1
|
Thye T, Krumkamp R, Lusingu JPA, Ofori LA, Minja DTR, Flieger A, Gesase S, Phillips R, Simon S, Obiri-Danso K, Akenten CW, Mbwana J, Paintsil E, Ascofare OM, Jaeger A, Lamshöft M, Eibach D, Loag W, Berg S, May J, Dekker D. Non-typhoidal Salmonella transmission reservoirs in Sub-Saharan Africa: a genomic assessment from a one health perspective. Antimicrob Resist Infect Control 2025; 14:46. [PMID: 40361223 PMCID: PMC12070793 DOI: 10.1186/s13756-025-01561-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND In sub-Saharan Africa, invasive non-typhoidal Salmonella disease, characterized by bloodstream infections with high mortality rates, poses a significant public health burden. In Africa, Salmonella enterica, which are typically livestock- associated pathogens in industrialised countries, have genetically evolved and anthroponotic transmission has been proposed for S. Typhimurium ST313. In this study, we investigated the hypothesis of an exclusively anthroponotic transmission reservoir of Salmonella enterica ST313 and aimed to identify reservoirs for other Salmonella spp., shedding light on their occurrence in different ecological niches. METHODS This study used a One Health approach and Salmonella were isolated from humans, livestock and the environment, in Tanzania and in Ghana. Salmonella spp. were identified by biochemical methods and antibiotic susceptibility was tested. Isolates were subjected to whole genome sequencing. RESULTS Out of 9,086 collected samples, 222 Salmonella enterica were identified comprising 58 serovars. The highest level of antimicrobial resistance was found in humans with emerging fluroquinolone resistance and multidrug resistance being highest in isolates from blood cultures (24%, n/N = 11/46). For the invasive strains, the sequence types S. Typhimurium ST313 and ST19 were most common and ST313 was associated with multidrug resistance, followed by S. Enteritidis ST11 and ST147 and S. Dublin ST10. An overlap of sequence types amongst human-livestock and human-environmental strains was detected for S. Typhimurium ST19 but not found for ST313 and the two serovars Dublin and Enteritidis. CONCLUSIONS Our study adds further evidence of S. Typhimurium ST313 being restricted to a human reservoir and linked to multidrug resistance. Additionally, our study provides comprehensive insights into Salmonella genetic diversity and distribution among humans, animals and the environment in Ghana and in Tanzania. This sheds light on other potential reservoirs for infections, all of which show antimicrobial resistance. Further research into stool carriage is warranted, encompassing patients with invasive disease and those with and without diarrhoea, to identify transmission reservoirs in particular for invasive disease-causing strains. These findings underscore the need for integrated One Health approaches to effectively monitor and manage salmonellosis and mitigate public health risks. Continued research into the spread of Salmonella spp. and its evolution is crucial for targeted interventions and disease control.
Collapse
Affiliation(s)
- Thorsten Thye
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
| | - Ralf Krumkamp
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
- German Centre for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Inhoffenstr.7, Brunswick, 338124, Germany
| | - John P A Lusingu
- National Institute for Medical Research (NIMR), Bombo Rd., Tanga, Tanzania
| | - Linda Aurelia Ofori
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology (KNUST), KNUST Campus, Kumasi, Ghana
| | - Daniel T R Minja
- National Institute for Medical Research (NIMR), Bombo Rd., Tanga, Tanzania
| | - Antje Flieger
- Robert Koch Institute (RKI), National Reference Center for Salmonella and Other Bacterial Enteric Pathogens, Burgstraße 37, 38855, Wernigerode, Germany
| | - Samwel Gesase
- National Institute for Medical Research (NIMR), Bombo Rd., Tanga, Tanzania
| | - Richard Phillips
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Asnogya Rd., Kumasi, Ghana
| | - Sandra Simon
- Robert Koch Institute (RKI), National Reference Center for Salmonella and Other Bacterial Enteric Pathogens, Burgstraße 37, 38855, Wernigerode, Germany
| | - Kwasi Obiri-Danso
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology (KNUST), KNUST Campus, Kumasi, Ghana
| | - Charity Wiafe Akenten
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Asnogya Rd., Kumasi, Ghana
- One Health Bacteriology Research Group, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
| | - Joyce Mbwana
- National Institute for Medical Research (NIMR), Bombo Rd., Tanga, Tanzania
| | - Ellis Paintsil
- One Health Bacteriology Research Group, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
| | - Oumou Maiga Ascofare
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Asnogya Rd., Kumasi, Ghana
| | - Anna Jaeger
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
| | - Maike Lamshöft
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
- German Centre for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Inhoffenstr.7, Brunswick, 338124, Germany
| | - Daniel Eibach
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
| | - Wibke Loag
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
| | - Stefan Berg
- One Health Bacteriology Research Group, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
| | - Jürgen May
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany
- German Centre for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems, Inhoffenstr.7, Brunswick, 338124, Germany
| | - Denise Dekker
- One Health Bacteriology Research Group, Bernhard Nocht Institute for Tropical Medicine (BNITM), Bernhard-Nochtstr. 74, 20359, Hamburg, Germany.
| |
Collapse
|
2
|
Boudrioua A, Joiner JD, Grin I, Kronenberger T, Korotkov VS, Steinchen W, Kohler A, Schminke S, Schulte JC, Pietsch M, Naini A, Kalverkamp S, Hotop SK, Coyle T, Piselli C, Coles M, Rox K, Marschal M, Bange G, Flieger A, Poso A, Brönstrup M, Hartmann MD, Wagner S. Discovery of synthetic small molecules targeting the central regulator of Salmonella pathogenicity. SCIENCE ADVANCES 2025; 11:eadr5235. [PMID: 40215303 PMCID: PMC11988454 DOI: 10.1126/sciadv.adr5235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 03/04/2025] [Indexed: 04/14/2025]
Abstract
The enteric pathogen Salmonella enterica serovar Typhimurium relies on the activity of effector proteins to invade, replicate, and disseminate into host epithelial cells and other tissues, thereby causing disease. Secretion and injection of effector proteins into host cells is mediated by dedicated secretion systems, which hence represent major virulence determinants. Here, we report the identification of a synthetic small molecule with drug-like properties, C26, which suppresses the secretion of effector proteins and consequently hinders bacterial invasion of eukaryotic cells. C26 binds to and inhibits HilD, the transcriptional regulator of the major secretion systems. Although sharing the same binding pocket as the previously described long-chain fatty acid ligands, C26 inhibits HilD with a unique binding mode and a distinct mechanism. We provide evidence of intramacrophage activity and present analogs with improved potency and suitability as scaffolds to develop antivirulence agents against Salmonella infections in humans and animals.
Collapse
Affiliation(s)
- Abdelhakim Boudrioua
- Section of Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- German Center for Infection Research (DZIF), partner-site Tübingen, 72076 Tübingen, Germany
| | - Joe D. Joiner
- Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany
| | - Iwan Grin
- Section of Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- German Center for Infection Research (DZIF), partner-site Tübingen, 72076 Tübingen, Germany
| | - Thales Kronenberger
- German Center for Infection Research (DZIF), partner-site Tübingen, 72076 Tübingen, Germany
- Institute of Medical Microbiology and Hygiene, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
| | - Vadim S. Korotkov
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Wieland Steinchen
- Center for Synthetic Microbiology, Philipps University of Marburg, Karl-von-Frisch-Str. 14, 35043 Marburg, Germany
- Department of Chemistry, Philipps University of Marburg, Hans Meerwein-Str. 4, 35043 Marburg, Germany
| | - Alexander Kohler
- Section of Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- German Center for Infection Research (DZIF), partner-site Tübingen, 72076 Tübingen, Germany
| | - Sophie Schminke
- Section of Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - Julia-Christina Schulte
- Section of Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- German Center for Infection Research (DZIF), partner-site Tübingen, 72076 Tübingen, Germany
| | - Michael Pietsch
- Unit for Enteropathogenic Bacteria and Legionella (FG11) and National Reference Centre for Salmonella and other Bacterial Enterics, Robert Koch Institute (RKI), Burgstr. 37, 38855 Wernigerode, Germany
| | - Arun Naini
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Simon Kalverkamp
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Sven-Kevin Hotop
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Travis Coyle
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Claudio Piselli
- German Center for Infection Research (DZIF), partner-site Tübingen, 72076 Tübingen, Germany
- Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany
| | - Murray Coles
- Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany
| | - Katharina Rox
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Matthias Marschal
- Institute of Medical Microbiology and Hygiene, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - Gert Bange
- Center for Synthetic Microbiology, Philipps University of Marburg, Karl-von-Frisch-Str. 14, 35043 Marburg, Germany
- Department of Chemistry, Philipps University of Marburg, Hans Meerwein-Str. 4, 35043 Marburg, Germany
| | - Antje Flieger
- Unit for Enteropathogenic Bacteria and Legionella (FG11) and National Reference Centre for Salmonella and other Bacterial Enterics, Robert Koch Institute (RKI), Burgstr. 37, 38855 Wernigerode, Germany
| | - Antti Poso
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery (TüCAD2), University of Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Institute of Organic Chemistry and Biomolecular Drug Research Centre (BMWZ), Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Marcus D. Hartmann
- German Center for Infection Research (DZIF), partner-site Tübingen, 72076 Tübingen, Germany
- Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Samuel Wagner
- Section of Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- German Center for Infection Research (DZIF), partner-site Tübingen, 72076 Tübingen, Germany
- Excellence Cluster “Controlling Microbes to Fight Infections” (CMFI), Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| |
Collapse
|
3
|
Zhou H, Jia C, Shen P, Huang C, Teng L, Wu B, Wang Z, Wang H, Xiao Y, Baker S, Weill FX, Li Y, Yue M. Genomic census of invasive nontyphoidal Salmonella infections reveals global and local human-to-human transmission. Nat Med 2025:10.1038/s41591-025-03644-4. [PMID: 40205197 DOI: 10.1038/s41591-025-03644-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 03/05/2025] [Indexed: 04/11/2025]
Abstract
Extraintestinal infections caused by Enterobacteriaceae represent a global concern, further exacerbated by the growing prevalence of antimicrobial resistance (AMR). Among these, invasive nontyphoidal Salmonella (iNTS) infections have become increasingly challenging to manage, and their global spread remains poorly understood. Here we compiled 1,115 patient records and generated a comprehensive genomic dataset on iNTS. Age and sex emerged as significant risk factors, with Salmonella Enteritidis identified as a major cause. We observed serovar-specific AMR patterns, with notable resistance to fluoroquinolones and third-generation cephalosporins. A global phylogenomic analysis of Enteritidis revealed three distinct clades, highlighting the accumulation of AMR determinants during its international spread. Importantly, our genomic and transmission analyses suggest that iNTS infections may involve human-to-human transmission, with diarrheal patients acting as potential intermediaries, deviating from typical zoonotic pathways. Collectively, our newly generated cohort and iNTS genomic dataset provide a framework for precise local iNTS burden and underscore emerging transmission trends.
Collapse
Affiliation(s)
- Haiyang Zhou
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, People's Republic of China
- Hainan Institute of Zhejiang University, Sanya, People's Republic of China
| | - Chenghao Jia
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, People's Republic of China
- Hainan Institute of Zhejiang University, Sanya, People's Republic of China
| | - Ping Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Chenghu Huang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, People's Republic of China
- Hainan Institute of Zhejiang University, Sanya, People's Republic of China
| | - Lin Teng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, People's Republic of China
| | - Beibei Wu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, People's Republic of China
| | - Zining Wang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, People's Republic of China
- Hainan Institute of Zhejiang University, Sanya, People's Republic of China
| | - Haoqiu Wang
- Affiliated Hangzhou Xixi Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yonghong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Stephen Baker
- School of Clinical Medicine, University of Cambride, Cambridge, UK
| | - François-Xavier Weill
- Institut Pasteur, Université Paris Cité, Unité des Bactéries pathogènes entériques, Paris, France
| | - Yan Li
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Hainan Institute of Zhejiang University, Sanya, People's Republic of China
| | - Min Yue
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, People's Republic of China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, People's Republic of China.
- Hainan Institute of Zhejiang University, Sanya, People's Republic of China.
| |
Collapse
|
4
|
Worley MJ. Salmonella Type III Secretion System Effectors. Int J Mol Sci 2025; 26:2611. [PMID: 40141253 PMCID: PMC11942329 DOI: 10.3390/ijms26062611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Salmonella is estimated to infect between 200 million and over 1 billion people per year. The exact number is not known, as many cases go unreported. Integral to the pathogenesis of Salmonella, as well as numerous other Gram-negative pathogens, is its type III effectors. Salmonella possesses two distinct type III secretion systems, encoded by Salmonella pathogenicity island-1 and Salmonella pathogenicity island-2. Together, they secrete at least 49 type III effectors into host cells that are collectively responsible for many of the virulence attributes of this pathogen. These virulence factors facilitate the invasion of host cells, induce and attenuate inflammation, and change the migratory properties of infected phagocytes, among other things. The effects of all type III effectors on Salmonella virulence are discussed.
Collapse
Affiliation(s)
- Micah J Worley
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
5
|
Jia C, Huang C, Zhou H, Zhou X, Wang Z, Siddique A, Kang X, Cao Q, Huang Y, He F, Li Y, Yue M. Avian-specific Salmonella transition to endemicity is accompanied by localized resistome and mobilome interaction. eLife 2025; 13:RP101241. [PMID: 40035424 PMCID: PMC11879110 DOI: 10.7554/elife.101241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
Bacterial regional demonstration after global dissemination is an essential pathway for selecting distinct finesses. However, the evolution of the resistome during the transition to endemicity remains unaddressed. Using the most comprehensive whole-genome sequencing dataset of Salmonella enterica serovar Gallinarum (S. Gallinarum) collected from 15 countries, including 45 newly recovered samples from two related local regions, we established the relationship among avian-specific pathogen genetic profiles and localization patterns. Initially, we revealed the international transmission and evolutionary history of S. Gallinarum to recent endemicity through phylogenetic analysis conducted using a spatiotemporal Bayesian framework. Our findings indicate that the independent acquisition of the resistome via the mobilome, primarily through plasmids and transposons, shapes a unique antimicrobial resistance profile among different lineages. Notably, the mobilome-resistome combination among distinct lineages exhibits a geographical-specific manner, further supporting a localized endemic mobilome-driven process. Collectively, this study elucidates resistome adaptation in the endemic transition of an avian-specific pathogen, likely driven by the localized farming style, and provides valuable insights for targeted interventions.
Collapse
Affiliation(s)
- Chenghao Jia
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
| | - Chenghu Huang
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
- Hainan Institute of Zhejiang UniversityNingboChina
| | - Haiyang Zhou
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
- Hainan Institute of Zhejiang UniversityNingboChina
| | - Xiao Zhou
- Ningbo Academy of Agricultural SciencesNingboChina
| | - Zining Wang
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
- Hainan Institute of Zhejiang UniversityNingboChina
| | - Abubakar Siddique
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
- Hainan Institute of Zhejiang UniversityNingboChina
| | - Xiamei Kang
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
| | - Qianzhe Cao
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
| | - Yingying Huang
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
- Hainan Institute of Zhejiang UniversityNingboChina
| | - Fang He
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
- ZJU-Xinchang Joint Innovation Centre (TianMu Laboratory), Gaochuang Hi-Tech ParkZhejiangChina
| | - Yan Li
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
- Hainan Institute of Zhejiang UniversityNingboChina
| | - Min Yue
- Department of Veterinary Medicine, Zhejiang University College of Animal SciencesHangzhouChina
- Hainan Institute of Zhejiang UniversityNingboChina
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of SciencesHangzhouChina
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| |
Collapse
|
6
|
Zhao K, Jin J, Liao Y, Liu A, Liu W, Wu W. IS 15DIV-flanked composite transposon harboring bla NDM-5 in multidrug-resistant Salmonella Typhimurium. iScience 2025; 28:111720. [PMID: 39877901 PMCID: PMC11772981 DOI: 10.1016/j.isci.2024.111720] [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: 09/23/2024] [Revised: 10/29/2024] [Accepted: 12/28/2024] [Indexed: 01/31/2025] Open
Abstract
Multidrug-resistant Salmonella Typhimurium has emerged as a global public health concern. Asymptomatic gastrointestinal carriage is a key factor in the spread of antibiotic-resistant bacteria. However, it is challenging to obtain direct evidence of in vivo transfer of mobile genetic elements (MGEs). Here, we found that MGEs harboring bla NDM-5 were transferred from asymptomatic Escherichia coli (LS20223694) to Salmonella Typhimurium (LS20223695) in a child. BLAST analyses demonstrated that the IS15DIV-flanked composite transposon of pLS20223695_NDM5 showed high similarity with pLS20223694_NDM5 and two previously reported plasmids, suggesting the possibility of genetic recombination. Besides, conjugation experiments showed that the transconjugant carried the incompatibility group I1 (IncI1)-I(alpha) plasmid replicon with the bla NDM-5 and sul1 genes, indicating that pLS20223695_NDM5 is a conjugative plasmid with transferability. Our study provides insights into the genetic basis of an IS15DIV-flanked composite transposon in Salmonella Typhimurium.
