted with rheumatoid arthritis.
Collapse
Affiliation(s)
- Chaowen Deng
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| | | | - Nan Lou
- Department of Orthopedics, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Fanfan Xing
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
24
|
Meireles D, Pombinho R, Cabanes D. Signals behind Listeria monocytogenes virulence mechanisms. Gut Microbes 2024; 16:2369564. [PMID: 38979800 PMCID: PMC11236296 DOI: 10.1080/19490976.2024.2369564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/13/2024] [Indexed: 07/10/2024] Open
Abstract
The tight and coordinated regulation of virulence gene expression is crucial to ensure the survival and persistence of bacterial pathogens in different contexts within their hosts. Considering this, bacteria do not express virulence factors homogenously in time and space, either due to their associated fitness cost or to their detrimental effect at specific infection stages. To efficiently infect and persist into their hosts, bacteria have thus to monitor environmental cues or chemical cell-to-cell signaling mechanisms that allow their transition from the external environment to the host, and therefore adjust gene expression levels, intrinsic biological activities, and appropriate behaviors. Listeria monocytogenes (Lm), a major Gram-positive facultative intracellular pathogen, stands out for its adaptability and capacity to thrive in a wide range of environments. Because of that, Lm presents itself as a significant concern in food safety and public health, that can lead to potentially life-threatening infections in humans. A deeper understanding of the intricate bacterial virulence mechanisms and the signals that control them provide valuable insights into the dynamic interplay between Lm and the host. Therefore, this review addresses the role of some crucial signals behind Lm pathogenic virulence mechanisms and explores how the ability to assimilate and interpret these signals is fundamental for pathogenesis, identifying potential targets for innovative antimicrobial strategies.
Collapse
Affiliation(s)
- Diana Meireles
- Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Group of Molecular Microbiology, IBMC, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar – ICBAS, Porto, Portugal
| | - Rita Pombinho
- Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Group of Molecular Microbiology, IBMC, Porto, Portugal
| | - Didier Cabanes
- Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Group of Molecular Microbiology, IBMC, Porto, Portugal
| |
Collapse
|
25
|
Wu J, McAuliffe O, O'Byrne CP. A novel RofA-family transcriptional regulator, GadR, controls the development of acid resistance in Listeria monocytogenes. mBio 2023; 14:e0171623. [PMID: 37882515 PMCID: PMC10746197 DOI: 10.1128/mbio.01716-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
IMPORTANCE The ability to survive the acidic conditions found in the stomach is crucial for the food-borne pathogen Listeria monocytogenes to gain access to the mammalian gastrointestinal tract. Little is currently known about how acid resistance is regulated in this pathogen and why this trait is highly variable between strains. Here, we used comparative genomics to identify a novel RofA-family transcriptional regulator, GadR, that controls the development of acid resistance. The RofA family of regulators was previously found only in a small group of bacterial pathogens, including streptococci, where they regulate virulence properties. We show that gadR encodes the dominant regulator of acid resistance in L. monocytogenes and that its sequence variability accounts for previously observed differences between strains in this trait. Together, these findings significantly advance our understanding of how this important pathogen copes with acid stress and suggest a potential molecular target to aid its control in the food chain.
Collapse
Affiliation(s)
- Jialun Wu
- Bacterial Stress Response Group, Microbiology, Ryan Institute, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Olivia McAuliffe
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
- School of Biological Sciences, Queen’s University Belfast, Belfast, Northern Ireland
| | - Conor P. O'Byrne
- Bacterial Stress Response Group, Microbiology, Ryan Institute, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| |
Collapse
|
26
|
Ramos GLPA, Duarte MCKH, Nascimento JS, Cruz AG, Sant'Ana AS. Quantitative microbiological risk assessment for the occurrence of listeriosis in Brazil due to the consumption of milk processed by pasteurization or thermosonication. Int J Food Microbiol 2023; 407:110424. [PMID: 37806011 DOI: 10.1016/j.ijfoodmicro.2023.110424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 08/02/2023] [Accepted: 10/01/2023] [Indexed: 10/10/2023]
Abstract
This study aimed to estimate the risk of listeriosis from the consumption of pasteurized milk in Brazil, comparing conventional treatment with the technology of thermosonication. The Quantitative Microbiological Risk Assessment (QMRA) model was developed, covering the entire milk production chain, from milking to the moment of consumption. In general, higher risks were observed in association with higher initial concentrations of the pathogen and the vulnerable population. The highest risk predicted (3.67 × 10-5) was related to the scenario considering the initial concentration range of L. monocytogenes between 4 and 6 log CFU/mL, with conventional treatment and considering the vulnerable population, resulting in one case of listeriosis every 27,248 servings. When considering thermosonication treatment, lower risks have been predicted. The scenario analysis indicated that the steps related to storage conditions in retail and at the consumer's home (post-processing steps) are the most influential in the associated risk, in all scenarios. The predictive parameters of inactivation related to the applied treatment also have a considerable influence on the risk. The results point to the influence of the stages of the dairy production chain and the thermosonication treatment applied in the food safety of milk, subsidizing information for industrial application and for regulatory agencies.
