1
|
Uzzal Hossain M, Khan Tanvir N, Naimur Rahman ABZ, Mahmud Chowdhury Z, Shahadat Hossain M, Dey S, Bhattacharjee A, Ahammad I, Salma Zohora U, Hashem A, Chandra Das K, Ara Keya C, Salimullah M. From sequence to significance: A thorough investigation of the distinctive genome features uncovered in C. Werkmanii strain NIB003. Gene 2025; 933:148965. [PMID: 39332601 DOI: 10.1016/j.gene.2024.148965] [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: 06/24/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Citrobacter werkmanii (C. werkmanii), an opportunistic urinary bacterium that causes diarrhea, is poorly understood. Our research focuses on genetic features that are crucial to disease development, such as pathogenic interactions, antibiotic resistance, virulence genes and genetic variation. Following its morphological, biochemical, and molecular identification, the whole genome of C. werkmanii strain NIB003 was sequenced in Bangladesh for the first time. Despite having around 80% whole genome conservation, the research shows that the Bangladeshi strain forms a separate phylogenetic cluster. This emphasises the genetic variability within C. werkmanii, resulting in particular modifications at the strain level and changes in its ability to cause disease. The results of the genetic diversity analysis indicate that the Bangladeshi sequenced genome is more diverse than the other strains due to the existence of unique features, such as the presence of t-RNA binding domain and N-6 adenine-specific DNA methylases.
Collapse
Affiliation(s)
- Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh
| | - Neyamat Khan Tanvir
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh; Dept. of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - A B Z Naimur Rahman
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh; Dept. of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Zeshan Mahmud Chowdhury
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh
| | - Md Shahadat Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh; Dept. of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Shajib Dey
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh; Dept. of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | - Arittra Bhattacharjee
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh
| | - Ishtiaque Ahammad
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh
| | - Umme Salma Zohora
- Dept. of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Abu Hashem
- Microbial Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh
| | - Keshob Chandra Das
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh
| | - Chaman Ara Keya
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka-1229, Bangladesh
| | - Md Salimullah
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh; Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka-1349, Bangladesh.
| |
Collapse
|
2
|
Arjun OK, Sethi M, Parida D, Dash J, Kumar Das S, Prakash T, Senapati S. Comprehensive physiological and genomic characterization of a potential probiotic strain, Lactiplantibacillus plantarum ILSF15, isolated from the gut of tribes of Odisha, India. Gene 2024; 931:148882. [PMID: 39182659 DOI: 10.1016/j.gene.2024.148882] [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: 03/18/2024] [Revised: 08/11/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Characterizing probiotic features of organisms isolated from diverse environments can lead to the discovery of novel strains with promising functional features and health attributes. The present study attempts to characterize a novel probiotic strain isolated from the gut of the tribal population of Odisha, India. Based on 16S rRNA-based phylogeny, the strain was identified as a species of the Lactiplantibacillus genus and was named Lactiplantibacillus plantarum strain ILSF15. The current investigation focuses on elucidating this strain's genetic and physiological properties associated with probiotic attributes such as biosafety risk, host adaptation/survival traits, and beneficial functional features. The novel strain was observed, in vitro, exhibiting features such as acid/bile tolerance, adhesion to the host enteric epithelial cells, cholesterol assimilation, and pathogen exclusion, indicating its ability to survive the harsh environment of the human GIT and resist the growth of harmful microorganisms. Additionally, the L. plantarum ILSF15 strain was found to harbor genes associated with the metabolism and synthesis of various bioactive molecules, including amino acids, carbohydrates, lipids, and vitamins, highlighting the organism's ability to efficiently utilize diverse resources and contribute to the host's nutrition and health. Several genes involved in host adaptation/survival strategies and host-microbe interactions were also identified from the ILSF15 genome. Moreover, L. plantarum strains, in general, were found to have an open pangenome characterized by high genetic diversity and the absence of specific lineages associated with particular habitats, signifying its versatile nature and potential applications in probiotic and functional food industries.
Collapse
Affiliation(s)
- O K Arjun
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Himachal Pradesh 175005, India
| | - Manisha Sethi
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha 751023, India
| | - Deepti Parida
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha 751023, India
| | - Jayalaxmi Dash
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha 751023, India
| | - Suraja Kumar Das
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha 751023, India
| | - Tulika Prakash
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Himachal Pradesh 175005, India.
| | | |
Collapse
|
3
|
Wu F, Yang B, Xiao Y, Ren LL, Chen HY, Hu XL, Pan YY, Chen YS, Li HR. psk1 virulence gene-induced pulmonary and systemic tuberculosis in a young woman with normal immune function: A case report. World J Clin Cases 2024; 12:6826-6833. [DOI: 10.12998/wjcc.v12.i35.6826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 07/02/2024] [Accepted: 07/23/2024] [Indexed: 10/24/2024] Open
Abstract
BACKGROUND Tuberculosis is a chronic infectious disease and an important public health problem. Despite progress in controlling tuberculosis, the incidence of tuberculosis in China is still very high, with 895000 new cases annually. This case report describes the investigation of a case of severe disseminated tuberculosis in a young adult with normal immune function, conducted to ascertain why a Mycobacterium tuberculosis (M. tuberculosis) strain caused such severe disease.