Collapse
Affiliation(s)
- Kaiting Zhao
- Department of Hematology, Lishui People’s Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Jing Jin
- Department of Clinical Laboratory, Lishui People’s Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Yuan Liao
- Department of Clinical Laboratory, Lishui People’s Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Aixia Liu
- Department of Clinical Laboratory, Lishui People’s Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Wugao Liu
- Department of Clinical Laboratory, Lishui People’s Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Weiping Wu
- Department of Clinical Laboratory, Lishui People’s Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| |
Collapse
|
7
|
Zhou N, Ding Y, He T, Sun Y, Chen H, Huang M, Li T. Characterization and Protective Efficacy of a Salmonella Typhimurium ATCC 14028 sptP Mutant as a Live Attenuated Vaccine Candidate. Vaccines (Basel) 2025; 13:150. [PMID: 40006697 PMCID: PMC11860608 DOI: 10.3390/vaccines13020150] [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: 12/07/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Salmonella Typhimurium poses a substantial health risk to both humans and animals. This study evaluated the potential of using the Salmonella Typhimurium ΔsptP mutant as a live-attenuated vaccine candidate by constructing it through homologous recombination and assessing its key biological properties, including growth characteristics, immunogenicity, and protective efficacy. METHODS We generated the ΔsptP mutant through targeted gene deletion, ensuring the preservation of the bacterial strain's growth and stability. In vitro and in vivo assays were performed to compare the invasive capabilities between the mutant and the wild-type strains. Specifically, we examined the invasion into RAW264.7 murine macrophages and mice. Furthermore, the virulence of the mutant was evaluated by determining the median lethal dose (LD50). To evaluate immunogenicity and protection, mice were immunized with 2 × 104 CFUs of the ΔsptP mutant, followed by a booster immunization, and then challenged with a virulent strain. RESULTS The ΔsptP mutant exhibited no significant changes in growth characteristics or genetic stability compared to the wild-type strain. However, it demonstrated a significantly diminished capacity for invasion in both murine macrophages and mice. The LD50 for the mutant was 39.92-fold higher than that of the wild-type, indicating a marked reduction in virulence. Mice immunized with the ΔsptP mutant and administered a booster immunization exhibited 87.5% protection against challenge with a virulent strain, as compared to the PBS control group. Moreover, the mutant induced IgG antibody levels comparable to those induced by the wild-type strain. CONCLUSIONS The ΔsptP mutant of Salmonella Typhimurium exhibits markedly reduced virulence while retaining robust immunogenicity and protective efficacy. These findings suggest that the ΔsptP mutant is a promising candidate for a live-attenuated vaccine, potentially providing an effective strategy to prevent Salmonella Typhimurium infections.
Collapse
Affiliation(s)
- Nanlong Zhou
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| | - Yonghui Ding
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| | - Ting He
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| | - Yuling Sun
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| | - Hongfang Chen
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| | - Meiling Huang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Tiansen Li
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| |
Collapse
|
8
|
Schwieters A, Cole AL, Rego E, Gao C, Kebriaei R, Wysocki VH, Gunn JS, Ahmer BMM. MtlD as a therapeutic target for intestinal and systemic bacterial infections. J Bacteriol 2025; 207:e0048024. [PMID: 39727397 PMCID: PMC11784389 DOI: 10.1128/jb.00480-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
The ability to treat infections is threatened by the rapid emergence of antibiotic resistance among pathogenic microbes. Therefore, new antimicrobials are needed. Here we evaluate mannitol-1-phosphate 5-dehydrogenase (MtlD) as a potential new drug target. In many bacteria, mannitol is transported into the cell and phosphorylated by MtlA, the EIICBA component of a phosphoenolpyruvate-dependent sugar phosphotransferase system. MtlD catalyzes the conversion of mannitol-1-phosphate (Mtl-1P) to fructose-6-phosphate, which enters the glycolytic pathway. Mutants lacking mtlD are sensitive to mannitol due to accumulation of Mtl-1P. Here, we constructed mtlD mutants in four different bacterial species (Cronobacter sakazakii, Pseudomonas aeruginosa, five serovars of Salmonella enterica, and three strains of Escherichia coli), confirming and quantifying their mannitol sensitivity. The quantification of mannitol sensitivity in vitro was complicated by an inoculum effect and a resumption of growth following mannitol intoxication. The rate of resumption at different mannitol concentrations and cell population densities is fairly constant and reveals what is likely an intoxication processing rate. Provision of mannitol in drinking water, or by intraperitoneal injection, dramatically attenuates infection of a Salmonella enterica serovar Typhimurium mtlD mutant in mouse models of both gastroenteritis and systemic infection. Using CC003/Unc mice, we find that a mtlD mutant of Salmonella enterica serovar Typhi is also attenuated by provision of mannitol in drinking water. Therefore, we postulate that MtlD could be a valuable new therapeutic target. IMPORTANCE The ability to treat infections is threatened by the rapid emergence of antibiotic resistance. Mannitol is a polyol used in human medicine and the food industry. During catabolism of mannitol, many bacteria transport mannitol across the inner membrane forming the toxic intermediate mannitol-1-phosphate (Mtl-1P). Mtl-1P must be processed by mannitol dehydrogenase (MtlD) or it accumulates intracellularly, causing growth attenuation. We test and confirm here that mtlD mutants of Escherichia coli (including UPEC, and EHEC), Salmonella (including serovars Typhi, and Paratyphi A, B, and C), Cronobacter, and Pseudomonas experience mannitol sensitivity in vitro. Furthermore, providing mannitol in drinking water can alleviate both gastrointestinal and systemic Salmonella infections in mice. This suggests that inhibition of MtlD could be a viable antimicrobial strategy.
Collapse
Affiliation(s)
- Andrew Schwieters
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| | - Allysa L. Cole
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
- Center for Microbial Pathogenesis, Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Emily Rego
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| | - Chengyu Gao
- Campus Chemical Instrument Center, The Ohio State University, Columbus, Ohio, USA
| | - Razieh Kebriaei
- Department of Outcomes and Translational Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Vicki H. Wysocki
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
- National Resource for Native MS-Guided Structural Biology, Columbus, Ohio, USA
| | - John S. Gunn
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
- Center for Microbial Pathogenesis, Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Brian M. M. Ahmer
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
9
|
Perez-Sepulveda BM, Hinton JCD. Microbe Profile: Salmonella Typhimurium: the master of the art of adaptation. MICROBIOLOGY (READING, ENGLAND) 2025; 171:001521. [PMID: 39786350 PMCID: PMC11893365 DOI: 10.1099/mic.0.001521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025]
Abstract
Salmonella Typhimurium is a major Salmonella serovar that is found globally. It is responsible for outbreaks of self-limiting gastroenteritis that are broadly linked to the industrialization of food production. S. Typhimurium is a pathogen with a broad host range and remarkable metabolic versatility. The ∼5 Mb genome includes the pSLT virulence plasmid and has a characteristic prophage repertoire. The major virulence determinants are encoded by a variety of pathogenicity islands. Emerging multidrug-resistant lineages of epidemics of S. Typhimurium are currently causing bloodstream infections in sub-Saharan Africa. The versatility and adaptability of S. Typhimurium pose an important public health challenge.
Collapse
Affiliation(s)
- Blanca M. Perez-Sepulveda
- Clinical Infection, Microbiology & Immunology Department, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Jay C. D. Hinton
- Clinical Infection, Microbiology & Immunology Department, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
10
|
Li Y, Pulford CV, Díaz P, Perez-Sepulveda BM, Duarte C, Predeus AV, Wiesner M, Heavens D, Low R, Schudoma C, Montaño A, Hall N, Moreno J, Hinton JCD. Potential links between human bloodstream infection by Salmonella enterica serovar Typhimurium and international transmission to Colombia. PLoS Negl Trop Dis 2025; 19:e0012801. [PMID: 39874387 PMCID: PMC11790238 DOI: 10.1371/journal.pntd.0012801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 02/03/2025] [Accepted: 12/19/2024] [Indexed: 01/30/2025] Open
Abstract
Salmonella enterica serovar Typhimurium is a prevalent food-borne pathogen that is usually associated with gastroenteritis infection. S. Typhimurium is also a major cause of bloodstream infections in sub-Saharan Africa, and is responsible for invasive non-typhoidal Salmonella (iNTS) disease. The pathogen also causes bloodstream infection in Colombia, but there has been a lack of information about the S. Typhimurium isolates that were responsible. Here, we investigated the genomic characteristics of 270 S. Typhimurium isolates from bloodstream infection patients in Colombia, collected between 1997 and 2017. We used whole-genome sequencing to analyse multidrug-resistant (MDR) profiles, plasmid distribution, and to define phylogenetic relationships. The study identified the distinct sequence types and phylogenetic clusters of S. Typhimurium prevalent in Colombia. The majority of isolates (90.8%) were ST19, which is distinct from the iNTS-associated S. Typhimurium in sub-Saharan Africa (ST313). The two prominent clusters of MDR S. Typhimurium were either DT104 or closely related to the LT2 reference strain. We used a phylogenetic approach to associate the Colombian clusters with global S. Typhimurium isolates from public databases. By putting the Colombian S. Typhimurium isolates in the context of the global spread of DT104, ST313 and LT2-related variants, we found that the Colombian clusters were introduced to the country via multiple independent events that were consistent with international transmission. We suggest that the acquisition of quinolone and chloramphenicol resistance by the Colombian S. Typhimurium isolates was driven by horizontal gene transfer. Three ST313 isolates that caused bloodstream infection in Colombia were identified. These ST313 isolates were related to the Malawian ST313 lineage 3 & UK ST313, and shared a similarly high invasiveness index. To our knowledge, this is the first report of ST313 in Colombia.
Collapse
Affiliation(s)
- Yan Li
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Caisey V. Pulford
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Paula Díaz
- Grupo de Microbiología, Instituto Nacional de Salud, Bogotá, Colombia
| | - Blanca M. Perez-Sepulveda
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carolina Duarte
- Grupo de Microbiología, Instituto Nacional de Salud, Bogotá, Colombia
| | - Alexander V. Predeus
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Magdalena Wiesner
- Grupo de Microbiología, Instituto Nacional de Salud, Bogotá, Colombia
| | | | - Ross Low
- Earlham Institute, Norwich, United Kingdom
| | | | - Angeline Montaño
- Grupo de Microbiología, Instituto Nacional de Salud, Bogotá, Colombia
| | - Neil Hall
- Earlham Institute, Norwich, United Kingdom
- School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Jaime Moreno
- Grupo de Microbiología, Instituto Nacional de Salud, Bogotá, Colombia
| | - Jay C. D. Hinton
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
11
|
Chen J, Huang L, An H, Wang Z, Kang X, Yin R, Jia C, Jin X, Yue M. One Health approach probes zoonotic non-typhoidal Salmonella infections in China: A systematic review and meta-analysis. J Glob Health 2024; 14:04256. [PMID: 39620281 PMCID: PMC11610537 DOI: 10.7189/jogh.14.04256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024] Open
Abstract
Background Zoonotic infections, particularly those caused by non-typhoidal Salmonella (NTS), pose a significant disease burden. However, there is a notable lack of comprehensive and integrated studies employing the One Health approach to address Salmonella prevalence. In this study, we aimed to analyse NTS spatiotemporal prevalence, serovar distribution, and antimicrobial resistance (AMR) across China. Methods We conducted a systematic review and meta-analysis to understand the dynamics of NTS in a One Health context in China. We searched the CNKI, Wanfang, and PubMed databases for Chinese and English peer-reviewed articles published before 1 January 2022 dealing with Salmonella in the context of China. We examined the dynamic prevalence along the food chain, the risk of dominant serovars and the carriers' regional contribution by principal component analysis, and the AMR burden before and after the ban on using antimicrobials as feed additives across five decades. We used the inverse variance index as an indicator of the inconsistency across studies, and we adopted the restricted maximum likelihood model due to high heterogeneity for analysis with a 95% confidence level for the pooled prevalence estimate. Results Based on 562 retrieved high-quality studies during 1967-2021, representing 5 052 496 samples overall and 80 536 positive samples for NTS isolates, the overall average prevalence was 7.35% (95% confidence interval (CI) = 0.069-0.087), which was regionally relatively higher in northern China (8.19%; 95% CI = 0.078-0.117) than in southern China (6.94%; 95% CI = 0.067-0.088). Poultry was the primary vehicle for serovars Enteritidis and Indiana (especially in the north), while swine and ruminants for Typhimurium and Derby were the first to highlight the regional livestock contribution to serovar prevalence. The overall AMR rate was 73.63% (95% CI = 0.68-0.99), decreasing after the ban on excessive use of feed-based antibiotics in livestock since 2020, with a relatively low resistance towards front-line and last-resort drugs. Conclusions Our study emphasises the importance of adopting a One Health framework to better understand the zoonotic nature of human NTS and highlights the dominant serovars on food contamination and human infection. The similarity in AMR patterns between poultry and human isolates further emphasises the integrated approach for evaluating disease burden and implementing targeted interventions.
Collapse
Affiliation(s)
- Jiaqi Chen
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, Hangzhou; University of Chinese Academy of Sciences, China
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Linlin Huang
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Hongli An
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
- Hainan Institute of Zhejiang University, Sanya, China
| | - Zining Wang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, Hangzhou; University of Chinese Academy of Sciences, China
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Xiamei Kang
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Rui Yin
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Chenghao Jia
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Xiuyan Jin
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Min Yue
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, Hangzhou; University of Chinese Academy of Sciences, China
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
- Hainan Institute of Zhejiang University, Sanya, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Dai Y, Liu R, Yue Y, Song N, Jia H, Ma Z, Gao X, Zhang M, Yuan X, Liu Q, Liu X, Li B, Wang W. A c-di-GMP binding effector STM0435 modulates flagellar motility and pathogenicity in Salmonella. Virulence 2024; 15:2331265. [PMID: 38532247 PMCID: PMC10978029 DOI: 10.1080/21505594.2024.2331265] [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: 09/14/2023] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Flagella play a crucial role in the invasion process of Salmonella and function as a significant antigen that triggers host pyroptosis. Regulation of flagellar biogenesis is essential for both pathogenicity and immune escape of Salmonella. We identified the conserved and unknown function protein STM0435 as a new flagellar regulator. The ∆stm0435 strain exhibited higher pathogenicity in both cellular and animal infection experiments than the wild-type Salmonella. Proteomic and transcriptomic analyses demonstrated dramatic increases in almost all flagellar genes in the ∆stm0435 strain compared to wild-type Salmonella. In a surface plasmon resonance assay, purified STM0435 protein-bound c-di-GMP had an affinity of ~8.383 µM. The crystal structures of apo-STM0435 and STM0435&c-di-GMP complex were determined. Structural analysis revealed that R33, R137, and D138 of STM0435 were essential for c-di-GMP binding. A Salmonella with STM1987 (GGDEF protein) or STM4264 (EAL protein) overexpression exhibits completely different motility behaviours, indicating that the binding of c-di-GMP to STM0435 promotes its inhibitory effect on Salmonella flagellar biogenesis.
Collapse
Affiliation(s)
- Yuanji Dai
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ruirui Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yingying Yue
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Nannan Song
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Haihong Jia
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Zhongrui Ma
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xueyan Gao
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Min Zhang
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xilu Yuan
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Qing Liu
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoyu Liu
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Bingqing Li
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Key Lab for Biotech-Drugs of National Health Commission, Shandong First Medical University, Jinan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong, China
| | - Weiwei Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
13
|
Thakur S, Baines SL, Sia CM, Valcanis M, Judd LM, Howden BP, Newton HJ, Ingle DJ. Genomic epidemiology and phenotypic characterisation of Salmonella enterica serovar Panama in Victoria, Australia. PLoS Negl Trop Dis 2024; 18:e0012666. [PMID: 39565816 PMCID: PMC11616866 DOI: 10.1371/journal.pntd.0012666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/04/2024] [Accepted: 10/30/2024] [Indexed: 11/22/2024] Open
Abstract
Salmonella enterica serovar Panama, a causative agent of non-typhoidal salmonellosis (NTS), is one of several serovars that causes invasive NTS disease (iNTS) in humans. S. Panama is an understudied pathogen, with its pathobiology poorly understood. It is a predominant iNTS serovar in Australia, a high-income country with high rates of salmonellosis, where S. Panama has been documented to have a high odds ratio (13.9-15.26) for causing iNTS. This study investigates the genomic epidemiology and antimicrobial resistance profiles of all S. Panama isolates recovered in Victoria, Australia, between 2000 and 2021. We examined the infection dynamics of S. Panama in seven isolates, representing the genetic diversity of the study population. Two sub-lineages, encompassed within a previously described Asian lineage, were identified. Multi-drug resistance (resistance to ≥3 drug classes) was detected in 46 (51.7%) Australian isolates. The plasmid-mediated colistin resistance gene, mcr1.1, was detected in one Australian S. Panama isolate, carried by an IncI plasmid previously reported in Salmonella and Escherichia coli isolates collected from poultry in South-East Asia. Examination of the intracellular replication dynamics of S. Panama isolates demonstrated diverse phenotypes. In THP-1 derived macrophages, despite low host cell uptake, S. Panama showed higher replication rates over time compared to S. enterica serovar Typhimurium. However, a causative genotype could not be identified to explain this observed phenotype. This study provides insights into the S. Panama isolates circulating in Australia over two-decades, finding that 78% were linked to international travel suggesting importation in Australia. It shows MDR was common in this iNTS serovar, and colistin resistance reported for the first time. It provides the first data on the host-pathogen interactions of S. Panama in Australia, which will aid our collective understanding of the pathobiology of S. Panama and iNTS serovars more broadly.