Collapse
Affiliation(s)
- Gustavo Luis P A Ramos
- Fluminense Federal University (UFF), Faculty of Veterinary Medicine, Niterói, RJ, Brazil; Federal Institute of Education, Science, and Technology of Rio de Janeiro (IFRJ), Food Department, Rio de Janeiro, RJ, Brazil.
| | | | - Janaína S Nascimento
- Federal Institute of Education, Science, and Technology of Rio de Janeiro (IFRJ), Food Department, Rio de Janeiro, RJ, Brazil
| | - Adriano G Cruz
- Federal Institute of Education, Science, and Technology of Rio de Janeiro (IFRJ), Food Department, Rio de Janeiro, RJ, Brazil
| | - Anderson S Sant'Ana
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
27
|
Raman V, Deshpande CP, Khanduja S, Howell LM, Van Dessel N, Forbes NS. Build-a-bug workshop: Using microbial-host interactions and synthetic biology tools to create cancer therapies. Cell Host Microbe 2023; 31:1574-1592. [PMID: 37827116 DOI: 10.1016/j.chom.2023.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
Many systemically administered cancer therapies exhibit dose-limiting toxicities that reduce their effectiveness. To increase efficacy, bacterial delivery platforms have been developed that improve safety and prolong treatment. Bacteria are a unique class of therapy that selectively colonizes most solid tumors. As delivery vehicles, bacteria have been genetically modified to express a range of therapies that match multiple cancer indications. In this review, we describe a modular "build-a-bug" method that focuses on five design characteristics: bacterial strain (chassis), therapeutic compound, delivery method, immune-modulating features, and genetic control circuits. We emphasize how fundamental research into gut microbe pathogenesis has created safe bacterial therapies, some of which have entered clinical trials. The genomes of gut microbes are fertile grounds for discovery of components to improve delivery and modulate host immune responses. Future work coupling these delivery vehicles with insights from gut microbes could lead to the next generation of microbial cancer therapy.
Collapse
Affiliation(s)
- Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA; Ernest Pharmaceuticals, LLC, Hadley, MA, USA
| | - Chinmay P Deshpande
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Shradha Khanduja
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Lars M Howell
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | | | - Neil S Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA; Molecular and Cell Biology Program, University of Massachusetts, Amherst, Amherst, MA, USA; Institute for Applied Life Science, University of Massachusetts, Amherst, Amherst, MA, USA.
| |
Collapse
|
28
|
de Oliveira LF, Filho DM, Marques BL, Maciel GF, Parreira RC, do Carmo Neto JR, Da Silva PEF, Guerra RO, da Silva MV, Santiago HDC, Birbrair A, Kihara AH, Dias da Silva VJ, Glaser T, Resende RR, Ulrich H. Organoids as a novel tool in modelling infectious diseases. Semin Cell Dev Biol 2023; 144:87-96. [PMID: 36182613 DOI: 10.1016/j.semcdb.2022.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/04/2022] [Indexed: 11/23/2022]
Abstract
Infectious diseases worldwide affect human health and have important societal impacts. A better understanding of infectious diseases is urgently needed. In vitro and in vivo infection models have brought notable contributions to the current knowledge of these diseases. Organoids are multicellular culture systems resembling tissue architecture and function, recapitulating many characteristics of human disease and elucidating mechanisms of host-infectious agent interactions in the respiratory and gastrointestinal systems, the central nervous system and the skin. Here, we discuss the applicability of the organoid technology for modeling pathogenesis, host response and features, which can be explored for the development of preventive and therapeutic treatments.
Collapse
Affiliation(s)
- Lucas Felipe de Oliveira
- Departamento de Fisiologia, Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil; Instituto Nacional de Ciência e Tecnologia de Medicina Regenerativa, Rio de Janeiro, RJ, Brazil
| | - Daniel Mendes Filho
- Departamento de Fisiologia, Escola Médica de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Bruno Lemes Marques
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal deGoiás, Goiânia, GO, Brazil
| | | | | | - José Rodrigues do Carmo Neto
- Departamento de Biociência e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | - Rhanoica Oliveira Guerra
- Departamento de Microbiologia, Imunologia eParasitologia, Instituto de Ciências Naturais e Biológicas, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Marcos Vinicius da Silva
- Departamento de Microbiologia, Imunologia eParasitologia, Instituto de Ciências Naturais e Biológicas, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Helton da Costa Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Radiology, Columbia University Medical Center, New York, NY, USA; Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Belo Horizonte, MG, Brazil
| | - Alexandre H Kihara
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Valdo José Dias da Silva
- Departamento de Fisiologia, Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil; Instituto Nacional de Ciência e Tecnologia de Medicina Regenerativa, Rio de Janeiro, RJ, Brazil
| | - Talita Glaser
- Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Belo Horizonte, MG, Brazil
| | - Henning Ulrich
- Instituto Nacional de Ciência e Tecnologia de Medicina Regenerativa, Rio de Janeiro, RJ, Brazil; Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil.
| |
Collapse
|
29
|
Nandi I, Aroeti B. Mitogen-Activated Protein Kinases (MAPKs) and Enteric Bacterial Pathogens: A Complex Interplay. Int J Mol Sci 2023; 24:11905. [PMID: 37569283 PMCID: PMC10419152 DOI: 10.3390/ijms241511905] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 08/13/2023] Open
Abstract
Diverse extracellular and intracellular cues activate mammalian mitogen-activated protein kinases (MAPKs). Canonically, the activation starts at cell surface receptors and continues via intracellular MAPK components, acting in the host cell nucleus as activators of transcriptional programs to regulate various cellular activities, including proinflammatory responses against bacterial pathogens. For instance, binding host pattern recognition receptors (PRRs) on the surface of intestinal epithelial cells to bacterial pathogen external components trigger the MAPK/NF-κB signaling cascade, eliciting cytokine production. This results in an innate immune response that can eliminate the bacterial pathogen. However, enteric bacterial pathogens evolved sophisticated mechanisms that interfere with such a response by delivering virulent proteins, termed effectors, and toxins into the host cells. These proteins act in numerous ways to inactivate or activate critical components of the MAPK signaling cascades and innate immunity. The consequence of such activities could lead to successful bacterial colonization, dissemination, and pathogenicity. This article will review enteric bacterial pathogens' strategies to modulate MAPKs and host responses. It will also discuss findings attempting to develop anti-microbial treatments by targeting MAPKs.