CASE SUMMARY A previously healthy 28-year-old woman presented to our hospital with a 1-month history of fever and fatigue. She was diagnosed with severe disseminated pulmonary tuberculosis, spinal tuberculosis with paravertebral abscesses, and tuberculous meningitis. M. tuberculosis was isolated from bronchoalveolar lavage fluid. She was treated with standard antituberculous therapy and underwent debridement, bone graft, and internal fixation surgery for spinal tuberculosis. She responded to therapy and regained her ability to walk following the surgery. We analysed the whole-genome sequence of the strain and designated it BLM-A21. Additional M. tuberculosis genomes were selected from the Virulence Factor Database (http://www.mgc.ac.cn/cgi-bin/VFs/genus.cgi?Genus=Mycobacterium) for comparison. An evolutionary tree of the BLM-A21 strain was built using PhyML maximum likelihood software. Further gene analysis revealed that, except for the pks1 gene, BLM-A21 had similar virulence genes to the CDC 1551 and H37Rv strains, which have lower dissemination.
CONCLUSION We speculate that the pks1 virulence gene in BLM-A21 may be the key virulence gene responsible for the widespread dissemination of M. tuberculosis infection in this previously healthy adult with normal immune function.
Collapse
Affiliation(s)
- Fan Wu
- Department of Respiratory and Critical Care Medicine, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian Province, China
| | - Bin Yang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yan Xiao
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Li-Li Ren
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hong-Yi Chen
- Emergency Department, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian Province, China
| | - Xin-Lan Hu
- Clinical Microbiology Laboratory, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian Province, China
| | - Yan-Yu Pan
- Infection Department, The 900th Hospital of the PLA Joint Support Force, Fuzhou 350001, Fujian Province, China
| | - Yu-Sheng Chen
- Department of Respiratory and Critical Care Medicine, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian Province, China
| | - Hong-Ru Li
- Department of Infectious Diseases, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
4
|
Wang H, Gu Y, Ju C, Li Y, Chen X, Zhou G, Zhang X, Liu C, Chen J, Han Y, Zhang J, Shao Z, Zhang M. Genetic characteristics and potential pathogenic agents in Campylobacter upsaliensis based on genomic analysis. Emerg Microbes Infect 2024; 13:2294857. [PMID: 38085548 PMCID: PMC10810667 DOI: 10.1080/22221751.2023.2294857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 12/11/2023] [Indexed: 01/26/2024]
Abstract
Campylobacter upsaliensis was the most common Campylobacter species in pets' gastrointestinal tracts and has been isolated from patients with bacteremia, hemolytic-uremic syndrome, spontaneous abortion, and Guillain-Barré syndrome. However, the genetic characteristics and the full extent of its significance as a human pathogen remain to be fully understood. This study involved an investigation for genomic analysis of 154 strains from different sources and additional antimicrobial resistance profiles of 26 strains for this species. The genomes contained 1,558-1,971 CDS and the genome sizes were estimated to vary from 1.53 Mb to 1.86 Mb, with an average GC content of 34.71%. The entire analyzed genomes could be divided into three clades (A, B, and C) based on ANI and phylogenomic analysis. Significantly, nearly all strains in Clade B were isolated from patient samples, and the virulence-related sequences FlgD, GmhA, and CdtC might serve as determining factors for the classification of Clade B. Half of the tested isolates had MIC values over 64 μg mL-1 for nalidixic acid, gentamicin, and streptomycin. Isolates from pets in China carried more resistant elements in the genomes. This study both provided a comprehensive profile of C. upsaliensis for its genomic features and suggested some pathogenic agents for human infection with this species.
Collapse
Affiliation(s)
- Hairui Wang
- 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, People’s Republic of China
| | - Yixin Gu
- 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, People’s Republic of China
| | - Changyan Ju
- Nanshan Center for Disease Control and Prevention, Shenzhen, People’s Republic of China
| | - Ying Li
- Shunyi District Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Xiaoli Chen
- 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, People’s Republic of China
| | - Guilan Zhou
- 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, People’s Republic of China
| | - Xin Zhang
- 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, People’s Republic of China
| | - Chang Liu
- 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, People’s Republic of China
| | - Jing Chen
- Meilianzhonghe Veterinary Hospital Companion Branch, Beijing, People’s Republic of China
| | - Yue Han
- Meilianzhonghe Veterinary Hospital Jingxi Branch, Beijing, People’s Republic of China
| | - Jianzhong Zhang
- 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, People’s Republic of China
| | - Zhujun Shao
- 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, People’s Republic of China
| | - Maojun Zhang
- 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, People’s Republic of China
| |
Collapse
|
5
|
Wang Q, Chen J, Qi W, Bai Y, Mao J, Qu J. Dam construction alters the traits of health-related microbes along the Yangtze River. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 953:176077. [PMID: 39244052 DOI: 10.1016/j.scitotenv.2024.176077] [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: 06/06/2024] [Revised: 08/23/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Dams, constructed globally for energy production and water conservation, fragment rivers, and modify flow regimes, thereby altering the composition of biological communities and ecosystem functions. Despite the extensive use of dams, few studies have explored their potential health impacts, particularly concerning changes in health-related genes, such as antibiotic resistance genes (ARGs) and virulence factor genes (VFGs), and their hosts (i.e., ARB and potential pathogens). Understanding these health-related effects is crucial because they can impact human health through water quality and pathogen prevalence. In this study, we investigated the planktonic microbial community in the Three Gorges Reservoir (TGR) and adjacent upstream and downstream areas of the Yangtze River during both the dry and wet season. Our metagenomic analysis showed that dam construction significantly decreased the abundance of ARGs, but it had an insignificant effect on VFGs. The observed reduction in ARGs abundance could be mainly attributed to the decrease abundance of the major ARGs carrier - Limnohabitansin the TGR and downstream areas due to high grazing pressure and fitness cost. Conversely, the abundance of microbes carrying VFGs (potential pathogens) remained stable from upstream to the dam reservoir, which may explain the negligible impact on VFG abundance. Overall, our results provide a detailed understanding of the ecological health implications of dam construction in large river ecosystems.