Collapse
Affiliation(s)
- Samriddhi Thakur
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
- Infection Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Sarah L. Baines
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Cheryll M. Sia
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
| | - Mary Valcanis
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
| | - Louise M. Judd
- Innovation Hub, Centre for Pathogen Genomics, University of Melbourne, Parkville, Victoria Australia
| | - Benjamin P. Howden
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
- Innovation Hub, Centre for Pathogen Genomics, University of Melbourne, Parkville, Victoria Australia
| | - Hayley J. Newton
- Infection Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Danielle J. Ingle
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Graf FE, Goodman RN, Gallichan S, Forrest S, Picton-Barlow E, Fraser AJ, Phan MD, Mphasa M, Hubbard ATM, Musicha P, Schembri MA, Roberts AP, Edwards T, Lewis JM, Feasey NA. Molecular mechanisms of re-emerging chloramphenicol susceptibility in extended-spectrum beta-lactamase-producing Enterobacterales. Nat Commun 2024; 15:9019. [PMID: 39424629 PMCID: PMC11489765 DOI: 10.1038/s41467-024-53391-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024] Open
Abstract
Infections with Enterobacterales (E) are increasingly difficult to treat due to antimicrobial resistance. After ceftriaxone replaced chloramphenicol (CHL) as empiric therapy for suspected sepsis in Malawi in 2004, extended-spectrum beta-lactamase (ESBL)-E rapidly emerged. Concurrently, resistance to CHL in Escherichia coli and Klebsiella spp. decreased, raising the possibility of CHL re-introduction. However, many phenotypically susceptible isolates still carry CHL acetyltransferase (cat) genes. To understand the molecular mechanisms and stability of this re-emerging CHL susceptibility we use a combination of genomics, phenotypic susceptibility assays, experimental evolution, and functional assays for CAT activity. Here, we show that of 840 Malawian E. coli and Klebsiella spp. isolates, 31% have discordant CHL susceptibility genotype-phenotype, and we select a subset of 42 isolates for in-depth analysis. Stable degradation of cat genes by insertion sequences leads to re-emergence of CHL susceptibility. Our study suggests that CHL could be reintroduced as a reserve agent for critically ill patients with ESBL-E infections in Malawi and similar settings and highlights the ongoing challenges in inferring antimicrobial resistance from sequence data.
Collapse
Affiliation(s)
- Fabrice E Graf
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Richard N Goodman
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Sarah Gallichan
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Sally Forrest
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Esther Picton-Barlow
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Alice J Fraser
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Minh-Duy Phan
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Madalitso Mphasa
- Malawi-Liverpool Wellcome Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Alasdair T M Hubbard
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Patrick Musicha
- Malawi-Liverpool Wellcome Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Mark A Schembri
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Adam P Roberts
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Thomas Edwards
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Joseph M Lewis
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Malawi-Liverpool Wellcome Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Nicholas A Feasey
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Malawi-Liverpool Wellcome Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- School of Medicine, University of St Andrews, St Andrews, UK
| |
Collapse
|
15
|
Xiang Y, Zhu K, Min K, Zhang Y, Liu J, Liu K, Han Y, Li X, Du X, Wang X, Huang Y, Li X, Peng Y, Yang C, Liu H, Liu H, Li X, Wang H, Wang C, Wang Q, Jia H, Yang M, Wang L, Wu Y, Cui Y, Chen F, Yang H, Baker S, Xu X, Yang J, Song H, Qiu S. Characterization of a Salmonella enterica serovar Typhimurium lineage with rough colony morphology and multidrug resistance. Nat Commun 2024; 15:6123. [PMID: 39033143 PMCID: PMC11271444 DOI: 10.1038/s41467-024-50331-y] [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: 08/28/2023] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is a major cause of salmonellosis, and the emergence of multidrug-resistant pathovariants has become a growing concern. Here, we investigate a distinct rough colony variant exhibiting a strong biofilm-forming ability isolated in China. Whole-genome sequencing on 2,212 Chinese isolates and 1,739 publicly available genomes reveals the population structure and evolutionary history of the rough colony variants. Characterized by macro, red, dry, and rough (mrdar) colonies, these variants demonstrate enhanced biofilm formation at 28 °C and 37 °C compared to typical rdar colonies. The mrdar variants exhibit extensive multidrug resistance, with significantly higher resistance to at least five classes of antimicrobial agents compared to non-mrdar variants. This resistance is primarily conferred by an IncHI2 plasmid harboring 19 antimicrobial resistance genes. Phylogenomic analysis divides the global collections into six lineages. The majority of mrdar variants belong to sublineage L6.5, which originated from Chinese smooth colony strains and possibly emerged circa 1977. Among the mrdar variants, upregulation of the csgDEFG operons is observed, probably due to a distinct point mutation (-44G > T) in the csgD gene promoter. Pangenome and genome-wide association analyses identify 87 specific accessory genes and 72 distinct single nucleotide polymorphisms associated with the mrdar morphotype.
Collapse
Affiliation(s)
- Ying Xiang
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Kunpeng Zhu
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
- Kaifeng Center for Disease Control and Prevention, Kaifeng, China
| | - Kaiyuan Min
- State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yaowen Zhang
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
- Daxing Center for Disease Control and Prevention, Beijing, China
| | - Jiangfeng Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Kangkang Liu
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Yiran Han
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Xinge Li
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Xinying Du
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Xin Wang
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Ying Huang
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Xinping Li
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Yuqian Peng
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Chaojie Yang
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Hongbo Liu
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Hongbo Liu
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Xiaoying Li
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Hui Wang
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Chao Wang
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Qi Wang
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Huiqun Jia
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Mingjuan Yang
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Ligui Wang
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China
| | - Yarong Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yujun Cui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Fei Chen
- CAS Key Laboratory of Genome Sciences & Information, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Stephen Baker
- University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Xuebin Xu
- Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China.
| | - Juntao Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Hongbin Song
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China.
| | - Shaofu Qiu
- Center for Disease Control and Prevention of Chinese PLA, Beijing, China.
| |
Collapse
|
16
|
Sima CM, Buzilă ER, Trofin F, Păduraru D, Luncă C, Duhaniuc A, Dorneanu OS, Nastase EV. Emerging Strategies against Non-Typhoidal Salmonella: From Pathogenesis to Treatment. Curr Issues Mol Biol 2024; 46:7447-7472. [PMID: 39057083 PMCID: PMC11275306 DOI: 10.3390/cimb46070442] [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: 06/25/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Even with the intensive efforts by public health programs to control and prevent it, non-typhoidal Salmonella (NTS) infection remains an important public health challenge. It is responsible for approximately 150 million illnesses and 60,000 deaths worldwide annually. NTS infection poses significant risks with high rates of morbidity and mortality, leading to potential short- and long-term complications. There is growing concern among health authorities about the increasing incidence of antimicrobial resistance, with multidrug resistance totaling 22.6% in Europe, highlighting an urgent need for new therapeutic approaches. Our review aims to provide a comprehensive overview of NTS infection. We outline the molecular mechanisms involved in the pathogenesis of NTS infection, as well as the events leading to invasive NTS infection and the subsequent complications associated with it. Given the widespread implications of antimicrobial resistance, our review also presents the global landscape of resistance, including multidrug resistance, and delve into the underlying mechanisms driving this resistance. The rising rates of antibiotic resistance frequently lead to treatment failures, emphasizing the importance of investigating alternative therapeutic options. Therefore, in this review we also explore potential alternative therapies that could offer promising approaches to treating NTS infections.
Collapse
Affiliation(s)
- Cristina Mihaela Sima
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania;
| | - Elena Roxana Buzilă
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
- Iasi Regional Center for Public Health, National Institute of Public Health, 700465 Iasi, Romania
| | - Felicia Trofin
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
| | - Diana Păduraru
- “Dr. C.I. Parhon” Clinical Hospital, 700503 Iasi, Romania;
| | - Cătălina Luncă
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
- “Sf. Maria” Children Emergency Hospital, 700309 Iasi, Romania
| | - Alexandru Duhaniuc
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
- Iasi Regional Center for Public Health, National Institute of Public Health, 700465 Iasi, Romania
| | - Olivia Simona Dorneanu
- Department of Preventive Medicine and Interdisciplinarity—Microbiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.S.); (E.R.B.); (F.T.); (C.L.); (A.D.)
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania;
| | - Eduard Vasile Nastase
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania;
- Department of Internal Medicine II—Infectious Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
17
|
Turnbull JD, Dicks J, Adkin R, Dickinson A, Kaushal D, Semowo M, McGregor H, Alexander S. Notification of bacterial strains made available by the United Kingdom National Collection of Type Cultures in 2022. Access Microbiol 2024; 6:000756.v3. [PMID: 39130739 PMCID: PMC11316570 DOI: 10.1099/acmi.0.000756.v3] [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: 12/13/2023] [Accepted: 05/03/2024] [Indexed: 08/13/2024] Open
Abstract
Here, we report on the one hundred and twenty-five bacterial strains made available by the National Collection of Type Cultures in 2022 alongside a commentary on the strains, their provenance and significance.
Collapse
Affiliation(s)
- Jake David Turnbull
- The National Collection of Type Cultures, UK Health Security Agency, 61 Colindale Avenue, Colindale, London, NW9 5EQ, UK
| | - Jo Dicks
- The National Collection of Type Cultures, UK Health Security Agency, 61 Colindale Avenue, Colindale, London, NW9 5EQ, UK
| | - Rachael Adkin
- The National Collection of Type Cultures, UK Health Security Agency, 61 Colindale Avenue, Colindale, London, NW9 5EQ, UK
| | - Alexander Dickinson
- The National Collection of Type Cultures, UK Health Security Agency, 61 Colindale Avenue, Colindale, London, NW9 5EQ, UK
| | - Dorota Kaushal
- The National Collection of Type Cultures, UK Health Security Agency, 61 Colindale Avenue, Colindale, London, NW9 5EQ, UK
| | - Mojisola Semowo
- The National Collection of Type Cultures, UK Health Security Agency, 61 Colindale Avenue, Colindale, London, NW9 5EQ, UK
| | - The NCTC 2022 Depositors Cohort
- The NCTC 2022 Depositors Cohort consists of individuals who deposited strains into the NCTC and those instrumental in preparing the strains for submission to the NCTC. The NCTC 2022 Depositors Cohort are: Kathy Bernard (ex. National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada), Marie Chattaway (Gastrointestinal Bacteria Reference Unit, UK Health Security Agency, Colindale, London, UK), Ka Lip Chew (Department of Laboratory Medicine, National University Hospital, Singapore, Singapore), Rachel Gilroy (ex. Microbes in the Food Chain Group, Quadram Institute, Norwich Research Park, Norwich, UK), Harriet Gooch (John Innes Centre, Norwich, UK), Thi Thu Hao Van (Royal Melbourne Institute of Technology, Bundoora Campus, Bundoora, Victoria, Australia), Jane Hawkey (Monash Central Clinical School, The Burnet Institute, Melbourne, Australia), Jay Hinton (Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, UK), Katie Hopkins (Antimicrobial Resistance & Mechanisms Service, Antimicrobial Resistance and Healthcare Associated Infections Unit, UK Health Security Agency, Colindale, London, UK), Claire Jenkins (Gastrointestinal Bacteria Reference Unit, Public Health England, Colindale, London, UK), Rob Mariman (Rijksinstituut voor Volksgezondheid en Milieu (RIVM), the National Institute for Public Health and the Environment, Bilthoven, The Netherlands), Despoina Mavridou (Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA), Mark Pallen (Quadram Institute, Norwich Research Park, Norwich, UK), Gavin Paterson (Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, UK), Blanca Perez Sepulveda (Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, UK), Zeli Shen (Massachusetts Institute of Technology, Cambridge, Massachusetts, USA), Sho Shimada (Toho University, Faculty of Medicine, Omorinishi, Ota-ku, Tokyo), Sooyeon Song (Department of Animal Science, Jeonbuk National University, Backje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea), Dmitriy Volokhov (US Food and Drug Administration, Center for Biologics Evaluation and Research, Silver Spring, Maryland, USA), Thomas Wood (Pennsylvania State University, University Park, Pennsylvania, USA)
| | - Hannah McGregor
- The National Collection of Type Cultures, UK Health Security Agency, 61 Colindale Avenue, Colindale, London, NW9 5EQ, UK
| | - Sarah Alexander
- The National Collection of Type Cultures, UK Health Security Agency, 61 Colindale Avenue, Colindale, London, NW9 5EQ, UK
| |
Collapse
|
18
|
Osbelt L, Almási ÉDH, Wende M, Kienesberger S, Voltz A, Lesker TR, Muthukumarasamy U, Knischewski N, Nordmann E, Bielecka AA, Giralt-Zúñiga M, Kaganovitch E, Kühne C, Baier C, Pietsch M, Müsken M, Greweling-Pils MC, Breinbauer R, Flieger A, Schlüter D, Müller R, Erhardt M, Zechner EL, Strowig T. Klebsiella oxytoca inhibits Salmonella infection through multiple microbiota-context-dependent mechanisms. Nat Microbiol 2024; 9:1792-1811. [PMID: 38862602 PMCID: PMC11222139 DOI: 10.1038/s41564-024-01710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 04/22/2024] [Indexed: 06/13/2024]
Abstract
The Klebsiella oxytoca species complex is part of the human microbiome, especially during infancy and childhood. K. oxytoca species complex strains can produce enterotoxins, namely, tilimycin and tilivalline, while also contributing to colonization resistance (CR). The relationship between these seemingly contradictory roles is not well understood. Here, by coupling ex vivo assays with CRISPR-mutagenesis and various mouse models, we show that K. oxytoca provides CR against Salmonella Typhimurium. In vitro, the antimicrobial activity against various Salmonella strains depended on tilimycin production and was induced by various simple carbohydrates. In vivo, CR against Salmonella depended on toxin production in germ-free mice, while it was largely toxin-independent in mice with residual microbiota. This was linked to the relative levels of toxin-inducing carbohydrates in vivo. Finally, dulcitol utilization was essential for toxin-independent CR in gnotobiotic mice. Together, this demonstrates that nutrient availability is key to both toxin-dependent and substrate-driven competition between K. oxytoca and Salmonella.
Collapse
Affiliation(s)
- Lisa Osbelt
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
- ESF International Graduate School on Analysis, Imaging and Modelling of Neuronal and Inflammatory Processes, Otto-von-Guericke University, Magdeburg, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Éva D H Almási
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Marie Wende
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
- ESF International Graduate School on Analysis, Imaging and Modelling of Neuronal and Inflammatory Processes, Otto-von-Guericke University, Magdeburg, Germany
| | - Sabine Kienesberger
- Institute of Molecular Biosciences, University of Graz, BioTechMed-Graz, Graz, Austria
| | - Alexander Voltz
- Department Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Till R Lesker
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | | | - Nele Knischewski
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Elke Nordmann
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Agata A Bielecka
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - María Giralt-Zúñiga
- Institute for Biology-Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eugen Kaganovitch
- Institute for Biology-Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Caroline Kühne
- Institute for Biology-Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Claas Baier
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Michael Pietsch
- Division of Enteropathogenic Bacteria and Legionella (FG11)/National Reference Centre for Salmonella and other Bacterial Enteric Pathogens, Robert Koch Institute, Wernigerode, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Rolf Breinbauer
- BioTechMed-Graz, Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| | - Antje Flieger
- Division of Enteropathogenic Bacteria and Legionella (FG11)/National Reference Centre for Salmonella and other Bacterial Enteric Pathogens, Robert Koch Institute, Wernigerode, Germany
| | - Dirk Schlüter
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF),Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Rolf Müller
- Department Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- German Center for Infection Research (DZIF),Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Marc Erhardt
- Institute for Biology-Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
- Max Planck Unit for the Science of Pathogens, Berlin, Germany
| | - Ellen L Zechner
- Institute of Molecular Biosciences, University of Graz, BioTechMed-Graz, Graz, Austria
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
- German Center for Infection Research (DZIF),Partner Site Hannover-Braunschweig, Braunschweig, Germany.
- Center for Individualized Infection Medicine, Hannover, Germany.
| |
Collapse
|
19
|
Zhang Z, Hu M, Xu X, Lv C, Shi C. Dynamic antimicrobial resistance and phylogenomic structure of Salmonella Typhimurium from 2007 to 2019 in Shanghai, China. Microbiol Spectr 2024; 12:e0026224. [PMID: 38904374 PMCID: PMC11302141 DOI: 10.1128/spectrum.00262-24] [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: 01/28/2024] [Accepted: 05/11/2024] [Indexed: 06/22/2024] Open
Abstract
Salmonella enterica serovar Typhimurium is an important foodborne pathogen associated with human salmonellosis worldwide. A retrospective screening was performed to elucidate the prevalence, antimicrobial resistance, and phylogenomic characterization of this pathogen in Shanghai, China. S. Typhimurium isolates were selected from 2,211 serotyped Salmonella isolates collected during 2007-2019. Two hundred and seventy-seven S. Typhimurium isolates were detected in 15 of 16 districts in Shanghai. It was noted that 214 (77.3%) isolates were multi-drug resistant and 32 (11.6%) isolates were resistant to ciprofloxacin and 5 (1.8%) isolates were further resistant to ceftriaxone. Poisson generalized linear mixed model results showed that the multi-drug resistance (MDR) in 2017 and 2018 was significantly higher than that in 2010 (P<0.05), highlighting an increase in the risk of MDR. Phylogenetic results showed that a global data set of 401 sequenced S. Typhimurium isolates was classified into four clones (ST36, ST313, ST19, and ST34), which appeared in international clonal dissemination. The ST34 isolates from China fell into two clades, ST34C1 and ST34C2, the latter of which might originate from Shanghai, and then expanded nationally, accompanied by extended-spectrum β-lactamase gene blaCTX-M-14 and a mutation in quinolone resistance-determining region of the gyrA 87 site. Furthermore, blaCTX-M-14 linking to ISEcp1 upstream and ΔIS903B downstream was found in IncI (Gamma)-like plasmids, and the plasmid conjugation contributed to its horizontal transmission. To our knowledge, it is the first report of the epidemiological and phylogenetic characterization for S. Typhimurium including the emerged clade ST34C2 in Shanghai, warranting the necessity of surveillance for this high-risk pathogen. IMPORTANCE Our study uncovered a widespread distribution of Salmonella enterica serovar Typhimurium isolates in Shanghai accompanied by the increase in antimicrobial resistance (AMR) especially MDR during a 10-year period, which filled in the gap about a long period of continuous monitoring of AMR in this pathogen in Shanghai. Meanwhile, we identified a new clade ST34C2 of S. Typhimurium with the acquisition of IncI (Gamma)-like plasmids mediated by extended-spectrum β-lactamase gene blaCTX-M-14 as well as gyrA 87 mutation, which had not been reported before. It was noted that IncI (Gamma)-like plasmids were reported in S. Typhimurium for the first time and conjugation could accelerate the spread of antimicrobial resistance gene blaCTX-M-14. These findings on the epidemic, antimicrobial resistance, and phylogenomic characterization for S. Typhimurium provide valuable insights into its potential risk to public health and also the basis for AMR prevention and control strategies in Shanghai in the future.