Collapse
Affiliation(s)
| | - Benjamin Aroeti
- Department of Biological Chemistry, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190410, Israel;
| |
Collapse
|
30
|
OuYang X, Liu P, Zheng Y, Jiang H, Lv Q, Huang W, Hao H, Pian Y, Kong D, Jiang Y. TRIM32 reduced the recruitment of innate immune cells and the killing capacity of Listeria monocytogenes by inhibiting secretion of chemokines. Gut Pathog 2023; 15:32. [PMID: 37415157 DOI: 10.1186/s13099-023-00558-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/10/2023] [Indexed: 07/08/2023] Open
Abstract
Listeria monocytogenes (Lm) is a facultative, intracellular Gram-positive pathogenic bacterium that causes sepsis, a condition characterized by persistent excessive inflammation and organ dysfunction. However, the pathogenesis of Lm-induced sepsis is unknown. In this research, we discovered that TRIM32 is required for innate immune regulation during Lm infection. Trim32 deficiency remarkably reduced bacteremia and proinflammatory cytokine secretion in mice with severe Lm infection, preventing sepsis. Trim32-/- mice had a lower bacterial burden after Lm infection and survived significantly longer than wild-type (WT) mice, as well as lower serum levels of inflammatory cytokines TNF-α, IL-6, IL-18, IL-12p70, IFN-β, and IFN-γ at 1 day post infection (dpi) compared to WT mice. On the other hand, the chemokines CXCL1, CCL2, CCL7, and CCL5 were enhanced at 3 dpi in Trim32-/- mice than WT mice, reflecting increased recruitment of neutrophils and macrophages. Furthermore, Trim32-/- mice had higher levels of macrophage-associated iNOS to kill Lm. Collectively, our findings suggest that TRIM32 reduces innate immune cells recruitment and Lm killing capabilities via iNOS production.
Collapse
Affiliation(s)
- Xuan OuYang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Peng Liu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Yuling Zheng
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Hua Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Qingyu Lv
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Wenhua Huang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Huaijie Hao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | - Yaya Pian
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China.
| | - Decong Kong
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China.
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China.
| |
Collapse
|
31
|
Lin Y, Lu J, Yang Z, Wang T, Li H, Sha S, Liu Z, Zhao Y, Wang L. Comparative genomics reveals key molecular targets for mutant Pediococcus pentosaceus C23221 producing pediocin. Int J Biol Macromol 2023:125006. [PMID: 37224904 DOI: 10.1016/j.ijbiomac.2023.125006] [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: 04/23/2023] [Accepted: 05/19/2023] [Indexed: 05/26/2023]
Abstract
Listeria monocytogenes is a common microorganism that causes food spoilage. Pediocins are some biologically active peptides or proteins encoded by ribosomes, which have a strong antimicrobial activity against L. monocytogenes. In this study, the antimicrobial activity of previously isolated P. pentosaceus C-2-1 was enhanced by ultraviolet (UV) mutagenesis. A positive mutant strain P. pentosaceus C23221 was obtained after 8 rounds of UV irradiation with increased antimicrobial activity of 1448 IU/mL, which was 8.47 folds higher than that of wild-type C-2-1. The genome of strain C23221 and wild-type C-2-1 was compared with identify the key genes for higher activity. The genome of the mutant strain C23221 consists of a chromosome of 1,742,268 bp, with 2052 protein-coding genes, 4 rRNA operons, and 47 tRNA genes, which is 79,769 bp less than the original strain. Compared with strain C-2-1, a total of 19 deduced proteins involved in 47 genes are unique to C23221 analyzed by GO database; the specific ped gene related to bacteriocin biosynthesis were detected using antiSMASH in mutant C23221, indicating mutant C23221 produced a new bacteriocin under mutagenesis conditions. This study provides genetic basis for further constituting a rational strategy to genetically engineer wild-type C-2-1 into an overproducer.
Collapse
Affiliation(s)
- Yi Lin
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Jiawen Lu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Zilu Yang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Tianming Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Hongbiao Li
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Shenfei Sha
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Zhenmin Liu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai 200436, China
| | - Yueliang Zhao
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation, Shanghai Ocean University, Shanghai 201306, China
| | - Liping Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
32
|
Scarponi D, ranalli M, Bedetti L, Miselli F, Rossi K, Iughetti L, Lugli L, Berardi A. La listeriosi neonatale e materno-fetale. MEDICO E BAMBINO 2023; 42:177-182. [DOI: 10.53126/meb42177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2023]
Abstract
Listeriosis is a foodborne condition associated with the ingestion of food contaminated by Listeria monocytogenes that can cause serious health consequences in neonates and pregnant women. In these groups the incidence of the disease is higher due to the relative immunocompromised state and the particular Listeria monocytogenes tropism for the placenta. The paediatrician and the neonatologist must therefore be able to recognize the signs and symptoms of the disease promptly so as to set up an adequate treatment according to the most recent international guidelines. This article provides epidemiological data and describes the pathophysiology and clinical manifestations of maternal-foetal and neonatal listeriosis, as well as the therapeutic indications according to very recent recommendations.