Collapse
Affiliation(s)
- Qiaojuan Wang
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Science, Beijing 100049, China; Yangtze Eco-Environment Engineering Research Center, China Three Gorges Corporation, Beijing 100038, China
| | - Junwen Chen
- Center for Water and Ecology, Tsinghua University, Beijing 100084, China
| | - Weixiao Qi
- Center for Water and Ecology, Tsinghua University, Beijing 100084, China
| | - Yaohui Bai
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Jie Mao
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Jiuhui Qu
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| |
Collapse
|
6
|
Ogundare ST, Fasina FO, Makumbi JP, van der Zel GA, Geertsma PF, Kock MM, Smith AM, Ehlers MM. Epidemiology and antimicrobial resistance profiles of pathogenic Escherichia coli from commercial swine and poultry abattoirs and farms in South Africa: A One Health approach. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175705. [PMID: 39181266 DOI: 10.1016/j.scitotenv.2024.175705] [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: 06/14/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Pathogenic Escherichia coli (PEC) are important foodborne bacteria that can cause severe illness in humans. The PECs thrive within the intestines of humans as well as animals and may contaminate multiple ecosystems, including food and water, via faecal transmission. Abattoir and farm employees are at high risk of PEC exposure, which could translate to community risk through person-to-person contact. To determine the epidemiology and resistome of PECs in Gauteng and Limpopo provinces of South Africa, 198 swine faecal samples, 220 poultry cloacal swabs, 108 human hand swabs, 11 run-off water samples from abattoirs and farms were collected from four swine and five poultry commercial abattoirs and two swine farms. One effluent sample each was collected from four wastewater treatment plants (WWTP) and a tertiary hospital setting. Phenotypic and genotypic techniques were used including polymerase chain reaction, pulsed-field gel electrophoresis (PFGE) and whole genome sequencing (WGS). Results showed EHEC and EPEC prevalence was 4.1 % (22/542) and 20.8 % (113/542), respectively, with the O26 serogroup detected the most in PEC isolates. According to the PFGE dendrogram, isolates from poultry, human hand swabs and run-off water clustered together. Diverse virulence factors such as the novel stx2k subtype and eae genes were detected among the 36 representative PEC isolates according to WGS. The results showed that 66.7 % (24/36) of sequenced PECs presented with multi-drug resistance (MDR) to β-lactamase 13.9 % (5/36), aminoglycoside 61.1 % (22/36), tetracycline 41.7 % (15/36) and quinolones 38.9 % (14/36). No colistin nor carbapenem resistance was detected. Sequence types (STs) associated with MDR in this study were: ST752, ST189, ST206, ST10, ST48 and ST38. The findings highlight the threat of zoonotic pathogens to close human contacts and the need for enhanced surveillance to mitigate the spread of MDR foodborne PECs.
Collapse
Affiliation(s)
- Samuel T Ogundare
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| | - Folorunso O Fasina
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa; Food and Agriculture Organisation of the United Nations, FAO Headquarters, Rome, Italy
| | - John-Paul Makumbi
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Gerbrand A van der Zel
- Gauteng Department of Agriculture, Rural Development and Environment, Pretoria, South Africa
| | - Peter F Geertsma
- Gauteng Department of Agriculture, Rural Development and Environment, Pretoria, South Africa
| | - Marleen M Kock
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Department of Medical Microbiology, National Health Laboratory Service, Tshwane Academic Division, Pretoria, South Africa
| | - Anthony M Smith
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Centre for Enteric Diseases, National Institute for Communicable Diseases, Division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Marthie M Ehlers
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Department of Medical Microbiology, National Health Laboratory Service, Tshwane Academic Division, Pretoria, South Africa
| |
Collapse
|
7
|
Ananna NT, Shishir TA, Ahmed A, Al Sium SM, Shakil MS, Haque FKM, Hasanuzzaman M. Characterization of two lytic bacteriophages infecting carbapenem-resistant clinical Klebsiella pneumoniae in Dhaka, Bangladesh. Virus Res 2024; 350:199491. [PMID: 39491772 DOI: 10.1016/j.virusres.2024.199491] [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: 06/14/2024] [Revised: 10/03/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Bacteriophages or bacteria infecting viruses are genetically diverse. Due to the emergence of antimicrobial-resistant bacteria, lytic bacteriophages are gaining enormous attention for treating superbug infections. Klebsiella pneumoniae is one of the eight most significant nosocomial pathogens and is addressed as a critical priority pathogen by WHO, requiring alternative treatment options. We reported two highly lytic bacteriophages, Klebsiella phage Kpn BM7 and the novel Klebsiella phage Kpn BU9, isolated from hospital wastewater and exhibiting lytic activity against different clinical isolates. Whole-genome analysis revealed that phages BM7 and BU9 belong to class Caudoviricetes. Phage BM7, with a genome length of 170,558 bp, is a member of the genus Marfavirus and the species Marfavirus F48. While phage BU9, with a genome length of 60,450 bp, remains unclassified. Neither phage harbors any lysogenic, toxin, or antimicrobial resistance genes. Both phages can steadily survive up to 40 °C and at pH 5-7. The optimal MOI was 0.1 for BM7 and 1 for BU9, with short latent periods of 10 and 25 min and burst sizes of 85 PFU/cell and 12 PFU/cell, respectively. This is the first carbapenem-resistant K. pneumoniae targeting lytic phages to be reported from Bangladesh. This study suggests that BM7 and BU9 are potential candidates for targeting carbapenem-resistant K. pneumoniae.