Collapse
Affiliation(s)
- Zengfeng Zhang
- Department of Food Science & Technology, School of Agriculture and Biology, State Key Lab of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, China
| | - Mengjun Hu
- Department of Food Science & Technology, School of Agriculture and Biology, State Key Lab of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, China
| | - Xuebin Xu
- Laboratory of Microbiology, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Chao Lv
- Department of Animal Health and Food Safety, School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunlei Shi
- Department of Food Science & Technology, School of Agriculture and Biology, State Key Lab of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
20
|
Sun RY, Fang LX, Dai JJ, Chen KC, Ke BX, Sun J, Ke CW, Wai Chi Chan E, Liu YH, Chen S, Liao XP. Antimicrobial resistance and population genomics of emerging multidrug-resistant Salmonella 4,[5],12:i:- in Guangdong, China. mSystems 2024; 9:e0116423. [PMID: 38747582 PMCID: PMC11237462 DOI: 10.1128/msystems.01164-23] [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: 11/06/2023] [Accepted: 04/09/2024] [Indexed: 06/19/2024] Open
Abstract
Salmonella 4,[5],12:i:-, a monophasic variant of Salmonella Typhimurium, has emerged as a global cause of multidrug-resistant salmonellosis and has become endemic in many developing and developed countries, especially in China. Here, we have sequenced 352 clinical isolates in Guangdong, China, during 2009-2019 and performed a large-scale collection of Salmonella 4,[5],12:i:- with whole genome sequencing (WGS) data across the globe, to better understand the population structure, antimicrobial resistance (AMR) genomic characterization, and transmission routes of Salmonella 4,[5],12:i:- across Guangdong. Salmonella 4,[5],12:i:- strains showed broad genetic diversity; Guangdong isolates were found to be widely distributed among the global lineages. Of note, we identified the formation of a novel Guangdong clade (Bayesian analysis of population structure lineage 1 [BAPS1]) genetically diversified from the global isolates and likely emerged around 1990s. BAPS1 exhibits unique genomic features, including large pan-genome, decreased ciprofloxacin susceptibility due to mutation in gyrA and carriage of plasmid-mediated quinolone resistance (PMQR) genes, and the multidrug-resistant IncHI2 plasmid. Furthermore, high genetic similarity was found between strains collected from Guangdong, Europe, and North America, indicating the association with multiple introductions from overseas. These results suggested that global dissemination and local clonal expansion simultaneously occurred in Guangdong, China, and horizontally acquired resistance to first-line and last-line antimicrobials at local level, underlying emergences of extensive drug and pan-drug resistance. Our findings have increased the knowledge of global and local epidemics of Salmonella 4,[5],12:i:- in Guangdong, China, and provided a comprehensive baseline data set essential for future molecular surveillance.IMPORTANCESalmonella 4,[5],12:i:- has been regarded as the predominant pandemic serotype causing diarrheal diseases globally, while multidrug resistance (MDR) constitutes great public health concerns. This study provided a detailed and comprehensive genome-scale analysis of this important Salmonella serovar in the past decade in Guangdong, China. Our results revealed the complexity of two distinct transmission modes, namely global transmission and local expansion, circulating in Guangdong over a decade. Using phylogeography models, the origin of Salmonella 4,[5],12:i:- was predicted from two aspects, year and country, that is, Salmonella 4,[5],12:i:- emerged in 1983, and was introduced from the UK, and subsequently differentiated into the local endemic lineage circa 1991. Additionally, based on the pan-genome analysis, it was found that the gene accumulation rate in local endemic BAPS 1 lineage was higher than in other lineages, and the horizontal transmission of MDR IncHI2 plasmid associated with high resistance played a major role, which showed the potential threat to public health.
Collapse
Affiliation(s)
- Ruan-Yang Sun
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, Guangdong, China
| | - Liang-Xing Fang
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, China
| | - Jing-Jing Dai
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, Guangdong, China
| | - Kai-Chao Chen
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Bi-Xia Ke
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Jian Sun
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, China
| | - Chang-Wen Ke
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Edward Wai Chi Chan
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Ya-Hong Liu
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Sheng Chen
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Xiao-Ping Liao
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, China
| |
Collapse
|
21
|
Li P, Zhan L, Wang H, Yan Y, Jia M, Gao L, Sun Y, Zhu G, Chen Z. Prevalence and Antimicrobial Resistance Diversity of Salmonella Isolates in Jiaxing City, China. Antibiotics (Basel) 2024; 13:443. [PMID: 38786171 PMCID: PMC11117378 DOI: 10.3390/antibiotics13050443] [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: 04/12/2024] [Revised: 05/11/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024] Open
Abstract
Nontyphoidal Salmonella (NTS) is a cause of foodborne diarrheal diseases worldwide. Important emerging NTS serotypes that have spread as multidrug-resistant high-risk clones include S. Typhimurium monophasic variant and S. Kentucky. In this study, we isolated Salmonella in 5019 stool samples collected from patients with clinical diarrhea and 484 food samples. Antibiotic susceptibility testing and whole-genome sequencing were performed on positive strains. The detection rates of Salmonella among patients with diarrhea and food samples were 4.0% (200/5019) and 3.1% (15/484), respectively. These 215 Salmonella isolates comprised five main serotypes, namely S. Typhimurium monophasic variant, S. Typhimurium, S. London, S. Enteritidis, and S. Rissen, and were mainly resistant to ampicillin, tetracycline, chloramphenicol, and trimethoprim/sulfamethoxazole. The MDR rates of five major serotypes were 77.4%, 56.0%, 66.7%, 53.3%, and 80.0%, respectively. The most commonly acquired extended-spectrum β-lactamase-encoding genes were blaTEM-1B, blaOXA-10, and blaCTX-M-65. The S. Typhimurium monophasic variant strains from Jiaxing City belonged to a unique clone with broad antibiotic resistance. S. Kentucky isolates showed the highest drug resistance, and all were MDR strains. The discovery of high antibiotic resistance rates in this common foodborne pathogen is a growing concern; therefore, ongoing surveillance is crucial to effectively monitor this pathogen.
Collapse
Affiliation(s)
- Ping Li
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Center for Disease Control and Prevention, Jiaxing 314050, China; (P.L.)
| | - Li Zhan
- Institute of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China;
| | - Henghui Wang
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Center for Disease Control and Prevention, Jiaxing 314050, China; (P.L.)
| | - Yong Yan
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Center for Disease Control and Prevention, Jiaxing 314050, China; (P.L.)
| | - Miaomiao Jia
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Center for Disease Control and Prevention, Jiaxing 314050, China; (P.L.)
| | - Lei Gao
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Center for Disease Control and Prevention, Jiaxing 314050, China; (P.L.)
| | - Yangming Sun
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Center for Disease Control and Prevention, Jiaxing 314050, China; (P.L.)
| | - Guoying Zhu
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Center for Disease Control and Prevention, Jiaxing 314050, China; (P.L.)
| | - Zhongwen Chen
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Center for Disease Control and Prevention, Jiaxing 314050, China; (P.L.)
| |
Collapse
|
22
|
Nagasawa Y, Nakayama M, Kato Y, Ogawa Y, Aribam SD, Tsugami Y, Iwata T, Mikami O, Sugiyama A, Onishi M, Hayashi T, Eguchi M. A novel vaccine strategy using quick and easy conversion of bacterial pathogens to unnatural amino acid-auxotrophic suicide derivatives. Microbiol Spectr 2024; 12:e0355723. [PMID: 38385737 PMCID: PMC10986568 DOI: 10.1128/spectrum.03557-23] [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: 10/04/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Abstract
We propose a novel strategy for quick and easy preparation of suicide live vaccine candidates against bacterial pathogens. This method requires only the transformation of one or more plasmids carrying genes encoding for two types of biological devices, an unnatural amino acid (uAA) incorporation system and toxin-antitoxin systems in which translation of the antitoxins requires the uAA incorporation. Escherichia coli BL21-AI laboratory strains carrying the plasmids were viable in the presence of the uAA, whereas the free toxins killed these strains after the removal of the uAA. The survival time after uAA removal could be controlled by the choice of the uAA incorporation system and toxin-antitoxin systems. Multilayered toxin-antitoxin systems suppressed escape frequency to less than 1 escape per 109 generations in the best case. This conditional suicide system also worked in Salmonella enterica and E. coli clinical isolates. The S. enterica vaccine strains were attenuated with a >105 fold lethal dose. Serum IgG response and protection against the parental pathogenic strain were confirmed. In addition, the live E. coli vaccine strain was significantly more immunogenic and provided greater protection than a formalin-inactivated vaccine. The live E. coli vaccine was not detected after inoculation, presumably because the uAA is not present in the host animals or the natural environment. These results suggest that this strategy provides a novel way to rapidly produce safe and highly immunogenic live bacterial vaccine candidates. IMPORTANCE Live vaccines are the oldest vaccines with a history of more than 200 years. Due to their strong immunogenicity, live vaccines are still an important category of vaccines today. However, the development of live vaccines has been challenging due to the difficulties in achieving a balance between safety and immunogenicity. In recent decades, the frequent emergence of various new and old pathogens at risk of causing pandemics has highlighted the need for rapid vaccine development processes. We have pioneered the use of uAAs to control gene expression and to conditionally kill host bacteria as a biological containment system. This report proposes a quick and easy conversion of bacterial pathogens into live vaccine candidates using this containment system. The balance between safety and immunogenicity can be modulated by the selection of the genetic devices used. Moreover, the uAA-auxotrophy can prevent the vaccine from infecting other individuals or establishing the environment.
Collapse
Affiliation(s)
- Yuya Nagasawa
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Sapporo, Hokkaido, Japan
| | - Momoko Nakayama
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Yusuke Kato
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Yohsuke Ogawa
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Sapporo, Hokkaido, Japan
| | - Swarmistha Devi Aribam
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Yusaku Tsugami
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Sapporo, Hokkaido, Japan
| | - Taketoshi Iwata
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Osamu Mikami
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Sapporo, Hokkaido, Japan
| | - Aoi Sugiyama
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Sapporo, Hokkaido, Japan
| | - Megumi Onishi
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Sapporo, Hokkaido, Japan
| | - Tomohito Hayashi
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Sapporo, Hokkaido, Japan
| | - Masahiro Eguchi
- National Institute of Animal Health, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| |
Collapse
|
23
|
Kumwenda B, Canals R, Predeus AV, Zhu X, Kröger C, Pulford C, Wenner N, Lora LL, Li Y, Owen SV, Everett D, Hokamp K, Heyderman RS, Ashton PM, Gordon MA, Msefula CL, Hinton JCD. Salmonella enterica serovar Typhimurium ST313 sublineage 2.2 has emerged in Malawi with a characteristic gene expression signature and a fitness advantage. MICROLIFE 2024; 5:uqae005. [PMID: 38623411 PMCID: PMC11018118 DOI: 10.1093/femsml/uqae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/17/2024]
Abstract
Invasive non-typhoidal Salmonella (iNTS) disease is a serious bloodstream infection that targets immune-compromised individuals, and causes significant mortality in sub-Saharan Africa. Salmonella enterica serovar Typhimurium ST313 causes the majority of iNTS in Malawi. We performed an intensive comparative genomic analysis of 608 S. Typhimurium ST313 isolates dating between 1996 and 2018 from Blantyre, Malawi. We discovered that following the arrival of the well-characterized S. Typhimurium ST313 lineage 2 in 1999, two multidrug-resistant variants emerged in Malawi in 2006 and 2008, designated sublineages 2.2 and 2.3, respectively. The majority of S. Typhimurium isolates from human bloodstream infections in Malawi now belong to sublineages 2.2 or 2.3. To understand the emergence of the prevalent ST313 sublineage 2.2, we studied two representative strains, D23580 (lineage 2) and D37712 (sublineage 2.2). The chromosome of ST313 lineage 2 and sublineage 2.2 only differed by 29 SNPs/small indels and a 3 kb deletion of a Gifsy-2 prophage region including the sseI pseudogene. Lineage 2 and sublineage 2.2 had distinctive plasmid profiles. The transcriptome was investigated in 15 infection-relevant in vitro conditions and within macrophages. During growth in physiological conditions that do not usually trigger S. Typhimurium SPI2 gene expression, the SPI2 genes of D37712 were transcriptionally active. We identified down-regulation of flagellar genes in D37712 compared with D23580. Following phenotypic confirmation of transcriptomic differences, we discovered that sublineage 2.2 had increased fitness compared with lineage 2 during mixed growth in minimal media. We speculate that this competitive advantage is contributing to the emergence of sublineage 2.2 in Malawi.
Collapse
Affiliation(s)
- Benjamin Kumwenda
- School of Life Sciences and Allied Health Professions, Kamuzu University of Health Sciences Blantyre, Blantyre, 265, Malawi
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
- Malawi–Liverpool–Wellcome Programme, Blantyre, 3, Malawi
| | - Rocío Canals
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - Alexander V Predeus
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - Xiaojun Zhu
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - Carsten Kröger
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, D02 PN40, Ireland
| | - Caisey Pulford
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - Nicolas Wenner
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - Lizeth Lacharme Lora
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - Yan Li
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - Siân V Owen
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - Dean Everett
- Department of Public Health and Epidemiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, P.O. Box 127788, United Arab Emirates
| | - Karsten Hokamp
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, Dublin, D02 PN40, Ireland
| | - Robert S Heyderman
- Malawi–Liverpool–Wellcome Programme, Blantyre, 3, Malawi
- Research Department of Infection, Division of Infection & Immunity, University College London, London, WC1E 6BT, United Kingdom
| | | | - Melita A Gordon
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
- Malawi–Liverpool–Wellcome Programme, Blantyre, 3, Malawi
| | - Chisomo L Msefula
- School of Life Sciences and Allied Health Professions, Kamuzu University of Health Sciences Blantyre, Blantyre, 265, Malawi
- Malawi–Liverpool–Wellcome Programme, Blantyre, 3, Malawi
| | - Jay C D Hinton
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| |
Collapse
|
24
|
Baqar S, Bonavia A, Louis Bourgeois A, Campo JJ, Clifford A, Hanevik K, Hasso-Agopsowicz M, Hausdorff W, Kaminski R, MacLennan CA, Mantis N, Martin LB, Omore R, Pasetti M, Pavlinac P, Phalipon A, Poly F, Porter C, Ramasamy MN, Rogawski McQuade ET, Sztein MB, Walker R. The 2022 Vaccines Against Shigella and Enterotoxigenic Escherichia coli (VASE) Conference: Summary of breakout workshops. Vaccine 2024; 42:1445-1453. [PMID: 38036392 PMCID: PMC10953702 DOI: 10.1016/j.vaccine.2023.11.045] [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: 04/25/2023] [Revised: 08/25/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023]
Abstract
The global public health nonprofit organization PATH hosted the third Vaccines Against Shigella and Enterotoxigenic Escherichia coli (VASE) Conference in Washington, DC, from November 29 to December 1, 2022. This international gathering focused on cutting-edge research related to the development of vaccines against neglected diarrheal pathogens including Shigella, enterotoxigenic Escherichia coli (ETEC), Campylobacter, and non-typhoidal Salmonella. In addition to the conference's plenary content, the agenda featured ten breakout workshops on topics of importance to the enteric vaccine field. This unique aspect of VASE Conferences allows focused groups of attendees to engage in in-depth discussions on subjects of interest to the enteric vaccine development community. In 2022, the workshops covered a range of topics. Two focused on the public health value of enteric vaccines, with one examining how to translate evidence into policy and the other on the value proposition of potential combination vaccines against bacterial enteric pathogens. Two more workshops explored new tools for the development and evaluation of vaccines, with the first on integrating antigen/antibody technologies for mucosal vaccine and immunoprophylactic development, and the second on adjuvants specifically for Shigella vaccines for children in low- and middle-income countries. Another pair of workshops covered the status of vaccines against two emerging enteric pathogens, Campylobacter and invasive non-typhoidal Salmonella. The remaining four workshops examined the assessment of vaccine impact on acute and long-term morbidity. These included discussions on the nature and severity of intestinal inflammation; cellular immunity and immunological memory in ETEC and Shigella infections; clinical and microbiologic endpoints for Shigella vaccine efficacy studies in children; and intricacies of protective immunity to enteric pathogens. This article provides a brief summary of the presentations and discussions at each workshop in order to share these sessions with the broader enteric vaccine field.