Collapse
Affiliation(s)
- Davide Scarponi
- Scuola di Specializzazione in Pediatria, AOU Policlinico di Modena
| | - Marco ranalli
- Scuola di Specializzazione in Pediatria, AOU Policlinico di Modena
| | - Luca Bedetti
- Terapia Intensiva Neonatale, AOU Policlinico di Modena
| | | | - katia Rossi
- Terapia Intensiva Neonatale, AOU Policlinico di Modena
| | | | - Licia Lugli
- Terapia Intensiva Neonatale, AOU Policlinico di Modena
| | | |
Collapse
|
33
|
Zakrzewski AJ, Kurpas M, Zadernowska A, Chajęcka-Wierzchowska W, Fraqueza MJ. A Comprehensive Virulence and Resistance Characteristics of Listeria monocytogenes Isolated from Fish and the Fish Industry Environment. Int J Mol Sci 2023; 24:ijms24043581. [PMID: 36834997 PMCID: PMC9967382 DOI: 10.3390/ijms24043581] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
Listeria monocytogenes is an important pathogen, often associated with fish, that can adapt and survive in products and food processing plants, where it can persist for many years. It is a species characterized by diverse genotypic and phenotypic characteristics. Therefore, in this study, a total of 17 L. monocytogenes strains from fish and fish-processing environments in Poland were characterized for their relatedness, virulence profiles, and resistance genes. The Core Genome Multilocus Sequence Typing (cgMLST) analysis revealed that the most frequent serogroups were IIa and IIb; sequence types (ST) were ST6 and ST121; and clonal complexes (CC) were CC6 and CC121. Core genome multilocus sequence typing (cgMLST) analysis was applied to compare the present isolates with the publicly available genomes of L. monocytogenes strains recovered in Europe from humans with listeriosis. Despite differential genotypic subtypes, most strains had similar antimicrobial resistance profiles; however, some of genes were located on mobile genetic elements that could be transferred to commensal or pathogenic bacteria. The results of this study showed that molecular clones of tested strains were characteristic for L. monocytogenes isolated from similar sources. Nevertheless, it is worth emphasizing that they could present a major public health risk due to their close relation with strains isolated from human listeriosis.
Collapse
Affiliation(s)
| | - Monika Kurpas
- Department of Immunobiology and Environmental Microbiology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Anna Zadernowska
- Department of Industrial and Food Microbiology, University of Warmia and Mazrui, 10-726 Olsztyn, Poland
- Correspondence:
| | | | - Maria João Fraqueza
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| |
Collapse
|
34
|
Martín I, Córdoba JJ, Rodríguez A. Effect of acidic conditions on the growth and expression of two virulence genes of Listeria monocytogenes serotype 4b. Res Microbiol 2023; 174:104042. [PMID: 36740027 DOI: 10.1016/j.resmic.2023.104042] [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: 06/21/2022] [Revised: 01/10/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
In this work, the effect of the usual acidic conditions of dry-cured fermented foods (pH values between 4.5 and 6), on the growth and expression of the virulence genes, hly and inlA, of Listeria monocytogenes serotype 4b, was evaluated. To analyse the expression of the inlA gene, a novel real-time PCR (qPCR) method using SYBR® Green methodology was developed. L. monocytogenes levels increased as the pH did and they were kept constant throughout incubation time at pH 4.5. However, a significant increase in the relative expression of the virulence genes was detected in most of the acidic conditions in all the incubation times. The most pronounced upregulation of the relative expression of the virulence genes was found at pH 4.5. The efficient inlA-based qPCR method could be of interest to check changes in the expression of such virulence gene of this pathogenic bacterium in acidic environments.
Collapse
Affiliation(s)
- Irene Martín
- Higiene y Seguridad Alimentaria, Instituto Universitario de Investigación de Carne y Productos Cárnicos, Facultad de Veterinaria, Universidad de Extremadura, Avda. de las Ciencias, s/n. 10003-Cáceres, Spain
| | - Juan J Córdoba
- Higiene y Seguridad Alimentaria, Instituto Universitario de Investigación de Carne y Productos Cárnicos, Facultad de Veterinaria, Universidad de Extremadura, Avda. de las Ciencias, s/n. 10003-Cáceres, Spain
| | - Alicia Rodríguez
- Higiene y Seguridad Alimentaria, Instituto Universitario de Investigación de Carne y Productos Cárnicos, Facultad de Veterinaria, Universidad de Extremadura, Avda. de las Ciencias, s/n. 10003-Cáceres, Spain.
| |
Collapse
|
35
|
Tran BM, Linnik DS, Punter CM, Śmigiel WM, Mantovanelli L, Iyer A, O’Byrne C, Abee T, Johansson J, Poolman B. Super-resolving microscopy reveals the localizations and movement dynamics of stressosome proteins in Listeria monocytogenes. Commun Biol 2023; 6:51. [PMID: 36641529 PMCID: PMC9840623 DOI: 10.1038/s42003-023-04423-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
The human pathogen Listeria monocytogenes can cope with severe environmental challenges, for which the high molecular weight stressosome complex acts as the sensing hub in a complicated signal transduction pathway. Here, we show the dynamics and functional roles of the stressosome protein RsbR1 and its paralogue, the blue-light receptor RsbL, using photo-activated localization microscopy combined with single-particle tracking and single-molecule displacement mapping and supported by physiological studies. In live cells, RsbR1 is present in multiple states: in protomers with RsbS, large clusters of stressosome complexes, and in connection with the plasma membrane via Prli42. RsbL diffuses freely in the cytoplasm but forms clusters upon exposure to light. The clustering of RsbL is independent of the presence of Prli42. Our work provides a comprehensive view of the spatial organization and intracellular dynamics of the stressosome proteins in L. monocytogenes, which paves the way towards uncovering the stress-sensing mechanism of this signal transduction pathway.