Collapse
Affiliation(s)
- Nishat Tasnim Ananna
- Biotechnology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | - Tushar Ahmed Shishir
- Biotechnology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | - Akash Ahmed
- Microbiology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | | | - Md Salman Shakil
- Microbiology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | - Fahim Kabir Monjurul Haque
- Microbiology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | - Md Hasanuzzaman
- Microbiology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh.
| |
Collapse
|
8
|
Dong Y, Zhang Y, Xu F, Zou K. Extensive genomic characterization, pre-clinical probiotic evaluation, and safety analysis of Bifidobacterium longum subsp. longum BL21 isolated from infant feces. Microb Pathog 2024; 197:107100. [PMID: 39505088 DOI: 10.1016/j.micpath.2024.107100] [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: 05/12/2024] [Revised: 10/03/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE This study aimed to evaluate the safety and probiotic properties of Bifidobacterium longum subsp. longum BL21 isolated from infant feces for use as a commercial probiotic strain. METHODS Whole-genome sequencing; physiological and biochemical assessments; enzymatic assays; metabolite, antibiotic sensitivity, cell adhesion and cytotoxicity, and tolerance tests; and a 14-day oral toxicity study were conducted. RESULTS BL21 exhibited genetic integrity, and its genome lacked genes related to antibiotic resistance or virulence. It was found to be non-pathogenic, had efficient carbohydrate metabolism and mucin degradation ability, and was free from biogenic amines. It also showed susceptibility to antibiotics, strong cell adhesion, and resilience to adverse conditions. The aforementioned results confirm that BL21 is a functional probiotic strain with genetic stability, enzymatic capabilities, and non-pathogenic properties that mean it is safe for oral consumption, demonstrating that it is a promising candidate for probiotic applications. CONCLUSION The study demonstrates that BL21 is a genetically stable, non-pathogenic probiotic strain with metabolic potential. The strain lacks virulence and antibiotic resistance genes, and its resilience to gastrointestinal conditions, as well as the results of the 14-day oral toxicity study, suggest that BL21 is safe for oral consumption. However, further long-term studies and clinical trials are needed to confirm its safety and efficacy for therapeutic use.
Collapse
Affiliation(s)
- Yao Dong
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yinan Zhang
- Shanghai Institute of Quality Inspection and Technical Research, Shanghai, 200233, China
| | - Fei Xu
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, China
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
9
|
Wu Y, Wu Z, Gao Y, Fan Y, Dong Y, Zhang Y, Gai Z, Gu S. Comprehensive genomic analysis and evaluation of in vivo and in vitro safety of Heyndrickxia coagulans BC99. Sci Rep 2024; 14:26602. [PMID: 39496841 PMCID: PMC11535476 DOI: 10.1038/s41598-024-78202-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: 08/09/2024] [Accepted: 10/29/2024] [Indexed: 11/06/2024] Open
Abstract
This study aimed to evaluate the safety profile and beneficial effects of Heyndrickxia coagulans strain BC99 (BC99) for potential use in functional foods and pharmaceuticals. We began with whole genome sequencing of BC99, followed by a comprehensive safety assessment comprising genome analysis, hemolysis, cytotoxicity, antibiotic susceptibility tests, and cell adhesion and tolerance studies, along with acute and subacute oral toxicity studies in animal models. BC99 was isolated from a well-characterized collection originating from the feces of a healthy infant. Our results indicated no hemolytic activity on Columbia blood agar plates and broad antibiotic sensitivity, including to gentamicin, ampicillin, chloramphenicol, ciprofloxacin, and others. Cytotoxicity testing confirmed no adverse effects on HT-29 cells and significant adhesive properties to intestinal epithelial cells. Tolerance tests demonstrated over 90% viability of BC99 under simulated gastrointestinal conditions. In vivo studies in mice and rats confirmed the absence of adverse effects following oral administration. Collectively, these findings support BC99's robust tolerance to gastrointestinal environments, strong adhesion capabilities, and a broad spectrum of antibiotic resistance, underlining its potential as a safe and effective agent for gut microbiota modulation and host health enhancement.