Collapse
Affiliation(s)
| | - Aurelio Bonavia
- Bill & Melinda Gates Medical Research Institute, United States
| | | | | | | | - Kurt Hanevik
- University of Bergen, Norway; Norwegian National Advisory Unit for Tropical Infectious Diseases, Medical Department, Haukeland University Hospital, Norway
| | | | - William Hausdorff
- PATH, United States; Faculty of Medicine, Université Libre de Bruxelles, Belgium
| | | | - Calman A MacLennan
- Enteric and Diarrheal Diseases, Bill & Melinda Gates Foundation, United Kingdom; The Jenner Institute, United Kingdom
| | - Nicholas Mantis
- Wadsworth Center, New York State Department of Health, United States
| | | | - Richard Omore
- Kenya Medical Research Institute Center for Global Health Research, Kenya
| | | | | | | | | | - Chad Porter
- Naval Medical Research Command, United States
| | | | | | | | | |
Collapse
|
25
|
Ke Y, Teng L, Zhu Z, Lu W, Liu W, Zhou H, Yu Q, Ye L, Zhu P, Zhao G, Yue M. Genomic investigation and nationwide tracking of pediatric invasive nontyphoidal Salmonella in China. MLIFE 2024; 3:156-160. [PMID: 38827503 PMCID: PMC11139200 DOI: 10.1002/mlf2.12117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/29/2023] [Accepted: 02/05/2024] [Indexed: 06/04/2024]
Abstract
Invasive nontyphoidal Salmonella (iNTS) causes significant concern with ~15% morbidity, affecting populations mainly in African countries. However, iNTS infections among the Chinese pediatric population remain largely unknown. Here, we conducted a genomic investigation to study pediatric iNTS infections in a Chinese hospital. iNTS isolates accounted for 15.2% (18/119) of all nontyphoidal Salmonella (NTS) strains. Compared to non-iNTS isolates, iNTS isolates harbored a lower prevalence of antimicrobial-resistant genes of fluoroquinolones and β-lactams, as well as disinfectant determinants and plasmids, but carried a significantly higher prevalence of cdtB, faeCDE, and tcpC genes. Importantly, we detected an emerging serovar Goldcoast as the predominant iNTS serovar locally. By integrating 320 global Goldcoast genomes based on the One Health samplings, we conducted nationwide phylogenomic tracking and detected repeated human-to-human transmission events among iNTS cases caused by an underestimated serovar Goldcoast. Together, our exploratory genomic approach highlights a new trend in pediatric iNTS infections.
Collapse
Affiliation(s)
- Yefang Ke
- Department of Clinical LaboratoryNingbo Women and Children's HospitalNingboChina
| | - Lin Teng
- Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Zhe Zhu
- Department of Blood TransfusionNingbo No. 2 HospitalNingboChina
| | - Wenbo Lu
- Department of Clinical LaboratoryNingbo Women and Children's HospitalNingboChina
| | - Wenyuan Liu
- Department of Clinical LaboratoryNingbo Women and Children's HospitalNingboChina
| | - Haiyang Zhou
- Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Qi Yu
- Office of ScreeningNingbo Women and Children's HospitalNingboChina
| | - Lina Ye
- Department of Clinical LaboratoryNingbo Women and Children's HospitalNingboChina
| | - Pan Zhu
- Neonatal Intensive Care UnitNingbo Women and Children's HospitalNingboChina
| | - Guoping Zhao
- School of Life Science, Hangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhouChina
- CAS Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological SciencesChinese Academy of SciencesShanghaiChina
- Department of Microbiology and Microbial Engineering, School of Life SciencesFudan UniversityShanghaiChina
| | - Min Yue
- Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
- School of Life Science, Hangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhouChina
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
- Hainan Institute of Zhejiang UniversitySanyaChina
| |
Collapse
|
26
|
Nhu NTK, Rahman MA, Goh KGK, Kim SJ, Phan MD, Peters KM, Alvarez-Fraga L, Hancock SJ, Ravi C, Kidd TJ, Sullivan MJ, Irvine KM, Beatson SA, Sweet MJ, Irwin AD, Vukovic J, Ulett GC, Hasnain SZ, Schembri MA. A convergent evolutionary pathway attenuating cellulose production drives enhanced virulence of some bacteria. Nat Commun 2024; 15:1441. [PMID: 38383596 PMCID: PMC10881479 DOI: 10.1038/s41467-024-45176-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
Bacteria adapt to selective pressure in their immediate environment in multiple ways. One mechanism involves the acquisition of independent mutations that disable or modify a key pathway, providing a signature of adaptation via convergent evolution. Extra-intestinal pathogenic Escherichia coli (ExPEC) belonging to sequence type 95 (ST95) represent a global clone frequently associated with severe human infections including acute pyelonephritis, sepsis, and neonatal meningitis. Here, we analysed a publicly available dataset of 613 ST95 genomes and identified a series of loss-of-function mutations that disrupt cellulose production or its modification in 55.3% of strains. We show the inability to produce cellulose significantly enhances ST95 invasive infection in a rat model of neonatal meningitis, leading to the disruption of intestinal barrier integrity in newborn pups and enhanced dissemination to the liver, spleen and brain. Consistent with these observations, disruption of cellulose production in ST95 augmented innate immune signalling and tissue neutrophil infiltration in a mouse model of urinary tract infection. Mutations that disrupt cellulose production were also identified in other virulent ExPEC STs, Shigella and Salmonella, suggesting a correlative association with many Enterobacteriaceae that cause severe human infection. Together, our findings provide an explanation for the emergence of hypervirulent Enterobacteriaceae clones.
Collapse
Affiliation(s)
- Nguyen Thi Khanh Nhu
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - M Arifur Rahman
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
- Immunopathology Group, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
- QIMR Berghofer Medical Research Institute, Brisbane QLD, Australia
| | - Kelvin G K Goh
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Seung Jae Kim
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Minh-Duy Phan
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Kate M Peters
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Laura Alvarez-Fraga
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
- INRAE, Univ Montpellier, LBE, 102 Avenue des Etangs, Narbonne, 11100, France
| | - Steven J Hancock
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Chitra Ravi
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Timothy J Kidd
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
- Central Microbiology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Matthew J Sullivan
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
- School of Biological Sciences, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Katharine M Irvine
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
- Immunopathology Group, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Scott A Beatson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Adam D Irwin
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
- University of Queensland Centre for Clinical Research, Brisbane, Australia
- Queensland Children's Hospital, Brisbane, Australia
| | - Jana Vukovic
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| | - Glen C Ulett
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia.
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.
| | - Sumaira Z Hasnain
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.
- Immunopathology Group, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia.
| | - Mark A Schembri
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia.
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
27
|
Lamichhane B, Mawad AMM, Saleh M, Kelley WG, Harrington PJ, Lovestad CW, Amezcua J, Sarhan MM, El Zowalaty ME, Ramadan H, Morgan M, Helmy YA. Salmonellosis: An Overview of Epidemiology, Pathogenesis, and Innovative Approaches to Mitigate the Antimicrobial Resistant Infections. Antibiotics (Basel) 2024; 13:76. [PMID: 38247636 PMCID: PMC10812683 DOI: 10.3390/antibiotics13010076] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/24/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Salmonella is a major foodborne pathogen and a leading cause of gastroenteritis in humans and animals. Salmonella is highly pathogenic and encompasses more than 2600 characterized serovars. The transmission of Salmonella to humans occurs through the farm-to-fork continuum and is commonly linked to the consumption of animal-derived food products. Among these sources, poultry and poultry products are primary contributors, followed by beef, pork, fish, and non-animal-derived food such as fruits and vegetables. While antibiotics constitute the primary treatment for salmonellosis, the emergence of antibiotic resistance and the rise of multidrug-resistant (MDR) Salmonella strains have highlighted the urgency of developing antibiotic alternatives. Effective infection management necessitates a comprehensive understanding of the pathogen's epidemiology and transmission dynamics. Therefore, this comprehensive review focuses on the epidemiology, sources of infection, risk factors, transmission dynamics, and the host range of Salmonella serotypes. This review also investigates the disease characteristics observed in both humans and animals, antibiotic resistance, pathogenesis, and potential strategies for treatment and control of salmonellosis, emphasizing the most recent antibiotic-alternative approaches for infection control.
Collapse
Affiliation(s)
- Bibek Lamichhane
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Asmaa M. M. Mawad
- Botany and Microbiology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Mohamed Saleh
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - William G. Kelley
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Patrick J. Harrington
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Cayenne W. Lovestad
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Jessica Amezcua
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Mohamed M. Sarhan
- Faculty of Pharmacy, King Salman International University (KSIU), Ras Sudr 8744304, Egypt
| | - Mohamed E. El Zowalaty
- Veterinary Medicine and Food Security Research Group, Medical Laboratory Sciences Program, Faculty of Health Sciences, Abu Dhabi Women’s Campus, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Hazem Ramadan
- Hygiene and Zoonoses Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Melissa Morgan
- Department of Animal and Food Sciences, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Yosra A. Helmy
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| |
Collapse
|
28
|
Smith C, Smith E, Rydlova A, Varro R, Hinton JCD, Gordon MA, Choy RKM, Liu X, Pollard AJ, Chiu C, Cooke GS, Gibani MM. Protocol for the challenge non-typhoidal Salmonella (CHANTS) study: a first-in-human, in-patient, double-blind, randomised, safety and dose-escalation controlled human infection model in the UK. BMJ Open 2024; 14:e076477. [PMID: 38199617 PMCID: PMC10806722 DOI: 10.1136/bmjopen-2023-076477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
INTRODUCTION Invasive non-typhoidal Salmonella (iNTS) serovars are a major cause of community-acquired bloodstream infections in sub-Saharan Africa (SSA). In this setting, Salmonella enterica serovar Typhimurium accounts for two-thirds of infections and is associated with an estimated case fatality rate of 15%-20%. Several iNTS vaccine candidates are in early-stage assessment which-if found effective-would provide a valuable public health tool to reduce iNTS disease burden. The CHANTS study aims to develop a first-in-human Salmonella Typhimurium controlled human infection model, which can act as a platform for future vaccine evaluation, in addition to providing novel insights into iNTS disease pathogenesis. METHODS AND ANALYSIS This double-blind, safety and dose-escalation study will randomise 40-80 healthy UK participants aged 18-50 to receive oral challenge with one of two strains of S. Typhimurium belonging to the ST19 (strain 4/74) or ST313 (strain D23580) lineages. 4/74 is a global strain often associated with diarrhoeal illness predominantly in high-income settings, while D23580 is an archetypal strain representing invasive disease-causing isolates found in SSA. The primary objective is to determine the minimum infectious dose (colony-forming unit) required for 60%-75% of participants to develop clinical or microbiological features of systemic salmonellosis. Secondary endpoints are to describe and compare the clinical, microbiological and immunological responses following challenge. Dose escalation or de-escalation will be undertaken by continual-reassessment methodology and limited within prespecified safety thresholds. Exploratory objectives are to describe mechanisms of iNTS virulence, identify putative immune correlates of protection and describe host-pathogen interactions in response to infection. ETHICS AND DISSEMINATION Ethical approval has been obtained from the NHS Health Research Authority (London-Fulham Research Ethics Committee 21/PR/0051; IRAS Project ID 301659). The study findings will be disseminated in international peer-reviewed journals and presented at national/international stakeholder meetings. Study outcome summaries will be provided to both funders and participants. TRIAL REGISTRATION NUMBER NCT05870150.
Collapse
Affiliation(s)
- Christopher Smith
- Department of Infectious Disease, Imperial College London, London, UK
| | - Emma Smith
- Department of Infectious Disease, Imperial College London, London, UK
| | - Anna Rydlova
- Department of Infectious Disease, Imperial College London, London, UK
| | - Robert Varro
- Department of Infectious Disease, Imperial College London, London, UK
| | - Jay C D Hinton
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Melita A Gordon
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Malawi Liverpool Wellcome Trust Clinical Research Programme, Kamuzu University of Health Sciences, Blantyre, Southern Region, Malawi
| | | | - Xinxue Liu
- Oxford Vaccine Group, Department of Paediatrics, Oxford University, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, Oxford University, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Graham S Cooke
- Department of Infectious Disease, Imperial College London, London, UK
| | - Malick M Gibani
- Department of Infectious Disease, Imperial College London, London, UK
| |
Collapse
|
29
|
Ormsby MJ, White HL, Metcalf R, Oliver DM, Feasey NA, Quilliam RS. Enduring pathogenicity of African strains of Salmonella on plastics and glass in simulated peri-urban environmental waste piles. JOURNAL OF HAZARDOUS MATERIALS 2024; 461:132439. [PMID: 37734312 DOI: 10.1016/j.jhazmat.2023.132439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/23/2023]
Abstract
In low- and middle-income countries, plastic has become a major constituent of landfills and urban dump sites. Environmental plastic pollution can also provide a novel surface for the formation of microbial biofilm, which often includes pathogenic bacteria and viruses. Here, under conditions simulating a peri-urban waste pile typical of an African informal settlement, we aimed to determine if pathogenic Salmonella spp. can retain their virulence following a prolonged period of desiccation on the surfaces of environmental plastic and glass. We show that clinically (and environmentally) relevant strains of Salmonella including S. Enteritidis, S. Typhimurium and S. Typhi can persist on plastic and glass for at least 28-days and that temperature (which increases with the depth of an urban waste pile) is a key determinant of this survival. All three strains of Salmonella retained their pathogenicity (determined by using a Galleria mellonella model of infection) following their recovery from the plastisphere indicating that plastics in the environment can act as reservoirs for human pathogens and could facilitate their persistence for extended periods of time. Pathogens colonising environmental plastic waste therefore pose a heightened public health risk, particularly in areas where people are frequently exposed to plastic pollution.
Collapse
Affiliation(s)
- Michael J Ormsby
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK.
| | - Hannah L White
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| | - Rebecca Metcalf
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| | - David M Oliver
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| | - Nicholas A Feasey
- Malawi-Liverpool Wellcome Research Programme, Blantyre, Malawi; Kamuzu University of Health Sciences, Blantyre, Malawi; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Richard S Quilliam
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| |
Collapse
|
30
|
Yang J, Barrila J, Nauman EA, Nydam SD, Yang S, Park J, Gutierrez-Jensen AD, Castro CL, Ott CM, Buss K, Steel J, Zakrajsek AD, Schuff MM, Nickerson CA. Incremental increases in physiological fluid shear progressively alter pathogenic phenotypes and gene expression in multidrug resistant Salmonella. Gut Microbes 2024; 16:2357767. [PMID: 38783686 PMCID: PMC11135960 DOI: 10.1080/19490976.2024.2357767] [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: 02/20/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
The ability of bacteria to sense and respond to mechanical forces has important implications for pathogens during infection, as they experience wide fluid shear fluctuations in the host. However, little is known about how mechanical forces encountered in the infected host drive microbial pathogenesis. Herein, we combined mathematical modeling with hydrodynamic bacterial culture to profile transcriptomic and pathogenesis-related phenotypes of multidrug resistant S. Typhimurium (ST313 D23580) under different fluid shear conditions relevant to its transition from the intestinal tract to the bloodstream. We report that D23580 exhibited incremental changes in transcriptomic profiles that correlated with its pathogenic phenotypes in response to these progressive increases in fluid shear. This is the first demonstration that incremental changes in fluid shear forces alter stress responses and gene expression in any ST313 strain and offers mechanistic insight into how forces encountered by bacteria during infection might impact their disease-causing ability in unexpected ways.
Collapse
Affiliation(s)
- Jiseon Yang
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Jennifer Barrila
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA
| | - Eric A. Nauman
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| | - Seth D. Nydam
- Department of Animal Care & Technologies, Arizona State University, Tempe, AZ, USA
| | - Shanshan Yang
- Bioinformatics Core Facility, Bioscience, Knowledge Enterprise, Arizona State University, Tempe, AZ, USA
| | - Jin Park
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA
| | - Ami D. Gutierrez-Jensen
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Christian L. Castro
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- JES Tech, Houston, TX, USA
| | - C. Mark Ott
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX, USA
| | - Kristina Buss
- Bioinformatics Core Facility, Bioscience, Knowledge Enterprise, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA
| | - Jason Steel
- Bioinformatics Core Facility, Bioscience, Knowledge Enterprise, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA
| | - Anne D. Zakrajsek
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| | - Mary M. Schuff
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| | - Cheryl A. Nickerson
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
31
|
Fenske GJ, Pouzou JG, Pouillot R, Taylor DD, Costard S, Zagmutt FJ. The genomic and epidemiological virulence patterns of Salmonella enterica serovars in the United States. PLoS One 2023; 18:e0294624. [PMID: 38051743 DOI: 10.1371/journal.pone.0294624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
The serovars of Salmonella enterica display dramatic differences in pathogenesis and host preferences. We developed a process (patent pending) for grouping Salmonella isolates and serovars by their public health risk. We collated a curated set of 12,337 S. enterica isolate genomes from human, beef, and bovine sources in the US. After annotating a virulence gene catalog for each isolate, we used unsupervised random forest methods to estimate the proximity (similarity) between isolates based upon the genomic presentation of putative virulence traits We then grouped isolates (virulence clusters) using hierarchical clustering (Ward's method), used non-parametric bootstrapping to assess cluster stability, and externally validated the clusters against epidemiological virulence measures from FoodNet, the National Outbreak Reporting System (NORS), and US federal sampling of beef products. We identified five stable virulence clusters of S. enterica serovars. Cluster 1 (higher virulence) serovars yielded an annual incidence rate of domestically acquired sporadic cases roughly one and a half times higher than the other four clusters combined (Clusters 2-5, lower virulence). Compared to other clusters, cluster 1 also had a higher proportion of infections leading to hospitalization and was implicated in more foodborne and beef-associated outbreaks, despite being isolated at a similar frequency from beef products as other clusters. We also identified subpopulations within 11 serovars. Remarkably, we found S. Infantis and S. Typhimurium subpopulations that significantly differed in genome length and clinical case presentation. Further, we found that the presence of the pESI plasmid accounted for the genome length differences between the S. Infantis subpopulations. Our results show that S. enterica strains associated with highest incidence of human infections share a common virulence repertoire. This work could be updated regularly and used in combination with foodborne surveillance information to prioritize serovars of public health concern.