Collapse
Affiliation(s)
- Buu Minh Tran
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Dmitrii Sergeevich Linnik
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Christiaan Michiel Punter
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Wojciech Mikołaj Śmigiel
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Luca Mantovanelli
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Aditya Iyer
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| | - Conor O’Byrne
- Microbiology, School of Biological & Chemical Sciences, Ryan Institute, University of Galway, Galway, Ireland
| | - Tjakko Abee
- grid.4818.50000 0001 0791 5666Laboratory of Food Microbiology, Wageningen University & Research, Wageningen, the Netherlands
| | - Jörgen Johansson
- grid.12650.300000 0001 1034 3451Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Bert Poolman
- grid.4830.f0000 0004 0407 1981Department of Biochemistry, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
36
|
Wortel IMN, Kim S, Liu AY, Ibarra EC, Miller MJ. Listeria motility increases the efficiency of epithelial invasion during intestinal infection. PLoS Pathog 2022; 18:e1011028. [PMID: 36584235 PMCID: PMC9836302 DOI: 10.1371/journal.ppat.1011028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 01/12/2023] [Accepted: 11/28/2022] [Indexed: 12/31/2022] Open
Abstract
Listeria monocytogenes (Lm) is a food-borne pathogen that causes severe bacterial gastroenteritis, with high rates of hospitalization and mortality. Lm is ubiquitous in soil, water and livestock, and can survive and proliferate at low temperatures. Following oral ingestion of contaminated food, Lm crosses the epithelium through intestinal goblet cells in a mechanism mediated by Lm InlA binding host E-cadherin. Importantly, human infections typically occur with Lm growing at or below room temperature, which is flagellated and motile. Even though many important human bacterial pathogens are flagellated, little is known regarding the effect of Lm motility on invasion and immune evasion. Here, we used complementary imaging and computer modeling approaches to test the hypothesis that bacterial motility helps Lm locate and engage target cells permissive for invasion. Imaging explanted mouse and human intestine, we showed that Lm grown at room temperature uses motility to scan the epithelial surface and preferentially attach to target cells. Furthermore, we integrated quantitative parameters from our imaging experiments to construct a versatile "layered" cellular Potts model (L-CPM) that simulates host-pathogen dynamics. Simulated data are consistent with the hypothesis that bacterial motility enhances invasion by allowing bacteria to search the epithelial surface for their preferred invasion targets. Indeed, our model consistently predicts that motile bacteria invade twice as efficiently over the first hour of infection. We also examined how bacterial motility affected interactions with host cellular immunity. In a mouse model of persistent infection, we found that neutrophils migrated to the apical surface of the epithelium 5 hours post infection and interacted with Lm. Yet in contrast to the view that neutrophils "hunt" for bacteria, we found that these interactions were driven by motility of Lm-which moved at least ~50x faster than neutrophils. Furthermore, our L-CPM predicts that motile bacteria maintain their invasion advantage even in the presence of host phagocytes, with the balance between invasion and phagocytosis governed almost entirely by bacterial motility. In conclusion, our simulations provide insight into host pathogen interaction dynamics at the intestinal epithelial barrier early during infection.
Collapse
Affiliation(s)
- Inge M. N. Wortel
- Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, the Netherlands
| | - Seonyoung Kim
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Annie Y. Liu
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Enid C. Ibarra
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Mark J. Miller
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
37
|
Grima P, Urciuoli C, Simone G, Palazzo AG, Nuzzo M, Quarta M, Carraturo I, Maci AM, Marinaci S, Portaccio G, Guido M, Zizza A, Romano A. Fatal Listeria monocytogenes septicemia and meningitis complicated by Candida glabrata fungemia: a case report. Curr Med Res Opin 2022; 38:2119-2121. [PMID: 36053118 DOI: 10.1080/03007995.2022.2120689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Listeria monocytogenes is a Gram-positive bacteria and etiological agent of listeriosis. It has the ability to colonize the intestinal lumen and cross the intestinal, blood-brain, and placental barriers, leading to invasive listeriosis responsible for septicemia and meningitis in subjects at risk such as patients with diabetes mellitus, the elderly, and immunocompromised individuals and, for maternal-neonatal infection in pregnant women. We report a rare case of L. monocytogenes septicemia and meningitis complicated by Candida glabrata fungemia on a patient with a history of type 2 diabetes mellitus, hypothyroidism, hypertension, chronic kidney failure, chronic ischemic vascular encephalopathy, and atrial fibrillation. Although adequate therapy was rapidly started with an initial partial clinical improvement, the patient suddenly experienced clinical worsening concomitantly with Candida septicemia resulting in a fatal outcome. To our knowledge, this is the first described case of an invasive L. monocytogenes infection complicated by Candida sepsis. We hypothesize that concomitant Candida infection may play a significant role in the pathogenesis and virulence of L. monocytogenes.
Collapse
Affiliation(s)
| | - Caterina Urciuoli
- Operative Unit of Infectious Diseases, "V. Fazzi" Hospital, Lecce, Italy
| | - Giuseppe Simone
- Operative Unit of Infectious Diseases, "V. Fazzi" Hospital, Lecce, Italy
| | | | - Milva Nuzzo
- Operative Unit of Infectious Diseases, "V. Fazzi" Hospital, Lecce, Italy
| | - Maurizio Quarta
- Operative Unit of Infectious Diseases, "V. Fazzi" Hospital, Lecce, Italy
| | | | - Anna Maria Maci
- Operative Unit of Infectious Diseases, "V. Fazzi" Hospital, Lecce, Italy
| | - Salvatore Marinaci
- Operative Unit of Infectious Diseases, "V. Fazzi" Hospital, Lecce, Italy
| | - Gerolamo Portaccio
- Operative Unit of Infectious Diseases, "V. Fazzi" Hospital, Lecce, Italy
| | - Marcello Guido
- Laboratory of Hygiene, Department of Biological and Environmental Sciences and Technologies, Faculty of Sciences, University of Salento, Lecce, Italy
| | - Antonella Zizza
- Institute of Clinical Physiology, National Research Council, Lecce, Italy
| | - Anacleto Romano
- Operative Unit of Infectious Diseases, "V. Fazzi" Hospital, Lecce, Italy
| |
Collapse
|
38
|
Li X, Shi X, Song Y, Yao S, Li K, Shi B, Sun J, Liu Z, Zhao W, Zhao C, Wang J. Genetic diversity, antibiotic resistance, and virulence profiles of Listeria monocytogenes from retail meat and meat processing. Food Res Int 2022; 162:112040. [DOI: 10.1016/j.foodres.2022.112040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/30/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
|
39
|
Fredriksson-Ahomaa M, Sauvala M, Kurittu P, Heljanko V, Heikinheimo A, Paulsen P. Characterisation of Listeria monocytogenes Isolates from Hunted Game and Game Meat from Finland. Foods 2022; 11:foods11223679. [PMID: 36429271 PMCID: PMC9689155 DOI: 10.3390/foods11223679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Listeria monocytogenes is an important foodborne zoonotic bacterium. It is a heterogeneous species that can be classified into lineages, serogroups, clonal complexes, and sequence types. Only scarce information exists on the properties of L. monocytogenes from game and game meat. We characterised 75 L. monocytogenes isolates from various game sources found in Finland between 2012 and 2020. The genetic diversity, presence of virulence and antimicrobial genes were studied with whole genome sequencing. Most (89%) of the isolates belonged to phylogenetic lineage (Lin) II and serogroup (SG) IIa. SGs IVb (8%) and IIb (3%) of Lin I were sporadically identified. In total, 18 clonal complexes and 21 sequence types (STs) were obtained. The most frequent STs were ST451 (21%), ST585 (12%) and ST37 (11%) found in different sample types between 2012 and 2020. We observed 10 clusters, formed by closely related isolates with 0-10 allelic differences. Most (79%) of the virulence genes were found in all of the L. monocytogenes isolates. Only fosX and lin were found out of 46 antimicrobial resistance genes. Our results demonstrate that potentially virulent and antimicrobial-sensitive L. monocytogenes isolates associated with human listeriosis are commonly found in hunted game and game meat in Finland.