Collapse
Affiliation(s)
- Ying Wu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, P.R. China
| | - Zhiyi Wu
- Department of Research and Development, Henan Animic Biotechnology Co., Ltd, Henan, China
| | - Yinyin Gao
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, P.R. China
| | - Yixuan Fan
- Department of Research and Development, Henan Animic Biotechnology Co., Ltd, Henan, China
| | - Yao Dong
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yinan Zhang
- Shanghai Institute of Quality Inspection and Technical Research, Shanghai, 200233, China
| | - Zhonghui Gai
- Department of Research and Development, Henan Animic Biotechnology Co., Ltd, Henan, China.
| | - Shaobin Gu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, P.R. China.
| |
Collapse
|
10
|
Yoshida H, Goto M, Tsuyuki Y, Kim JS, Takahashi T. Streptococcus canis transcriptomic modifications in host cell entry environments of human keratinocytes. BMC Genomics 2024; 25:1028. [PMID: 39497051 PMCID: PMC11533360 DOI: 10.1186/s12864-024-10974-z] [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: 08/06/2024] [Accepted: 10/29/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Streptococcus canis is a commensal bacterium in companion animals. This microorganism can infect humans who have been in deep contact with or bitten by pet dogs, suggesting that the skin/soft tissue is one of infection entry sites. To understand pathological process in human cells, we aimed to determine S. canis transcriptomic changes in invasive environments of human keratinocytes. METHODS We selected one isolate from candidates with whole-genome sequences, based on re-obtained cell invasion ability (CIA) data into human keratinocytes along with bacterial cytotoxicity. RNA-sequencing was conducted for the samples at baselines and 2 h/5 hr post-inoculation using NovaSeq 6000. Global/differential gene expression analyses [principal component analysis (PCA)/k-means clustering analysis/differentially expressed gene (DEG) analyses] were performed. We classified DEGs into their functional categories. To validate transcriptomic results, we did quantitative reverse transcription-polymerase chain reaction (qRT-PCR) assays. RESULTS FU1 isolate was selected from seven candidates, based on re-obtained CIA data with less cytotoxicity. Total read bases of 6.17-9.02 Gbp were obtained by RNA-sequencing. PCA and k-means clustering analysis indicated clustering according to their inoculation times. Volcano plots and Venn diagrams revealed that S. canis invasion into keratinocytes produced altered distributions of many genes. Gene ontology enrichment analysis showed most of the gene expressions were downregulated. DEG functional analysis showed the downregulated DEGs belonging to energy production and conversion/carbohydrate transport and metabolism/amino acid transport and metabolism/nucleotide transport and metabolism, with the upregulated DEGs belonging to transcription. qRT-PCR assays for downregulated/upregulated expressions of four genes (pgk-slo/opuAA-kdpB) validated transcriptomic results. CONCLUSION Our observations suggest that S. canis can downregulate its metabolism-associated gene expressions in human keratinocyte environments. The observed gene expression changes can imply the latent infection in human cells. Further investigation is needed to elucidate the underlying mechanisms for the latent infection.
Collapse
Affiliation(s)
- Haruno Yoshida
- Laboratory of Infectious Diseases, Graduate School of Infection Control Sciences and Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Mieko Goto
- Laboratory of Infectious Diseases, Graduate School of Infection Control Sciences and Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Yuzo Tsuyuki
- Laboratory of Infectious Diseases, Graduate School of Infection Control Sciences and Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
- Division of Clinical Laboratory, Sanritsu Zelkova Veterinary Laboratory, Tokyo, Japan
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Takashi Takahashi
- Laboratory of Infectious Diseases, Graduate School of Infection Control Sciences and Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan.
| |
Collapse
|
11
|
Gao FZ, Hu LX, Liu YS, Qiao LK, Chen ZY, Su JQ, He LY, Bai H, Zhu YG, Ying GG. Unveiling the overlooked small-sized microbiome in river ecosystems. WATER RESEARCH 2024; 265:122302. [PMID: 39178591 DOI: 10.1016/j.watres.2024.122302] [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: 06/25/2024] [Revised: 08/12/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024]
Abstract
Enriching microorganisms using a 0.22-μm pore size is a general pretreatment procedure in river microbiome research. However, it remains unclear the extent to which this method loses microbiome information. Here, we conducted a comparative metagenomics-based study on microbiomes with sizes over 0.22 μm (large-sized) and between 0.22 μm and 0.1 μm (small-sized) in a subtropical river. Although the absolute concentration of small-sized microbiome was about two orders of magnitude lower than that of large-sized microbiome, sequencing only large-sized microbiome resulted in a significant loss of microbiome diversity. Specifically, the microbial community was different between two sizes, and 347 genera were only detected in small-sized microbiome. Small-sized microbiome had much more diverse viral community than large-sized fraction. The viruses had abundant ecological functions and were hosted by 825 species of 169 families, including pathogen-related families. Small-sized microbiome had distinct antimicrobial resistance risks from large-sized microbiome, showing an enrichment of eight antibiotic resistance gene (ARG) types as well as the detection of 140 unique ARG subtypes and five enriched risk rank I ARGs. Draft genomes of five major resistant pathogens having diverse ecological and pollutant-degrading functions were only assembled in small-sized microbiome. These findings provide novel insights into river ecosystems, and highlight the overlooked small-sized microbiome in the environment.