Collapse
Affiliation(s)
- Gavin J Fenske
- EpiX Analytics, Fort Collins, Colorado, United States of America
| | - Jane G Pouzou
- EpiX Analytics, Fort Collins, Colorado, United States of America
| | - Régis Pouillot
- EpiX Analytics, Fort Collins, Colorado, United States of America
| | - Daniel D Taylor
- EpiX Analytics, Fort Collins, Colorado, United States of America
| | - Solenne Costard
- EpiX Analytics, Fort Collins, Colorado, United States of America
| | | |
Collapse
|
32
|
Hajra D, Nair AV, Chakravortty D. Decoding the invasive nature of a tropical pathogen of concern: The invasive non-Typhoidal Salmonella strains causing host-restricted extraintestinal infections worldwide. Microbiol Res 2023; 277:127488. [PMID: 37716125 DOI: 10.1016/j.micres.2023.127488] [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: 07/28/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/18/2023]
Abstract
Invasive-Non-Typhoidal Salmonella (iNTS) are the major cause of health concern in the low-income, under-developed nations in Africa and Asia that lack proper sanitation facilities. Around 5% of the NTS cases give rise to invasive, extraintestinal diseases leading to focal infections like osteomyelitis, meningitis, osteoarthritis, endocarditis and neonatal sepsis. iNTS serovars like S. Typhimurium, S. Enteritidis, S. Dublin, S. Choleraesuis show a greater propensity to become invasive than others which hints at the genetic basis of their emergence. The major risk factors attributing to the invasive diseases include immune-compromised individuals having co-infection with malaria or HIV, or suffering from malnutrition. The rampant use of antibiotics leading to the emergence of multi-drug resistant strains poses a great challenge in disease management. An extensive understanding of the iNTS pathogenesis and its epidemiology will open up avenues for the development of new vaccination and therapeutic strategies to restrict the spread of this neglected disease.
Collapse
Affiliation(s)
- Dipasree Hajra
- Department of Microbiology & Cell Biology, Indian Institute of Science, India
| | - Abhilash Vijay Nair
- Department of Microbiology & Cell Biology, Indian Institute of Science, India
| | | |
Collapse
|
33
|
Martins IM, Seribelli AA, Machado Ribeiro TR, da Silva P, Lustri BC, Hernandes RT, Falcão JP, Moreira CG. Invasive non-typhoidal Salmonella (iNTS) aminoglycoside-resistant ST313 isolates feature unique pathogenic mechanisms to reach the bloodstream. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 116:105519. [PMID: 37890808 DOI: 10.1016/j.meegid.2023.105519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
Invasive non-typhoidal Salmonella (iNTS) from the clonal type ST313 (S. Typhimurium ST313) is the primary cause of invasive salmonellosis in Africa. Recently, in Brazil, iNTS ST313 strains have been isolated from different sources, but there is a lack of understanding of the mechanisms behind how these gut bacteria can break the gut barrier and reach the patient's bloodstream. Here, we compare 13 strains of S. Typhimurium ST313, previously unreported isolates, from human blood cultures, investigating aspects of virulence and mechanisms of resistance. Initially, RNAseq analyses between ST13-blood isolate and SL1344 (ST19) prototype revealed 15 upregulated genes directly related to cellular invasion and replication, such as sopD2, sifB, and pipB. Limited information is available about S. Typhimurium ST313 pathogenesis and epidemiology, especially related to the global distribution of strains. Herein, the correlation of strains isolated from different sources in Brazil was employed to compare clinical and non-clinical isolates, a total of 22 genomes were studied by single nucleotide polymorphism (SNPs). The epidemiological analysis of 22 genomes of S. Typhimurium ST313 strains grouped them into three distinct clusters (A, B, and C) by SNP analysis, where cluster A comprised five, group B six, and group C 11. The 13 clinical blood isolates were all resistant to streptomycin, 92.3% of strains were resistant to ampicillin and 15.39% were resistant to kanamycin. The resistance genes acrA, acrB, mdtK, emrB, emrR, mdsA, and mdsB related to the production of efflux pumps were detected in all (100%) strains studied, similar to pathogenic traits investigated. In conclusion, we evidenced that S. Typhimurium ST313 strains isolated in Brazil have unique epidemiology. The elevated frequencies of virulence genes such as sseJ, sopD2, and pipB are a major concern in these Brazilian isolates, showing a higher pathogenic potential.
Collapse
Affiliation(s)
- Isabela Mancini Martins
- Faculdade de Ciências Farmacêuticas de Araraquara, Universidade Estadual Paulista- UNESP- Departamento de Ciências Biológicas, Araraquara, SP, Brazil
| | - Amanda Aparecida Seribelli
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo- USP, Ribeirão Preto, SP, Brazil
| | - Tamara R Machado Ribeiro
- Faculdade de Ciências Farmacêuticas de Araraquara, Universidade Estadual Paulista- UNESP- Departamento de Ciências Biológicas, Araraquara, SP, Brazil
| | - Patrick da Silva
- Faculdade de Ciências Farmacêuticas de Araraquara, Universidade Estadual Paulista- UNESP- Departamento de Ciências Biológicas, Araraquara, SP, Brazil
| | - Bruna Cardinali Lustri
- Faculdade de Ciências Farmacêuticas de Araraquara, Universidade Estadual Paulista- UNESP- Departamento de Ciências Biológicas, Araraquara, SP, Brazil
| | - Rodrigo T Hernandes
- Instituto de Biociências, Universidade Estadual Paulista- UNESP, Botucatu, SP, Brazil
| | - Juliana Pfrimer Falcão
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo- USP, Ribeirão Preto, SP, Brazil.
| | - Cristiano Gallina Moreira
- Faculdade de Ciências Farmacêuticas de Araraquara, Universidade Estadual Paulista- UNESP- Departamento de Ciências Biológicas, Araraquara, SP, Brazil; Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
34
|
Zhong L, Zhang M, Sun L, Yang Y, Wang B, Yang H, Shen Q, Xia Y, Cui J, Hang H, Ren Y, Pang B, Deng X, Zhan Y, Li H, Zhou Z. Distributed genotyping and clustering of Neisseria strains reveal continual emergence of epidemic meningococcus over a century. Nat Commun 2023; 14:7706. [PMID: 38001084 PMCID: PMC10673917 DOI: 10.1038/s41467-023-43528-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Core genome multilocus sequence typing (cgMLST) is commonly used to classify bacterial strains into different types, for taxonomical and epidemiological applications. However, cgMLST schemes require central databases for the nomenclature of new alleles and sequence types, which must be synchronized worldwide and involve increasingly intensive calculation and storage demands. Here, we describe a distributed cgMLST (dcgMLST) scheme that does not require a central database of allelic sequences and apply it to study evolutionary patterns of epidemic and endemic strains of the genus Neisseria. We classify 69,994 worldwide Neisseria strains into multi-level clusters that assign species, lineages, and local disease outbreaks. We divide Neisseria meningitidis into 168 endemic lineages and three epidemic lineages responsible for at least 9 epidemics in the past century. According to our analyses, the epidemic and endemic lineages experienced very different population dynamics in the past 100 years. Epidemic lineages repetitively emerged from endemic lineages, disseminated worldwide, and apparently disappeared rapidly afterward. We propose a stepwise model for the evolutionary trajectory of epidemic lineages in Neisseria, and expect that the development of similar dcgMLST schemes will facilitate epidemiological studies of other bacterial pathogens.
Collapse
Affiliation(s)
- Ling Zhong
- Pasteurien College, Suzhou Medical College, Soochow University, Suzhou, 215123, China
- Key Laboratory of Alkene-Carbon Fibers-Based Technology & Application for Detection of Major Infectious Diseases, Soochow University, Suzhou, 215123, China
| | - Menghan Zhang
- Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China
| | - Libing Sun
- Department of Pathology, East District of Suzhou Municipal Hospital, Suzhou, 215000, China
| | - Yu Yang
- Pasteurien College, Suzhou Medical College, Soochow University, Suzhou, 215123, China
| | - Bo Wang
- Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China
| | - Haibing Yang
- Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China
| | - Qiang Shen
- Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China
| | - Yu Xia
- Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China
| | - Jiarui Cui
- Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China
| | - Hui Hang
- Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China
| | - Yi Ren
- Iotabiome Biotechnology Inc, Suzhou, 215000, China
| | - Bo Pang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiangyu Deng
- Center for Food Safety, University of Georgia, Griffin, GA, USA
| | - Yahui Zhan
- Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China.
| | - Heng Li
- Pasteurien College, Suzhou Medical College, Soochow University, Suzhou, 215123, China.
- Key Laboratory of Alkene-Carbon Fibers-Based Technology & Application for Detection of Major Infectious Diseases, Soochow University, Suzhou, 215123, China.
- Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Soochow University, Suzhou, 215123, China.
| | - Zhemin Zhou
- Pasteurien College, Suzhou Medical College, Soochow University, Suzhou, 215123, China.
- Key Laboratory of Alkene-Carbon Fibers-Based Technology & Application for Detection of Major Infectious Diseases, Soochow University, Suzhou, 215123, China.
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| |
Collapse
|
35
|
Stenhouse GE, Keddy KH, Bengtsson RJ, Hall N, Smith AM, Thomas J, Iturriza-Gómara M, Baker KS. The genomic epidemiology of shigellosis in South Africa. Nat Commun 2023; 14:7715. [PMID: 38001075 PMCID: PMC10673971 DOI: 10.1038/s41467-023-43345-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Shigellosis, a leading cause of diarrhoeal mortality and morbidity globally, predominantly affects children under five years of age living in low- and middle-income countries. While whole genome sequence analysis (WGSA) has been effectively used to further our understanding of shigellosis epidemiology, antimicrobial resistance, and transmission, it has been under-utilised in sub-Saharan Africa. In this study, we applied WGSA to large sub-sample of surveillance isolates from South Africa, collected from 2011 to 2015, focussing on Shigella flexneri 2a and Shigella sonnei. We find each serotype is epidemiologically distinct. The four identified S. flexneri 2a clusters having distinct geographical distributions, and antimicrobial resistance (AMR) and virulence profiles, while the four sub-Clades of S. sonnei varied in virulence plasmid retention. Our results support serotype specific lifestyles as a driver for epidemiological differences, show AMR is not required for epidemiological success in S. flexneri, and that the HIV epidemic may have promoted Shigella population expansion.
Collapse
Affiliation(s)
- George E Stenhouse
- Clinical Infection, Microbiology, and Immunology, University of Liverpool, Liverpool, UK.
| | | | - Rebecca J Bengtsson
- Clinical Infection, Microbiology, and Immunology, University of Liverpool, Liverpool, UK
| | - Neil Hall
- Earlham Institute, Norwich Research Park, NR4 7UZ, Norwich, UK
| | - Anthony M Smith
- Centre for Enteric Diseases, National Institute for Communicable Diseases (NICD), Division of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Juno Thomas
- Centre for Enteric Diseases, National Institute for Communicable Diseases (NICD), Division of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
| | - Miren Iturriza-Gómara
- Clinical Infection, Microbiology, and Immunology, University of Liverpool, Liverpool, UK
| | - Kate S Baker
- Clinical Infection, Microbiology, and Immunology, University of Liverpool, Liverpool, UK.
- Department of Genetics, University of Cambridge, CB23EH, Cambridge, UK.
| |
Collapse
|
36
|
Hanumunthadu B, Kanji N, Owino N, Ferreira Da Silva C, Robinson H, White R, Ferruzzi P, Nakakana U, Canals R, Pollard AJ, Ramasamy M. Salmonella Vaccine Study in Oxford (SALVO) trial: protocol for an observer-participant blind randomised placebo-controlled trial of the iNTS-GMMA vaccine within a European cohort. BMJ Open 2023; 13:e072938. [PMID: 37963701 PMCID: PMC10649500 DOI: 10.1136/bmjopen-2023-072938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 10/03/2023] [Indexed: 11/16/2023] Open
Abstract
INTRODUCTION Invasive non-typhoidal Salmonellosis (iNTS) is mainly caused by Salmonella enterica serovars Typhimurium and Enteritidis and is estimated to result in 77 500 deaths per year, disproportionately affecting children under 5 years of age in sub-Saharan Africa. Invasive non-typhoidal Salmonellae serovars are increasingly acquiring resistance to first-line antibiotics, thus an effective vaccine would be a valuable tool in reducing morbidity and mortality from infection. While NTS livestock vaccines are in wide use, no licensed vaccines exist for use in humans. Here, a first-in-human study of a novel vaccine (iNTS-GMMA) containing S. Typhimurium and S. Enteritidis Generalised Modules for Membrane Antigens (GMMA) outer membrane vesicles is presented. METHOD AND ANALYSIS The Salmonella Vaccine Study in Oxford is a randomised placebo-controlled participant-observer blind phase I study of the iNTS-GMMA vaccine. Healthy adult volunteers will be randomised to receive three intramuscular injections of the iNTS-GMMA vaccine, containing equal quantities of S. Typhimurium and S. Enteritidis GMMA particles adsorbed on Alhydrogel, or an Alhydrogel placebo at 0, 2 and 6 months. Participants will be sequentially enrolled into three groups: group 1, 1:1 randomisation to low dose iNTS-GMMA vaccine or placebo; group 2, 1:1 randomisation to full dose iNTS-GMMA vaccine or placebo; group 3, 2:1 randomisation to full dose or lower dose (dependant on DSMC reviews of groups 1 and 2) iNTS-GMMA vaccine or placebo.The primary objective is safety and tolerability of the vaccine. The secondary objective is immunogenicity as measured by O-antigen based ELISA. Further exploratory objectives will characterise the expanded human immune profile. ETHICS AND DISSEMINATION Ethical approval for this study has been obtained from the South Central-Oxford A Research Ethics Committee (Ethics REF:22/SC/0059). Appropriate documentation and regulatory approvals have been acquired. Results will be disseminated via peer-reviewed articles and conferences. TRIAL REGISTRATION NUMBER EudraCT Number: 2020-000510-14.
Collapse
Affiliation(s)
- Brama Hanumunthadu
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | - Nasir Kanji
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | - Nelly Owino
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | - Carla Ferreira Da Silva
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | - Hannah Robinson
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
- NIHR Oxford Biomedical Research Centre, Oxford, Oxfordshire, UK
| | - Rachel White
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | | | | | - Rocio Canals
- GSK Vaccines Institute for Global Health, Siena, Italy
| | - Andrew J Pollard
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| | - Maheshi Ramasamy
- Department of Paediatrics, University of Oxford, Oxford Vaccine Group, Oxford, Oxfordshire, UK
| |
Collapse
|
37
|
Bergkessel M, Forte B, Gilbert IH. Small-Molecule Antibiotic Drug Development: Need and Challenges. ACS Infect Dis 2023; 9:2062-2071. [PMID: 37819866 PMCID: PMC10644355 DOI: 10.1021/acsinfecdis.3c00189] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Indexed: 10/13/2023]
Abstract
The need for new antibiotics is urgent. Antimicrobial resistance is rising, although currently, many more people die from drug-sensitive bacterial infections. The continued evolution of drug resistance is inevitable, fueled by pathogen population size and exposure to antibiotics. Additionally, opportunistic pathogens will always pose a threat to vulnerable patients whose immune systems cannot efficiently fight them even if they are sensitive to available antibiotics, according to clinical microbiology tests. These problems are intertwined and will worsen as human populations age, increase in density, and experience disruptions such as war, extreme weather events, or declines in standard of living. The development of appropriate drugs to treat all the world's bacterial infections should be a priority, and future success will likely require combinations of multiple approaches. However, the highest burden of bacterial infection is in Low- and Middle-Income Countries, where limited medical infrastructure is a major challenge. For effectively managing infections in these contexts, small-molecule-based treatments offer significant advantages. Unfortunately, support for ongoing small-molecule antibiotic discovery has recently suffered from significant challenges related both to the scientific difficulties in treating bacterial infections and to market barriers. Nevertheless, small-molecule antibiotics remain essential and irreplaceable tools for fighting infections, and efforts to develop novel and improved versions deserve ongoing investment. Here, we first describe the global historical context of antibiotic treatment and then highlight some of the challenges surrounding small-molecule development and potential solutions. Many of these challenges are likely to be common to all modalities of antibacterial treatment and should be addressed directly.
Collapse
Affiliation(s)
- Megan Bergkessel
- Division
of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, U.K.
| | - Barbara Forte
- Drug
Discovery Unit and Wellcome Centre for Anti-Infectives Research, Division
of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, U.K.
| | - Ian H. Gilbert
- Drug
Discovery Unit and Wellcome Centre for Anti-Infectives Research, Division
of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, U.K.
| |
Collapse
|
38
|
Worley MJ. Salmonella Bloodstream Infections. Trop Med Infect Dis 2023; 8:487. [PMID: 37999606 PMCID: PMC10675298 DOI: 10.3390/tropicalmed8110487] [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: 09/19/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023] Open
Abstract
Salmonella is a major foodborne pathogen of both animals and humans. This bacterium is responsible for considerable morbidity and mortality world-wide. Different serovars of this genus cause diseases ranging from self-limiting gastroenteritis to a potentially fatal systemic disease known as enteric fever. Gastrointestinal infections with Salmonella are usually self-limiting and rarely require medical intervention. Bloodstream infections, on the other hand, are often fatal even with hospitalization. This review describes the routes and underlying mechanisms of the extraintestinal dissemination of Salmonella and the chronic infections that sometimes result. It includes information on the pathogenicity islands and individual virulence factors involved in systemic dissemination as well as a discussion of the host factors that mediate susceptibility. Also, the major outbreaks of invasive Salmonella disease in the tropics are described.