Collapse
Affiliation(s)
- Maria Fredriksson-Ahomaa
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
- Correspondence:
| | - Mikaela Sauvala
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Paula Kurittu
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Viivi Heljanko
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Annamari Heikinheimo
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland
- Microbiology Unit, Finnish Food Authority, 60100 Seinäjoki, Finland
| | - Peter Paulsen
- Unit of Food Hygiene and Technology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, 1210 Vienna, Austria
| |
Collapse
|
40
|
Magagna G, Finazzi G, Filipello V. Newly Designed Primers for the Sequencing of the inlA Gene of Lineage I and II Listeria monocytogenes Isolates. Int J Mol Sci 2022; 23:ijms232214106. [PMID: 36430584 PMCID: PMC9698914 DOI: 10.3390/ijms232214106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022] Open
Abstract
Listeria monocytogenes is a major human foodborne pathogen responsible for listeriosis. The virulence factor Internalin A (inlA) has a key role in the invasion of L. monocytogenes into the human intestinal epithelium, and the presence of premature stop-codons (PMSC) mutations in the inlA gene sequence is correlated with attenuated virulence. The inlA sequencing process is carried out by dividing the gene into three sections which are then reassembled to obtain the full gene. The primers available however were only able to entirely amplify the lineage II isolates. In this study, we present a set of new primers which allow inlA sequencing of isolates belonging to both lineages, since lineage I isolates are the ones most frequently associated to clinical cases. Using newly designed primers, we assessed the presence of inlA PMSCs in food, food processing environments and clinical isolates.
Collapse
Affiliation(s)
- Giulia Magagna
- Food Safety Department, Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna (IZSLER), Via A. Bianchi 9, 25124 Brescia, Italy
- Correspondence: ; Tel.: +39-0302-2906-11
| | - Guido Finazzi
- Food Safety Department, Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna (IZSLER), Via A. Bianchi 9, 25124 Brescia, Italy
- Centro di Referenza Nazionale per i Rischi Emergenti in Sicurezza Alimentare—CRESA, Via A. Bianchi 9, 25124 Brescia, Italy
| | - Virginia Filipello
- Food Safety Department, Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna (IZSLER), Via A. Bianchi 9, 25124 Brescia, Italy
| |
Collapse
|
41
|
De Simone G, di Masi A, Ascenzi P. Strategies of Pathogens to Escape from NO-Based Host Defense. Antioxidants (Basel) 2022; 11:2176. [PMID: 36358549 PMCID: PMC9686644 DOI: 10.3390/antiox11112176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 06/22/2024] Open
Abstract
Nitric oxide (NO) is an essential signaling molecule present in most living organisms including bacteria, fungi, plants, and animals. NO participates in a wide range of biological processes including vasomotor tone, neurotransmission, and immune response. However, NO is highly reactive and can give rise to reactive nitrogen and oxygen species that, in turn, can modify a broad range of biomolecules. Much evidence supports the critical role of NO in the virulence and replication of viruses, bacteria, protozoan, metazoan, and fungi, thus representing a general mechanism of host defense. However, pathogens have developed different mechanisms to elude the host NO and to protect themselves against oxidative and nitrosative stress. Here, the strategies evolved by viruses, bacteria, protozoan, metazoan, and fungi to escape from the NO-based host defense are overviewed.