Collapse
Affiliation(s)
- Fang-Zhou Gao
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical, Chemistry of Environment, South China Normal University, Guangzhou, China; School of Environment, South China Normal University, University Town, Guangzhou, China
| | - Li-Xin Hu
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical, Chemistry of Environment, South China Normal University, Guangzhou, China; School of Environment, South China Normal University, University Town, Guangzhou, China
| | - You-Sheng Liu
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical, Chemistry of Environment, South China Normal University, Guangzhou, China; School of Environment, South China Normal University, University Town, Guangzhou, China
| | - Lu-Kai Qiao
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical, Chemistry of Environment, South China Normal University, Guangzhou, China; School of Environment, South China Normal University, University Town, Guangzhou, China
| | - Zi-Yin Chen
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical, Chemistry of Environment, South China Normal University, Guangzhou, China; School of Environment, South China Normal University, University Town, Guangzhou, China
| | - Jian-Qiang Su
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Liang-Ying He
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical, Chemistry of Environment, South China Normal University, Guangzhou, China; School of Environment, South China Normal University, University Town, Guangzhou, China
| | - Hong Bai
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical, Chemistry of Environment, South China Normal University, Guangzhou, China; School of Environment, South China Normal University, University Town, Guangzhou, China
| | - Yong-Guan Zhu
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Guang-Guo Ying
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical, Chemistry of Environment, South China Normal University, Guangzhou, China; School of Environment, South China Normal University, University Town, Guangzhou, China.
| |
Collapse
|
12
|
Yan Z, Li Y, Ni Y, Xia X, Zhang Y, Wu Y, Zhang J, Chen G, Li R, Zhang R. Plasmid-borne tigecycline resistance gene tet(X4) in Salmonella enterica and Escherichia coli isolates from a pediatric patient with diarrhea. Drug Resist Updat 2024; 77:101145. [PMID: 39226862 DOI: 10.1016/j.drup.2024.101145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024]
Affiliation(s)
- Zelin Yan
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yan Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Yingling Ni
- Clinical Microbiology Laboratory, Laboratory of Yuhuan Traditional Chinese Medicine Hospital, Taizhou, Zhejiang 317600, China
| | - Xiaoni Xia
- Laboratory Department of the First People's Hospital of Daishan County, Zhoushan, Zhejiang 316200, China
| | - Yanyan Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yuchen Wu
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Jing Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Gongxiang Chen
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Ruichao Li
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225000, China.
| | - Rong Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
13
|
Jarocki VM, Li D, Bogema DR, Yam J, Jenkins C, Hai FI, Djordjevic SP. Comparative genomic analysis of ESBL-selected and non-selected Escherichia coli in Australian wastewater: Elucidating differences in diversity, antimicrobial resistance, and virulence profiles. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 949:175079. [PMID: 39094658 DOI: 10.1016/j.scitotenv.2024.175079] [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: 03/13/2024] [Revised: 07/07/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
Extended-spectrum β-lactamases (ESBLs)-producing E. coli have been proposed as an indicator bacterium for antimicrobial resistance (AMR) surveillance within a OneHealth framework. However, it is important to understand the effects and potential biases ESBL-selection has on E. coli populations. Utilising whole genome sequencing, this study compared 80 ESBL-selected E. coli isolates with 201 non-selected isolates from Australian wastewater. The findings revealed significant variations between these cohorts in genetic diversity, AMR profiles, and carriage of virulence-associated genes (VAGs), plasmids, and the transmissible Locus of Stress Tolerance (tLST), a genomic island that imparts resistance to extreme heat and chlorination. The study highlights the predominance of certain sequence types (STs), particularly ST131 (75 % clade A), in ESBL-selected isolates (40 % vs 2 %) and overall the ESBL-selected isolates were largely multidrug-resistant (MDR), predominantly carrying genes for resistance to aminoglycosides, extended-spectrum β-lactams, fluoroquinolone, macrolides, sulphonamides/trimethoprim, and tetracyclines. The ESBLs identified were almost exclusively blaCTX-M genes, most commonly blaCTX-M-15 > blaCTX-M-27 > blaCTX-M-14. These were predominately carried on IncF plasmids or chromosomally (always ISEcp1 associated), in equal numbers. In contrast, 80 % of non-selected isolates carried no acquired ARGs, and none carried blaCTX-M genes. In both cohorts, extraintestinal pathogenic E. coli (ExPEC) was the dominate pathotype (35 % total) with few (4 % total) intestinal pathogenic E. coli pathotypes identified (aEPEC > ETEC > EAEC). Nevertheless, some clinically important genes were only identified in the non-selected group, namely tigecycline-resistance gene tet(X4) and AmpC ESBL blaCMY-2. Additionally, the presence of tLST, associated with higher metal resistance gene carriage (Ag, As, Cu, Hg, Ni), in a substantial portion of non-selected isolates (20 % vs 0 %), underscores environmental pressures shaping bacterial populations in wastewater ecosystems. These insights are important for developing comprehensive, less biased genomic surveillance strategies to understand and manage public health threats posed by pathogenic E. coli and AMR.
Collapse
Affiliation(s)
- Veronica M Jarocki
- Australian Institute for Microbiology & Infection, University of Technology Sydney, NSW, Australia; Australian Centre for Genomic Epidemiological Microbiology, University of Technology Sydney, NSW, Australia; Cooperative Research Centre for Solving Antimicrobial resistance in Agribusiness, Food and Environment (CRC SAAFE), South Australia, Australia.