Collapse
Affiliation(s)
- Micah J Worley
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
39
|
Van Puyvelde S, de Block T, Sridhar S, Bawn M, Kingsley RA, Ingelbeen B, Beale MA, Barbé B, Jeon HJ, Mbuyi-Kalonji L, Phoba MF, Falay D, Martiny D, Vandenberg O, Affolabi D, Rutanga JP, Ceyssens PJ, Mattheus W, Cuypers WL, van der Sande MAB, Park SE, Kariuki S, Otieno K, Lusingu JPA, Mbwana JR, Adjei S, Sarfo A, Agyei SO, Asante KP, Otieno W, Otieno L, Tahita MC, Lompo P, Hoffman IF, Mvalo T, Msefula C, Hassan-Hanga F, Obaro S, Mackenzie G, Deborggraeve S, Feasey N, Marks F, MacLennan CA, Thomson NR, Jacobs J, Dougan G, Kariuki S, Lunguya O. A genomic appraisal of invasive Salmonella Typhimurium and associated antibiotic resistance in sub-Saharan Africa. Nat Commun 2023; 14:6392. [PMID: 37872141 PMCID: PMC10593746 DOI: 10.1038/s41467-023-41152-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/23/2023] [Indexed: 10/25/2023] Open
Abstract
Invasive non-typhoidal Salmonella (iNTS) disease manifesting as bloodstream infection with high mortality is responsible for a huge public health burden in sub-Saharan Africa. Salmonella enterica serovar Typhimurium (S. Typhimurium) is the main cause of iNTS disease in Africa. By analysing whole genome sequence data from 1303 S. Typhimurium isolates originating from 19 African countries and isolated between 1979 and 2017, here we show a thorough scaled appraisal of the population structure of iNTS disease caused by S. Typhimurium across many of Africa's most impacted countries. At least six invasive S. Typhimurium clades have already emerged, with ST313 lineage 2 or ST313-L2 driving the current pandemic. ST313-L2 likely emerged in the Democratic Republic of Congo around 1980 and further spread in the mid 1990s. We observed plasmid-borne as well as chromosomally encoded fluoroquinolone resistance underlying emergences of extensive-drug and pan-drug resistance. Our work provides an overview of the evolution of invasive S. Typhimurium disease, and can be exploited to target control measures.
Collapse
Affiliation(s)
- Sandra Van Puyvelde
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK.
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium.
| | | | - Sushmita Sridhar
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Division of Infectious Disease, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Matt Bawn
- Quadram Institute Bioscience, Norwich, UK
- Earlham Institute, Norwich, UK
- Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Robert A Kingsley
- Quadram Institute Bioscience, Norwich, UK
- School of Biological Science, University of East Anglia, Norwich, UK
| | - Brecht Ingelbeen
- Institute of Tropical Medicine, Antwerp, Belgium
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Mathew A Beale
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Hyon Jin Jeon
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
- International Vaccine Institute, 1 Gwanak-ro, Seoul, 08826, Republic of Korea
- Madagascar Institute for Vaccine Research, University of Antananarivo, Antananarivo, Madagascar
| | - Lisette Mbuyi-Kalonji
- Department of Medical Biology, University Teaching Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
- National Institute for Biomedical Research, Kinshasa, Democratic Republic of the Congo
| | - Marie-France Phoba
- Department of Medical Biology, University Teaching Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
- National Institute for Biomedical Research, Kinshasa, Democratic Republic of the Congo
| | - Dadi Falay
- Department of Pediatrics, University Hospital of Kisangani, Kisangani, Democratic Republic of the Congo
| | - Delphine Martiny
- Department of Microbiology, Laboratoire Hospitalier Universitaire de Bruxelles-Universitair Laboratorium Brussel (LHUB-ULB), Université Libre de Bruxelles (ULB), 1000, Brussels, Belgium
- Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000, Mons, Belgium
| | - Olivier Vandenberg
- Department of Microbiology, Laboratoire Hospitalier Universitaire de Bruxelles-Universitair Laboratorium Brussel (LHUB-ULB), Université Libre de Bruxelles (ULB), 1000, Brussels, Belgium
- Division of Infection and Immunity, Faculty of Medical Sciences, University College London, London, UK
| | - Dissou Affolabi
- Centre National Hospitalier Universitaire Hubert Koutoukou Maga, Cotonou, Benin
| | - Jean Pierre Rutanga
- Institute of Tropical Medicine, Antwerp, Belgium
- College of Science and Technology, University of Rwanda, Kigali, Rwanda
| | - Pieter-Jan Ceyssens
- National Reference Center for Salmonella, Unit of Human Bacterial Diseases, Sciensano, J. Wytsmanstraat 14, B-1050, Brussels, Belgium
| | - Wesley Mattheus
- National Reference Center for Salmonella, Unit of Human Bacterial Diseases, Sciensano, J. Wytsmanstraat 14, B-1050, Brussels, Belgium
| | - Wim L Cuypers
- Institute of Tropical Medicine, Antwerp, Belgium
- Department of Computer Science, University of Antwerp, Antwerp, Belgium
| | - Marianne A B van der Sande
- Institute of Tropical Medicine, Antwerp, Belgium
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Se Eun Park
- International Vaccine Institute, 1 Gwanak-ro, Seoul, 08826, Republic of Korea
- Yonsei University Graduate School of Public Health, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Simon Kariuki
- Kenya Medical Research Institute/Centre for Global Health Research, Kisumu, Kenya
| | - Kephas Otieno
- Kenya Medical Research Institute/Centre for Global Health Research, Kisumu, Kenya
| | - John P A Lusingu
- National Institute for Medical Research, Tanga, Tanzania
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, København, Denmark
| | - Joyce R Mbwana
- National Institute for Medical Research, Tanga, Tanzania
| | - Samuel Adjei
- University of Health & Allied Sciences, Ho, Volta Region, Ghana
| | - Anima Sarfo
- University of Health & Allied Sciences, Ho, Volta Region, Ghana
| | - Seth O Agyei
- University of Health & Allied Sciences, Ho, Volta Region, Ghana
| | - Kwaku P Asante
- Kintampo Health Research Centre, Research and Development Division, Ghana Health Service, Kintampo North Municipality, Ho, Volta Region, Ghana
| | | | | | - Marc C Tahita
- Institut de Recherche en Science de la Santé, Direction Régionale du Centre-Ouest/ClinicalResearch Unit of Nanoro, Nanoro, Burkina Faso
| | - Palpouguini Lompo
- Institut de Recherche en Science de la Santé, Direction Régionale du Centre-Ouest/ClinicalResearch Unit of Nanoro, Nanoro, Burkina Faso
| | | | - Tisungane Mvalo
- University of North Carolina Project, Lilongwe, Malawi
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chisomo Msefula
- Malawi Liverpool Wellcome Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Fatimah Hassan-Hanga
- Department of Paediatrics, Bayero University, Kano, Nigeria
- Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Stephen Obaro
- University of Nebraska Medical Center, Omaha, NE, USA
- International Foundation Against Infectious Diseases in Nigeria (IFAIN), Abuja, Nigeria
| | - Grant Mackenzie
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, The Gambia
- London School of Hygiene and Tropical Medicine, Keppel St, Bloomsbury, London, WC1E 7HT, UK
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | | | - Nicholas Feasey
- University of North Carolina Project, Lilongwe, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Florian Marks
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
- International Vaccine Institute, 1 Gwanak-ro, Seoul, 08826, Republic of Korea
- Madagascar Institute for Vaccine Research, University of Antananarivo, Antananarivo, Madagascar
- Heidelberg Institute of Global Health, University of Heidelberg, Heidelberg, Germany
| | - Calman A MacLennan
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Enteric and Diarrheal Diseases, Global Health, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Nicholas R Thomson
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- London School of Hygiene and Tropical Medicine, Keppel St, Bloomsbury, London, WC1E 7HT, UK
| | - Jan Jacobs
- Institute of Tropical Medicine, Antwerp, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Gordon Dougan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
| | - Samuel Kariuki
- Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Octavie Lunguya
- Department of Medical Biology, University Teaching Hospital of Kinshasa, Kinshasa, Democratic Republic of the Congo
- National Institute for Biomedical Research, Kinshasa, Democratic Republic of the Congo
| |
Collapse
|
40
|
Falay D, Hardy L, Bonebe E, Mattheus W, Ngbonda D, Lunguya O, Jacobs J. Intestinal carriage of invasive non-typhoidal Salmonella among household members of children with Salmonella bloodstream infection, Kisangani, DR Congo. Front Microbiol 2023; 14:1241961. [PMID: 37901802 PMCID: PMC10602682 DOI: 10.3389/fmicb.2023.1241961] [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: 06/18/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Invasive non-typhoidal Salmonella (iNTS), mainly Salmonella Typhimurium and Salmonella Enteritidis, causes a severe burden in sub-Saharan Africa; however, its reservoir (animal or environmental) is unclear. The present study assessed healthy household members of index patients for intestinal carriage of Salmonella. Methods Index patients were admitted to the University Hospital of Kisangani (DR Congo), and Salmonella was grown from blood cultures. Household members were asked to provide three stool samples for culture for Salmonella. Salmonella Typhimurium and S. Enteritidis isolates from index patients, and household members were assessed for genetic relatedness using the multiple-locus variable number of tandem repeat analysis (MLVA), and the multilocus sequence type (ST) was determined by whole genome sequencing. Results Between May 2016 and January 2020, 22 households were visited. The index patient serotypes were Typhimurium, Enteritidis, Typhi, and Paratyphi C; II:42:r:-; and I:7:y:- (n = 8, 7, 5, and each 1, respectively). The median (range) delay between the index patient and household sampling was 25 days (2 days to 7.3 months); 203 household members provided at least one stool sample. In all, 15 (7.3%) Salmonella carriers were found in nine of 22 households. For one index patient, the household comprised S. Typhimurium in four household members, including the index patient, sampled 27 days after bloodstream infection; the MLVA types of these five isolates were similar. They belonged to ST313 lineage 2 and were closely related [0-1 allelic distance (AD) among the stool isolates and eight AD with the blood culture isolate]. In another household, the stool culture of the index patient (obtained 67 days after bloodstream infection) grew S. Enteritidis of the same MLVA type; both isolates belonged to the ST11 Central/Eastern African clade and were closely related (three AD). Discussion The present study provides evidence of household clustering of S. Typhimurium ST313 and intestinal carriage of iNTS several weeks after bloodstream infection.
Collapse
Affiliation(s)
- Dadi Falay
- Department of Pediatrics, University Hospital of Kisangani, Kisangani, Democratic Republic of Congo
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Liselotte Hardy
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Edmonde Bonebe
- Department of Microbiology, National Institute for Biomedical Research, Kinshasa, Democratic Republic of Congo
| | - Wesley Mattheus
- Division of Human Bacterial Diseases, Sciensano, Uccle, Belgium
| | - Dauly Ngbonda
- Department of Pediatrics, University Hospital of Kisangani, Kisangani, Democratic Republic of Congo
| | - Octavie Lunguya
- Department of Microbiology, National Institute for Biomedical Research, Kinshasa, Democratic Republic of Congo
- Department of Microbiology, University Teaching Hospital of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Jan Jacobs
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| |
Collapse
|
41
|
Lewis ED, Ortega EF, Dao MC, Barger K, Mason JB, Leong JM, Osburne MS, Magoun L, Nepveux V FJ, Chishti AH, Schwake C, Quynh A, Gilhooly CH, Petty G, Guo W, Matuszek G, Pereira D, Reddy M, Wang J, Wu D, Meydani SN, Combs GF. Safe and effective delivery of supplemental iron to healthy adults: a two-phase, randomized, double-blind trial - the safe iron study. Front Nutr 2023; 10:1230061. [PMID: 37899826 PMCID: PMC10603204 DOI: 10.3389/fnut.2023.1230061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 08/28/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction The safety of novel forms of iron in healthy, iron-replete adults as might occur if used in population-based iron supplementation programs was examined. We tested the hypotheses that supplementation with nanoparticulate iron hydroxide adipate tartrate (IHAT), an iron-enriched Aspergillus oryzae product (ASP), or ferrous sulphate heptahydrate (FS) are safe as indicated by erythrocyte susceptibility to malarial infection, bacterial proliferation, and gut inflammation. Responses to FS administered daily or weekly, and with or without other micronutrients were compared. Methods Two phases of randomized, double-blinded trials were conducted in Boston, MA. Phase I randomized 160 volunteers to six treatments: placebo, IHAT, ASP, FS, and FS plus a micronutrient powder (MNP) administrated daily at 60 mg Fe/day; and FS administered as a single weekly dose of 420 mg Fe. Phase II randomized 86 volunteers to IHAT, ASP, or FS administered at 120 mg Fe/day. Completing these phases were 151 and 77 participants, respectively. The study was powered to detect effects on primary endpoints: susceptibility of participant erythrocytes to infection by Plasmodium falciparum, the proliferation potential of selected pathogenic bacteria in sera, and markers of gut inflammation. Secondary endpoints for which the study was not powered included indicators of iron status and gastrointestinal symptoms. Results Supplementation with any form of iron did not affect any primary endpoint. In Phase I, the frequency of gastrointestinal symptoms associated with FS was unaffected by dosing with MNP or weekly administration; but participants taking IHAT more frequently reported abdominal pain (27%, p < 0.008) and nausea (4%, p = 0.009) than those taking FS, while those taking ASP more frequently reported nausea (8%, p = 0.009). Surprisingly, only 9% of participants taking IHAT at 120 mg Fe/day (Phase II) reported abdominal pain and no other group reported that symptom. Discussion With respect to the primary endpoints, few differences were found when comparing these forms of iron, indicating that 28 days of 60 or 120 mg/day of IHAT, ASP, or FS may be safe for healthy, iron-replete adults. With respect to other endpoints, subjects receiving IHAT more frequently reported abdominal pain and nausea, suggesting the need for further study. Clinical Trial Registration ClinicalTrials.gov, NCT03212677; registered: 11 July 2017.
Collapse
Affiliation(s)
- Erin D. Lewis
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Edwin F. Ortega
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Maria Carlota Dao
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Kathryn Barger
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Joel B. Mason
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, United States
| | - Marcia S. Osburne
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, United States
| | - Loranne Magoun
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, United States
| | - Felix J. Nepveux V
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, United States
| | - Athar H. Chishti
- Department of Developmental, Molecular and Chemical Biology, Tufts University, Boston, MA, United States
| | - Christopher Schwake
- Department of Developmental, Molecular and Chemical Biology, Tufts University, Boston, MA, United States
| | - Anh Quynh
- Department of Developmental, Molecular and Chemical Biology, Tufts University, Boston, MA, United States
| | - Cheryl H. Gilhooly
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Gayle Petty
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Weimin Guo
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Gregory Matuszek
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Dora Pereira
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Manju Reddy
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
| | - Jifan Wang
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Dayong Wu
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Simin N. Meydani
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Gerald F. Combs
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| |
Collapse
|
42
|
Chung The H, Pham P, Ha Thanh T, Phuong LVK, Yen NP, Le SNH, Vu Thuy D, Chau TTH, Le Phuc H, Ngoc NM, Vi LL, Mather AE, Thwaites GE, Thomson NR, Baker S, Pham DT. Multidrug resistance plasmids underlie clonal expansions and international spread of Salmonella enterica serotype 1,4,[5],12:i:- ST34 in Southeast Asia. Commun Biol 2023; 6:1007. [PMID: 37789208 PMCID: PMC10547704 DOI: 10.1038/s42003-023-05365-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 09/15/2023] [Indexed: 10/05/2023] Open
Abstract
Salmonella enterica serotype 1,4,[5],12:i:- (Typhimurium monophasic variant) of sequence type (ST) 34 has emerged as the predominant pandemic genotype in recent decades. Despite increasing reports of resistance to antimicrobials in Southeast Asia, Salmonella ST34 population structure and evolution remained understudied in the region. Here we performed detailed genomic investigations on 454 ST34 genomes collected from Vietnam and diverse geographical sources to elucidate the pathogen's epidemiology, evolution and antimicrobial resistance. We showed that ST34 has been introduced into Vietnam in at least nine occasions since 2000, forming five co-circulating major clones responsible for paediatric diarrhoea and bloodstream infection. Most expansion events were associated with acquisitions of large multidrug resistance plasmids of IncHI2 or IncA/C2. Particularly, the self-conjugative IncA/C2 pST34VN2 (co-transferring blaCTX-M-55, mcr-3.1, and qnrS1) underlies local expansion and intercontinental spread in two separate ST34 clones. At the global scale, Southeast Asia was identified as a potential hub for the emergence and dissemination of multidrug resistant Salmonella ST34, and mutation analysis suggests of selection in antimicrobial responses and key virulence factors.
Collapse
Affiliation(s)
- Hao Chung The
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.
| | - Phuong Pham
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Tuyen Ha Thanh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | | | | | - Son-Nam H Le
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Duong Vu Thuy
- Children's Hospital No. 1, Ho Chi Minh City, Vietnam
| | | | - Hoang Le Phuc
- Children's Hospital No. 1, Ho Chi Minh City, Vietnam
| | | | - Lu Lan Vi
- The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Alison E Mather
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- University of East Anglia, Norwich, UK
| | - Guy E Thwaites
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Nicholas R Thomson
- London School of Hygiene and Tropical Medicine, London, UK
- The Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Stephen Baker
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Diseases (CITIID), University of Cambridge, Cambridge, UK
| | - Duy Thanh Pham
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
43
|
Smith C, Smith E, Chiu C, Hinton J, Perez Sepulveda B, Gordon M, Choy RK, Hill PW, Meiring JE, Darton TC, Carey ME, Cooke G, Gibani MM. The Challenge Non-Typhoidal Salmonella (CHANTS) Consortium: Development of a non-typhoidal Salmonella controlled human infection model: Report from a consultation group workshop, 05 July 2022, London, UK. Wellcome Open Res 2023; 8:111. [PMID: 37808389 PMCID: PMC10558987 DOI: 10.12688/wellcomeopenres.19012.2] [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] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Invasive non-typhoidal Salmonella disease (iNTS) is a major cause of morbidity and mortality globally, particularly as a cause of bloodstream infection in children and immunocompromised adults in sub-Saharan Africa. Vaccines to prevent non-typhoidal Salmonella (NTS) would represent a valuable public health tool in this setting to avert cases and prevent expansion of antimicrobial resistance. Several NTS and combination typhoidal-NTS vaccine candidates are in early-stage development, although the pathway to licensure is unclear due to challenges in conducting large phase III field trials. Controlled human infection models (CHIM) present an opportunity to accelerate vaccine development for a range of enteric pathogens. Several recent typhoidal Salmonella CHIMs have been conducted safely and have played pivotal roles in progressing vaccine candidates to pre-qualification and licensure. The Challenge Non-Typhoidal Salmonella (CHANTS) consortium has been formed with funding from the Wellcome Trust, to deliver the first NTS CHIM, which can act as a platform for future vaccine evaluation. This paper reports the conclusions of a consultation group workshop convened with key stakeholders. The aims of this meeting were to: (1) define the rationale for an NTS CHIM (2) map the NTS vaccine pipeline (3) refine study design and (4) establish potential future use cases.