Collapse
Affiliation(s)
| | | | - Paolo Ascenzi
- Laboratorio Interdipartimentale di Microscopia Elettronica, Via della Vasca Navale 79, 00146 Roma, Italy
| |
Collapse
|
42
|
Narayanan L, Ozdemir O, Alugubelly N, Ramachandran R, Banes M, Lawrence M, Abdelhamed H. Identification of genetic elements required for Listeria monocytogenes growth under limited nutrient conditions and virulence by a screening of transposon insertion library. Front Microbiol 2022; 13:1007657. [PMID: 36312968 PMCID: PMC9608667 DOI: 10.3389/fmicb.2022.1007657] [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: 07/30/2022] [Accepted: 09/22/2022] [Indexed: 01/29/2023] Open
Abstract
Listeria monocytogenes, the causative agent of listeriosis, displays a lifestyle ranging from saprophytes in the soil to pathogenic as a facultative intracellular parasite in host cells. In the current study, a random transposon (Tn) insertion library was constructed in L. monocytogenes strain F2365 and screened to identify genes and pathways affecting in vitro growth and fitness in minimal medium (MM) containing different single carbohydrate as the sole carbon source. About 2,000 Tn-mutants were screened for impaired growth in MM with one of the following carbon sources: glucose, fructose, mannose, mannitol, sucrose, glycerol, and glucose 6-phosphate (G6P). Impaired or abolished growth of L. monocytogenes was observed for twenty-one Tn-mutants with disruptions in genes encoding purine biosynthesis enzymes (purL, purC, purA, and purM), pyrimidine biosynthesis proteins (pyrE and pyrC), ATP synthase (atpI and atpD2), branched-chain fatty acids (BCFA) synthesis enzyme (bkdA1), a putative lipoprotein (LMOF2365_2387 described as LP2387), dUTPase family protein (dUTPase), and two hypothetical proteins. All Tn-mutants, except the atpD2 mutant, grew as efficiently as wild-type strain in a nutrient rich media. The virulence of twenty-one Tn-mutants was assessed in mice at 72 h following intravenous (IV) infection. The most attenuated mutants had Tn insertions in purA, hypothetical protein (LMOf2365_0064 described as HP64), bkdA1, dUTPase, LP2387, and atpD2, confirming the important role of these genes in pathogenesis. Six Tn-mutants were then tested for ability to replicate intracellularly in murine macrophage J774.1 cells. Significant intracellular growth defects were observed in two Tn-mutants with insertions in purA and HP64 genes, suggesting that an intact purine biosynthesis pathway is important for intracellular growth of L. monocytogens. These findings may not be fully generalized to all of L. monocytogenes strains due to their genetic diversity. In conclusion, Tn-mutagenesis identified that biosynthesis of purines, pyrimidines, ATP, and BCFA are important for L. monocytogens pathogenesis. Purine and pyrimidine auxotrophs play an important role in the pathogenicity in other bacterial pathogens, but our study also revealed new proteins essential for both growth in MM and L. monocytogenes strain F2365 virulence.
Collapse
Affiliation(s)
- Lakshmi Narayanan
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States,Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States
| | - Ozan Ozdemir
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States
| | - Navatha Alugubelly
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States
| | - Reshma Ramachandran
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States,Department of Poultry Science, Mississippi State University, Mississippi State, MS, United States
| | - Michelle Banes
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States
| | - Mark Lawrence
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States
| | - Hossam Abdelhamed
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States,*Correspondence: Hossam Abdelhamed,
| |
Collapse
|
43
|
Rahman S, Das AK. A subtractive proteomics and immunoinformatics approach towards designing a potential multi-epitope vaccine against pathogenic Listeriamonocytogenes. Microb Pathog 2022; 172:105782. [PMID: 36150556 DOI: 10.1016/j.micpath.2022.105782] [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: 04/28/2022] [Revised: 08/04/2022] [Accepted: 09/11/2022] [Indexed: 11/29/2022]
Abstract
Listeria monocytogenes is the causative agent of listeriosis, which is dangerous for pregnant women, the elderly or individuals with a weakened immune system. Individuals with leukaemia, cancer, HIV/AIDS, kidney transplant and steroid therapy suffer from immunological damage are menaced. World Health Organization (WHO) reports that human listeriosis has a high mortality rate of 20-30% every year. To date, no vaccine is available to treat listeriosis. Thereby, it is high time to design novel vaccines against L. monocytogenes. Here, we present computational approaches to design an antigenic, stable and safe vaccine against the L. monocytogenes that could help to control the infections associated with the pathogen. Three vital pathogenic proteins of L. monocytogenes, such as Listeriolysin O (LLO), Phosphatidylinositol-specific phospholipase C (PI-PLC), and Actin polymerization protein (ActA), were selected using a subtractive proteomics approach to design the multi-epitope vaccine (MEV). A total of 5 Cytotoxic T-lymphocyte (CTL) and 9 Helper T-lymphocyte (HTL) epitopes were predicted from these selected proteins. To design the multi-epitope vaccine (MEV) from the selected proteins, CTL epitopes were joined with the AAY linker, and HTL epitopes were joined with the GPGPG linker. Additionally, a human β-defensin-3 (hBD-3) adjuvant was added to the N-terminal side of the final MEV construct to increase the immune response to the vaccine. The final MEV was predicted to be antigenic, non-allergen and non-toxic in nature. Physicochemical property analysis suggested that the MEV construct is stable and could be easily purified through the E. coli expression system. This in-silico study showed that MEV has a robust binding interaction with Toll-like receptor 2 (TLR2), a key player in the innate immune system. Current subtractive proteomics and immunoinformatics study provides a background for designing a suitable, safe and effective vaccine against pathogenic L. monocytogenes.
Collapse
Affiliation(s)
- Shakilur Rahman
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, West Bengal, India
| | - Amit Kumar Das
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, West Bengal, India.
| |
Collapse
|
44
|
Sibanda T, Buys EM. Listeria monocytogenes Pathogenesis: The Role of Stress Adaptation. Microorganisms 2022; 10:microorganisms10081522. [PMID: 36013940 PMCID: PMC9416357 DOI: 10.3390/microorganisms10081522] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/08/2022] [Accepted: 07/19/2022] [Indexed: 12/13/2022] Open
Abstract
Adaptive stress tolerance responses are the driving force behind the survival ability of Listeria monocytogenes in different environmental niches, within foods, and ultimately, the ability to cause human infections. Although the bacterial stress adaptive responses are primarily a necessity for survival in foods and the environment, some aspects of the stress responses are linked to bacterial pathogenesis. Food stress-induced adaptive tolerance responses to acid and osmotic stresses can protect the pathogen against similar stresses in the gastrointestinal tract (GIT) and, thus, directly aid its virulence potential. Moreover, once in the GIT, the reprogramming of gene expression from the stress survival-related genes to virulence-related genes allows L. monocytogenes to switch from an avirulent to a virulent state. This transition is controlled by two overlapping and interlinked transcriptional networks for general stress response (regulated by Sigma factor B, (SigB)) and virulence (regulated by the positive regulatory factor A (PrfA)). This review explores the current knowledge on the molecular basis of the connection between stress tolerance responses and the pathogenesis of L. monocytogenes. The review gives a detailed background on the currently known mechanisms of pathogenesis and stress adaptation. Furthermore, the paper looks at the current literature and theories on the overlaps and connections between the regulatory networks for SigB and PrfA.