| | - Dmitriy Li
- Australian Institute for Microbiology & Infection, University of Technology Sydney, NSW, Australia; Australian Centre for Genomic Epidemiological Microbiology, University of Technology Sydney, NSW, Australia
| | - Daniel R Bogema
- Australian Centre for Genomic Epidemiological Microbiology, University of Technology Sydney, NSW, Australia; Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries, NSW, Australia
| | - Jerald Yam
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries, NSW, Australia
| | - Cheryl Jenkins
- Australian Centre for Genomic Epidemiological Microbiology, University of Technology Sydney, NSW, Australia; Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries, NSW, Australia
| | - Faisal I Hai
- School of Civil, Mining, Environmental and Architectural Engineering, University of Wollongong, NSW, Australia
| | - Steven P Djordjevic
- Australian Institute for Microbiology & Infection, University of Technology Sydney, NSW, Australia; Australian Centre for Genomic Epidemiological Microbiology, University of Technology Sydney, NSW, Australia.
| |
Collapse
|
14
|
Garrido-Palazuelos LI, Almanza-Orduño AA, Waseem M, Basheer A, Medrano-Félix JA, Mukthar M, Ahmed-Khan H, Shahid F, Aguirre-Sánchez JR. Immunoinformatic approach for multi-epitope vaccine design against Staphylococcus aureus based on hemolysin proteins. J Mol Graph Model 2024; 132:108848. [PMID: 39182254 DOI: 10.1016/j.jmgm.2024.108848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/09/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Staphylococcus aureus is a common bacterium that causes a variety of infections in humans. This microorganism produces several virulence factors, including hemolysins, which contribute to its disease-causing ability. The treatment of S. aureus infections typically involves the use of antibiotics. However, the emergence of antibiotic-resistant strains has become a major concern. Therefore, vaccination against S. aureus has gained attention as an alternative approach. Vaccination has the advantage of stimulating the immune system to produce specific antibodies that can neutralize bacteria and prevent infection. However, developing an effective vaccine against S. aureus has proven to be challenging. This study aimed to use in silico methods to design a multi-epitope vaccine against S. aureus infection based on hemolysin proteins. The designed vaccine contained four B-cell epitopes, four CTL epitopes, and four HTL epitopes, as well as the ribosomal protein L7/L12 and pan-HLA DR-binding epitope, included as adjuvants. Furthermore, the vaccine was non-allergenic and non-toxic with the potential to stimulate the TLR2-, TLR-4, and TLR-6 receptors. The predicted vaccine exhibited a high degree of antigenicity and stability, suggesting potential for further development as a viable vaccine candidate. The population coverage of the vaccine was 94.4 %, indicating potential widespread protection against S. aureus. Overall, these findings provide valuable insights into the design of an effective multi-epitope vaccine against S. aureus infection and pave the way for future experimental validations.
Collapse
Affiliation(s)
- Lennin Isaac Garrido-Palazuelos
- Universidad Autónoma de Occidente, Unidad Regional Los Mochis. Departamento Académico de Ciencias de la Salud. Blvd. Macario Gaxiola y Carretera Internacional, México 15, C.P. 81223, Los Mochis, Sinaloa, Mexico
| | - Arath Andrés Almanza-Orduño
- Universidad Autónoma de Occidente, Unidad Regional Los Mochis. Departamento Académico de Ciencias de la Salud. Blvd. Macario Gaxiola y Carretera Internacional, México 15, C.P. 81223, Los Mochis, Sinaloa, Mexico
| | - Maaz Waseem
- Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan; School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Amina Basheer
- Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan; Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - José Andrés Medrano-Félix
- Investigadoras e investigadores por México Centro de Investigación En Alimentación y Desarrollo A.C. Laboratorio Nacional para la Investigación en Inocuidad Alimentaria. Carretera a El Dorado km 5.5, Campo El Diez, 80110, Culiacán, Sinaloa, Mexico
| | - Mamuna Mukthar
- Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Haris Ahmed-Khan
- Department of Biotechnology, University of Mianwali, Punjab, 42200, Pakistan
| | - Fatima Shahid
- Department of Applied Physics, Faculty of Science & Technology, National University of Malaysia (UKM), Selangor Malaysia, Malaysia
| | - José Roberto Aguirre-Sánchez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA). Centro de Investigación en ALimentación y Desarrollo A.C. (CIAD) Unidad Culiacán, Sinaloa, México.