Collapse
Affiliation(s)
- Christopher Smith
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Emma Smith
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Jay Hinton
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
| | - Blanca Perez Sepulveda
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
| | - Melita Gordon
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | | | - Peter W.S. Hill
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - James E. Meiring
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Thomas C. Darton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Megan E. Carey
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Graham Cooke
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Malick M. Gibani
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - CHANTS Consortium
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- PATH, Seattle, Washington, 98121, USA
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
44
|
Smith C, Smith E, Chiu C, Hinton J, Perez Sepulveda B, Gordon M, Choy RK, Hill PW, Meiring JE, Darton TC, Carey ME, Cooke G, Gibani MM. The Challenge Non-Typhoidal Salmonella (CHANTS) Consortium: Development of a non-typhoidal Salmonella controlled human infection model: Report from a consultation group workshop, 05 July 2022, London, UK. Wellcome Open Res 2023; 8:111. [PMID: 37808389 PMCID: PMC10558987 DOI: 10.12688/wellcomeopenres.19012.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 04/21/2024] Open
Abstract
Invasive non-typhoidal Salmonella disease (iNTS) is a major cause of morbidity and mortality globally, particularly as a cause of bloodstream infection in children and immunocompromised adults in sub-Saharan Africa. Vaccines to prevent non-typhoidal Salmonella (NTS) would represent a valuable public health tool in this setting to avert cases and prevent expansion of antimicrobial resistance. Several NTS and combination typhoidal-NTS vaccine candidates are in early-stage development, although the pathway to licensure is unclear due to challenges in conducting large phase III field trials. Controlled human infection models (CHIM) present an opportunity to accelerate vaccine development for a range of enteric pathogens. Several recent typhoidal Salmonella CHIMs have been conducted safely and have played pivotal roles in progressing vaccine candidates to pre-qualification and licensure. The Challenge Non-Typhoidal Salmonella (CHANTS) consortium has been formed with funding from the Wellcome Trust, to deliver the first NTS CHIM, which can act as a platform for future vaccine evaluation. This paper reports the conclusions of a consultation group workshop convened with key stakeholders. The aims of this meeting were to: (1) define the rationale for an NTS CHIM (2) map the NTS vaccine pipeline (3) refine study design and (4) establish potential future use cases.
Collapse
Affiliation(s)
- Christopher Smith
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Emma Smith
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Jay Hinton
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
| | - Blanca Perez Sepulveda
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
| | - Melita Gordon
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | | | - Peter W.S. Hill
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - James E. Meiring
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Thomas C. Darton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
| | - Megan E. Carey
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Graham Cooke
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - Malick M. Gibani
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
| | - CHANTS Consortium
- Department of Infectious Disease, Imperial College London, London, W2 1PG, UK
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 3BX, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- PATH, Seattle, Washington, 98121, USA
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2TN, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
45
|
Nuanmuang N, Leekitcharoenphon P, Njage PMK, Gmeiner A, Aarestrup FM. An Overview of Antimicrobial Resistance Profiles of Publicly Available Salmonella Genomes with Sufficient Quality and Metadata. Foodborne Pathog Dis 2023; 20:405-413. [PMID: 37540138 PMCID: PMC10510693 DOI: 10.1089/fpd.2022.0080] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023] Open
Abstract
Salmonella enterica (S. enterica) is a commensal organism or pathogen causing diseases in animals and humans, as well as widespread in the environment. Antimicrobial resistance (AMR) has increasingly affected both animal and human health and continues to raise public health concerns. A decade ago, it was estimated that the increased use of whole genome sequencing (WGS) combined with sharing of public data would drastically change and improve the surveillance and understanding of Salmonella epidemiology and AMR. This study aimed to evaluate the current usefulness of public WGS data for Salmonella surveillance and to investigate the associations between serovars, antibiotic resistance genes (ARGs), and metadata. Out of 191,306 Salmonella genomes deposited in European Nucleotide Archive and NCBI databases, 47,452 WGS with sufficient minimum metadata (country, year, and source) of S. enterica were retrieved from 116 countries and isolated between 1905 and 2020. For in silico analysis of the WGS data, KmerFinder, SISTR, and ResFinder were used for species, serovars, and AMR identification, respectively. The results showed that the five common isolation sources of S. enterica are human (29.10%), avian (22.50%), environment (11.89%), water (9.33%), and swine (6.62%). The most common ARG profiles for each class of antimicrobials are β-lactam (blaTEM-1B; 6.78%), fluoroquinolone [(parC[T57S], qnrB19); 0.87%], folate pathway antagonist (sul2; 8.35%), macrolide [mph(A); 0.39%], phenicol (floR; 5.94%), polymyxin B (mcr-1.1; 0.09%), and tetracycline [tet(A); 12.95%]. Our study reports the first overview of ARG profiles in publicly available Salmonella genomes from online databases. All data sets from this study can be searched at Microreact.
Collapse
Affiliation(s)
- Narong Nuanmuang
- Research Group for Genomic Epidemiology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Pimlapas Leekitcharoenphon
- Research Group for Genomic Epidemiology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Patrick Murigu Kamau Njage
- Research Group for Genomic Epidemiology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Alexander Gmeiner
- Research Group for Genomic Epidemiology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Frank M. Aarestrup
- Research Group for Genomic Epidemiology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
46
|
Fu Y, M’ikanatha NM, Dudley EG. Whole-Genome Subtyping Reveals Population Structure and Host Adaptation of Salmonella Typhimurium from Wild Birds. J Clin Microbiol 2023; 61:e0184722. [PMID: 37249426 PMCID: PMC10281135 DOI: 10.1128/jcm.01847-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/12/2023] [Indexed: 05/31/2023] Open
Abstract
Within-host evolution of bacterial pathogens can lead to host-associated variants of the same species or serovar. Identification and characterization of closely related variants from diverse host species are crucial to public health and host-pathogen adaptation research. However, the work remained largely underexplored at a strain level until the advent of whole-genome sequencing (WGS). Here, we performed WGS-based subtyping and analyses of Salmonella enterica serovar Typhimurium (n = 787) from different wild birds across 18 countries over a 75-year period. We revealed seven avian host-associated S. Typhimurium variants/lineages. These lineages emerged globally over short timescales and presented genetic features distinct from S. Typhimurium lineages circulating among humans and domestic animals. We further showed that, in terms of virulence, host adaptation of these variants was driven by genome degradation. Our results provide a snapshot of the population structure and genetic diversity of S. Typhimurium within avian hosts. We also demonstrate the value of WGS-based subtyping and analyses in unravelling closely related variants at the strain level.
Collapse
Affiliation(s)
- Yezhi Fu
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania, USA
| | | | - Edward G. Dudley
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania, USA
- E. coli Reference Center, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
47
|
Chen K, Zhan Z, Li L, Li J, Zhou Z, Wang N, Sun D, Xu C, Chen P, Qu X, Liao M, Zhang J. BolA affects the biofilm formation ability, outer membrane permeability and virulence, thus is required for the adaptability of Salmonella enterica serotype Typhimurium to the harsh survival environment. Microbiol Res 2023; 274:127423. [PMID: 37295142 DOI: 10.1016/j.micres.2023.127423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023]
Abstract
Salmonella enterica serotype Typhimurium, an important foodborne pathogen with high adaptability to the host's internal and external survival environment, seriously threatens public health. Therefore, to understand the mechanism underlying the high adaptability, this study investigated the transcription factor BolA by constructing BolA deletion strain 269△BolA, complemented strain 269BolAR and overexpression strain 269BolA+ based on WT269. BolA significantly inhibited motility; at 6 h, the BolA overexpression strain (269BolA+) showed 91.2% and 90.7% lower motility than the wild type (WT269) and BolA deletion strain (269△BolA), respectively, by downregulating motility-related flagellar genes. BolA promoted biofilm formation; 269BolA+ showed 3.6-fold and 5.2-fold higher biofilm formation ability than WT269 and 269ΔBolA, respectively, by upregulation biofilm formation-related genes. BolA overexpression downregulated the outer membrane gene OmpF and upregulated OmpC, thereby regulating cell permeability, and reducing the antibacterial effect of vancomycin, which can destruct the outer membrane. BolA improved adaptability; 269△BolA showed higher susceptibility to eight antibiotics and 2.5- and 4-fold lower acid and oxidative stress tolerance, respectively, than WT269. In Caco-2 and HeLa cells, 269△BolA showed 2.8- and 3-fold lower cell adhesion ability, respectively, and 4- and 2-fold lower cell invasion ability, respectively, than WT269, through downregulation of the virulence genes. Thus, BolA expression promotes biofilm formation and balances the membrane permeability, thereby improving the resistance of the strains, and enhances its host cell invasion ability by upregulating bacterial virulence factors. Results of this study suggest that the BolA gene may serve as a potential target of therapeutic or preventative strategies to control Salmonella Typhimurium infections.
Collapse
Affiliation(s)
- Kaifeng Chen
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zeqiang Zhan
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Lili Li
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jiayi Li
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhouping Zhou
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Nanwei Wang
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Dage Sun
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Chenggang Xu
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Pengju Chen
- Henan Institute of Modern Chinese Veterinary Medicine, Henan 450000, China
| | - Xiaoyun Qu
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ming Liao
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Jianmin Zhang
- Center of Emerging and Zoonotic Diseases, National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
48
|
Vasicek EM, Gunn JS. Invasive Non-Typhoidal Salmonella Lineage Biofilm Formation and Gallbladder Colonization Vary But Do Not Correlate Directly with Known Biofilm-Related Mutations. Infect Immun 2023; 91:e0013523. [PMID: 37129526 PMCID: PMC10187132 DOI: 10.1128/iai.00135-23] [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: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/03/2023] Open
Abstract
Non-typhoidal Salmonella (NTS) serovars have a broad host range and cause gastroenteritis in humans. However, invasive NTS (iNTS) bloodstream infections have increased in the last decade, causing 60,000 deaths annually. Human-specific typhoidal Salmonella colonizes and forms biofilms on gallstones, resulting in chronic, asymptomatic infection. iNTS lineages are undergoing genomic reduction and may have adapted to person-to-person transmission via mutations in virulence, bile resistance, and biofilm formation. As such, we sought to determine the capacity of iNTS lineages for biofilm formation and the development of chronic infections in the gallbladder in our mouse model. Of the lineages tested (L1, L2, L3 and UK), only L2 and UK were defective for the rough, dry and red (RDAR) morphotype, correlating with the known bcsG (cellulose) mutation but not with csgD (curli) gene mutations. Biofilm-forming ability was assessed in vitro, which revealed a biofilm formation hierarchy of L3 > ST19 > UK > L1 = L2, which did not correlate directly with either the bcsG or the csgD mutation. By confocal microscopy, biofilms of L2 and UK had significantly less curli and cellulose, while L1 biofilms had significantly lower cellulose. All iNTS strains were able to colonize the mouse gallbladder, liver, and spleen in a similar manner, while L3 had a significantly higher bacterial load in the gallbladder and increased lethality. While there was iNTS lineage variability in biofilm formation, gallbladder colonization, and virulence in a chronic mouse model, all tested lineages were capable of colonization despite possessing biofilm-related mutations. Thus, iNTS strains may be unrecognized chronic pathogens in endemic settings.
Collapse
Affiliation(s)
- Erin M. Vasicek
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| | - John S. Gunn
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
49
|
Fong WY, Canals R, Predeus AV, Perez-Sepulveda B, Wenner N, Lacharme-Lora L, Feasey N, Wigley P, Hinton JCD. Genome-wide fitness analysis identifies genes required for in vitro growth and macrophage infection by African and global epidemic pathovariants of Salmonella enterica Enteritidis. Microb Genom 2023; 9:mgen001017. [PMID: 37219927 PMCID: PMC10272866 DOI: 10.1099/mgen.0.001017] [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: 09/08/2022] [Accepted: 03/17/2023] [Indexed: 05/24/2023] Open
Abstract
Salmonella enterica Enteritidis is the second most common serovar associated with invasive non-typhoidal Salmonella (iNTS) disease in sub-Saharan Africa. Previously, genomic and phylogenetic characterization of S . enterica Enteritidis isolates from the human bloodstream led to the discovery of the Central/Eastern African clade (CEAC) and West African clade, which were distinct from the gastroenteritis-associated global epidemic clade (GEC). The African S . enterica Enteritidis clades have unique genetic signatures that include genomic degradation, novel prophage repertoires and multi-drug resistance, but the molecular basis for the enhanced propensity of African S . enterica Enteritidis to cause bloodstream infection is poorly understood. We used transposon insertion sequencing (TIS) to identify the genetic determinants of the GEC representative strain P125109 and the CEAC representative strain D7795 for growth in three in vitro conditions (LB or minimal NonSPI2 and InSPI2 growth media), and for survival and replication in RAW 264.7 murine macrophages. We identified 207 in vitro -required genes that were common to both S . enterica Enteritidis strains and also required by S . enterica Typhimurium, S . enterica Typhi and Escherichia coli , and 63 genes that were only required by individual S . enterica Enteritidis strains. Similar types of genes were required by both P125109 and D7795 for optimal growth in particular media. Screening the transposon libraries during macrophage infection identified 177 P125109 and 201 D7795 genes that contribute to bacterial survival and replication in mammalian cells. The majority of these genes have proven roles in Salmonella virulence. Our analysis uncovered candidate strain-specific macrophage fitness genes that could encode novel Salmonella virulence factors.
Collapse
Affiliation(s)
- Wai Yee Fong
- Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Present address: Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, USA
| | - Rocío Canals
- Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Present address: GSK Vaccines Institute for Global Health S.R.L., Siena, Italy
| | - Alexander V. Predeus
- Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Present address: Wellcome Trust Sanger Institute, Cambridge, UK
| | - Blanca Perez-Sepulveda
- Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Nicolas Wenner
- Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Present address: Biozentrum, University of Basel, Basel, Switzerland
| | - Lizeth Lacharme-Lora
- Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Nicholas Feasey
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Malawi-Liverpool-Wellcome Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Paul Wigley
- Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK
- Present address: Bristol Veterinary School,University of Bristol, Langford Campus, UK
| | - Jay C. D. Hinton
- Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
50
|
Teklemariam AD, Al-Hindi RR, Albiheyri RS, Alharbi MG, Alghamdi MA, Filimban AAR, Al Mutiri AS, Al-Alyani AM, Alseghayer MS, Almaneea AM, Albar AH, Khormi MA, Bhunia AK. Human Salmonellosis: A Continuous Global Threat in the Farm-to-Fork Food Safety Continuum. Foods 2023; 12:foods12091756. [PMID: 37174295 PMCID: PMC10178548 DOI: 10.3390/foods12091756] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Salmonella is one of the most common zoonotic foodborne pathogens and a worldwide public health threat. Salmonella enterica is the most pathogenic among Salmonella species, comprising over 2500 serovars. It causes typhoid fever and gastroenteritis, and the serovars responsible for the later disease are known as non-typhoidal Salmonella (NTS). Salmonella transmission to humans happens along the farm-to-fork continuum via contaminated animal- and plant-derived foods, including poultry, eggs, fish, pork, beef, vegetables, fruits, nuts, and flour. Several virulence factors have been recognized to play a vital role in attaching, invading, and evading the host defense system. These factors include capsule, adhesion proteins, flagella, plasmids, and type III secretion systems that are encoded on the Salmonella pathogenicity islands. The increased global prevalence of NTS serovars in recent years indicates that the control approaches centered on alleviating the food animals' contamination along the food chain have been unsuccessful. Moreover, the emergence of antibiotic-resistant Salmonella variants suggests a potential food safety crisis. This review summarizes the current state of the knowledge on the nomenclature, microbiological features, virulence factors, and the mechanism of antimicrobial resistance of Salmonella. Furthermore, it provides insights into the pathogenesis and epidemiology of Salmonella infections. The recent outbreaks of salmonellosis reported in different clinical settings and geographical regions, including Africa, the Middle East and North Africa, Latin America, Europe, and the USA in the farm-to-fork continuum, are also highlighted.
Collapse
Affiliation(s)
- Addisu D Teklemariam
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rashad R Al-Hindi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Raed S Albiheyri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mona G Alharbi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mashail A Alghamdi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amani A R Filimban
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdullah S Al Mutiri
- Laboratory Department, Saudi Food and Drug Authority, Riyadh 12843, Saudi Arabia
| | - Abdullah M Al-Alyani
- Laboratory Department, Saudi Food and Drug Authority, Jeddah 22311, Saudi Arabia
| | - Mazen S Alseghayer
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Monitoring and Risk Assessment Department, Saudi Food and Drug Authority, Riyadh 13513, Saudi Arabia
| | - Abdulaziz M Almaneea
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Monitoring and Risk Assessment Department, Saudi Food and Drug Authority, Riyadh 13513, Saudi Arabia
| | - Abdulgader H Albar
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Microbiology and Medical Parasitology, Faculty of Medicine, Jeddah University, Jeddah 23218, Saudi Arabia
| | - Mohsen A Khormi
- Department of Biological Sciences, Faculty of Sciences, Jazan University, Jazan 82817, Saudi Arabia
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
- Purdue University Interdisciplinary Life Science Program (PULSe), West Lafayette, IN 47907, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|