Collapse
Affiliation(s)
- Thulani Sibanda
- Department of Consumer and Food Sciences, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa;
- Department of Applied Biology and Biochemistry, National University of Science and Technology, Bulawayo P.O. Box AC939, Zimbabwe
| | - Elna M. Buys
- Department of Consumer and Food Sciences, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa;
- Correspondence:
| |
Collapse
|
45
|
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
|
46
|
Muthulakshmi L, Suganya K, Murugan M, Annaraj J, Duraipandiyan V, Al Farraj DA, Elshikh MS, Juliet A, Pasupuleti M, Arockiaraj J. Antibiofilm efficacy of novel biogenic silver nanoparticles from Terminalia catappa against food-borne Listeria monocytogenes ATCC 15,313 and mechanisms investigation in-vivo and in-vitro. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2022; 34:102083. [DOI: 10.1016/j.jksus.2022.102083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2023]
|
47
|
Huitian G, Xiuai D, Chenglin Z, Xinfeng W, Yuanyuan L, Yanan S, Qihang C, Huiwen X. Transcriptional Analysis of Listeria monocytogenes Invasion of Macrophages. APPL BIOCHEM MICRO+ 2022. [DOI: 10.1134/s000368382203005x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
48
|
Cai G, Zhong F, Cao Q, Bai Y, Zou H, Gu J, Yuan Y, Zhu G, Liu Z, Bian J. ZEA and DON inhibited inflammation after L. monocytogenes infection and induced ribosomal hyperfunction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 236:113470. [PMID: 35395601 DOI: 10.1016/j.ecoenv.2022.113470] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 06/14/2023]
Abstract
The complex microbial community in food environment is a major problem of human or animal health and safety. Mycotoxins and food-borne bacteria can both induce inflammation in the body and cause a series of changes in biological functions. In this study, mice were gavaged with low doses of ZEA, DON, or ZEA + DON, and then infected with L. monocytogenes. A cytokine microarray, including 40 inflammation-related serum cytokines, and proteomics were used to verify the effects of ZEA, DON, and ZEA + DON on the host inflammation and biological function after L. monocytogenes infection. The results showed that mononucleosis after bacterial infection was inhibited by ZEA, DON, and ZEA + DON, while the balance of macrophage differentiation was shifted toward M2-type. ZEA, DON, and ZEA + DON decreased the levels of serum proinflammatory cytokines IL-1β and IL-12 after infection. In addition, the signal of the NF-κB pathway was inhibited. Proteomic results showed that ZEA, DON, and ZEA + DON led to biological dysfunction in ribosomal and metabolic cells, primarily leading to abnormal ribosomal hyperfunction. This study showed that ZEA, DON, and ZEA + DON can aggravate disease progression by inhibiting the inflammatory response following foodborne bacterial infection. These metabolites may also disrupt normal biological functions, which may lead to ribosomal hyperfunction, making bacterial clearance more difficult.
Collapse
Affiliation(s)
- Guodong Cai
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Fang Zhong
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Qianying Cao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Yuni Bai
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China.
| |
Collapse
|
49
|
Liu S, He X, Zhang T, Zhao K, Xiao C, Tong Z, Jin L, He N, Deng Y, Li S, Guo Y, Chen Z. Highly sensitive smartphone-based detection of Listeria monocytogenes using SYTO9. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.11.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
50
|
Gou H, Liu Y, Shi W, Nan J, Wang C, Sun Y, Cao Q, Wei H, Song C, Tian C, Wei Y, Xue H. The Characteristics and Function of Internalin G in Listeria monocytogenes. Pol J Microbiol 2022; 71:63-71. [PMID: 35635167 PMCID: PMC9152910 DOI: 10.33073/pjm-2022-009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/10/2022] [Indexed: 11/05/2022] Open
Abstract
In order to clarified characteristics and function of internalin G (inlG) in Listeria monocytogenes ATCC®19111 (1/2a) (LM), the immune protection of the inlG was evaluated in mice, the homologous recombination was used to construct inlG deletion strains, and their biological characteristics were studied by the transcriptomics analysis. As a result, the immunization of mice with the purified protein achieved a protective effect against bacterial infection. The deletion strain LM-AinlG was successfully constructed with genetic stability. The mouse infection test showed that the virulence of LM was decreased after the deletion of the inlG gene. The deletion strain showed enhanced adhesion to and invasion of Caco-2 cells. Compared to the wild strain, 18 genes were up-regulated, and 24 genes were down-regulated in the LM-AinlG. This study has laid a foundation for further research on the function of inlG and the pathogenesis of LM. In this study, immunization of mice with the purified inlG protein achieved a protective effect against Listeria monocytogenes infection. The virulence of LM-ΔinlG was decreased by mouse infection. However, the adhesion and invasion ability to Caco-2 cell were enhanced. Compared to the wild strain, 18 genes were up-regulated, and 24 genes were down-regulated in the LM-ΔinlG. This study has laid a foundation for further study of the function of the inlG and the listeriosis.
Collapse
Affiliation(s)
- Huitian Gou
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yuanyuan Liu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Wenjing Shi
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Jinyu Nan
- Jiuquan City Animal Control Disease Center, Jiuquan, China
| | - Chuan Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yanan Sun
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Qihang Cao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Huilin Wei
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Chen Song
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Changqing Tian
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yanquan Wei
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Huiwen Xue
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|