| |
Collapse
|
15
|
Ötkün S, Erdenliğ Gürbi Lek S. Whole-genome sequencing-based analysis of Brucella species isolated from ruminants in various regions of Türki̇ye. BMC Infect Dis 2024; 24:1220. [PMID: 39472798 PMCID: PMC11524016 DOI: 10.1186/s12879-024-09921-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/11/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Brucellosis, a zoonotic disease in Türkiye, which has significant direct and indirect impacts on the healthcare system and livestock. This study, which aimed to investigate the differences among Brucella spp. isolates originating from different regions of Türkiye, for implications for public health and veterinary medicine. METHOD Twenty-one isolates from ruminants and two isolates from humans obtained from various regions of Türkiye were utilized in the study. The isolates were identified and biotyped using traditional microbiological procedures, and whole-genome sequencing (WGS) was performed. This was followed by single nucleotide polymorphism (SNP)--based phylogenetic analysis and WGS-based analysis of virulence and resistance genes. Additionally, phenotypic antimicrobial resistance and phage susceptibilities were determined. The obtained data were then compared for concordance, ensuring the validity and reliability of the results. RESULTS Our study, employing culture methods, polymerase chain reaction (PCR), and WGS analyses, identified 11 Brucella melitensis (bv 3 (n = 9), one each bv 1 and bv 2) and 12 B. abortus (bv 3 (n = 11), bv 9 (n = 1)) isolates All B. abortus isolates were of bovine origin, while the B. melitensis isolates were from sheep (n = 7), goat (n = 1), ram (n = 1), and humans (n = 2). In the whole-genome SNP-based phylogenetic tree, all B. melitensis strains were found to be of the IIb subtype of genotype II associated with the Eastern Mediterranean lineage. Ten different genotypes were identified in the SNP analysis of the isolates, with a maximum SNP difference of 278 and a minimum SNP difference of 4 among these genotypes. According to the WGS-SNP-based phylogenetic tree of B. abortus isolates, they were grouped in clade C1. In the SNP analysis, where ten different genotypes were identified, the SNP difference among these genotypes was a maximum of 316 and a minimum of 6. In the in silico MLST analysis performed with WGS data, B. melitensis isolates were identified as ST8 and ST102 genotypes, while B. abortus isolates were identified as ST2 and ST3 genotypes. The dominant genotypes were ST8 for B. melitensis and ST2 for B. abortus, respectively. Virulence gene analysis conducted based on WGS data of the 23 B. abortus and B. melitensis isolates revealed 43 virulence gene-associated regions in all strains, irrespective of species, host, or isolation year. Although classical resistance-related genes were not detected by WGS-based antimicrobial resistance gene analysis, phenotypic resistance analysis revealed resistance to azithromycin, rifampin, and trimethoprim/sulfamethoxazole in B. abortus and B. melitensis isolates. CONCLUSION Both B. melitensis and B. abortus were circulating species in animals and human. The dominant genotypes were ST8 for B. melitensis and ST2 for B. abortus, respectively. All B. melitensis strains were found to be of the IIb subtype of genotype II associated with the Eastern Mediterranean lineage, while B. abortus isolates, they were grouped in clade C1. Further, a comprehensive study with a sufficient number of isolates covering all regions of Türkiye would provide more accurate information about the current epidemiological situation in the country.
Collapse
Affiliation(s)
- Songül Ötkün
- Departman of Veterinary Microbiology, Fakulty of Veterinary Medicine, Siirt University, Siirt, Türkiye.
| | - Sevil Erdenliğ Gürbi Lek
- Departman of Veterinary Microbiology, Fakulty of Veterinary Medicine, Harran University, Şanlıurfa, Türkiye
| |
Collapse
|
16
|
Maghembe RS, Magulye MA, Makaranga A, Nsubuga G, Sekyanzi S, Moto EA, Mwesigwa S, Katagirya E. Metagenome mining divulges virulent and multidrug resistant Pseudomonas aeruginosa ST242 and Klebsiella michiganensis ST∗1b23 coinfecting an 8-month-old meningitis infant under ICU in Kampala, Uganda, East Africa. Heliyon 2024; 10:e39455. [PMID: 39498086 PMCID: PMC11532849 DOI: 10.1016/j.heliyon.2024.e39455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024] Open
Abstract
Pediatric meningitis is a global health problem, with insufficiently known pathogens and antibiotic resistance (AMR) especially in low-resource settings. Here, we sought to uncover the virulence and AMR of pathogens associated with infant meningitis, treated with ceftriaxone, in Kampala, Uganda. In a bid to isolate Klebsiella oxytoca, we coincidentally recovered a co-culture and challenged it with antibiotic susceptibility testing (AST) on a panel of 14 antibiotics. We then combined metagenome binning with antiSMASH/PRISM genome mining to unveil the pathogens, their virulence and molecular targets in relation to meningitis. From AST, the co-culture exhibited resistance to multiple aminoglycosides, fluroquinolones, and β-lactams, including ceftriaxone, the inherently used drug. From metagenome annotation, the first bin was identified as Pseudomonas aeruginosa ST242 and the second as Klebsiella michiganensis ST∗1b23. Among others, P. aeruginosa ST242 virulence factors include type 3 and type 6 secretion systems, biofilm, and nonribosomal peptides (NRPs) of the pyoverdine synthase operon, targeting claudin-5, a component of the tight junctions of the blood-brain barrier (BBB). The P. aeruginosa ST242 genome portrays intrinsic resistance to beta-lactamases (blaOXA-50 and blaPAO), aminoglycosides [aph(3')-IIb)], fluoroquinolones (crpP), tetracycline (tmexD2) and fosfomycin (fosA), among others. From K. michiganensis ST∗1b23 genome mining we elucidated a yersiniabactin-related metabolite, targeting the ligand-binding domain of the human polymeric immunoglobulin receptor (pIgR) and other components of the BBB. The K. michiganensis ST∗1b23 chromosome encodes the genes blaOXY-1 and OqxA/B, conferring resistance to β-lactams, fluoroquinolones, and trimethoprim respectively. Notably, we found one frameshift and nine substitution mutations conferring carbapenem and cephalosporin resistance mechanisms. Overall, our findings strongly suggest coinfection and a mechanistic crosstalk between P. aeruginosa ST242 and K. michiganensis ST∗1b23 in the pathogenesis of meningitis in this case. Importantly, ceftriaxone could be an inappropriate treatment choice for these pathogens. Hence, serious surveillance and experimental studies will improve the management of pediatric meningitis.
Collapse
Affiliation(s)
- Reuben S. Maghembe
- Department of Microbiology and Parasitology, Faculty of Medicine, St. Francis University College of Health and Allied Sciences (SFUCHAS), P. O. Box 175, Ifakara, Tanzania
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- Department of Microbiology and Immunology, Faculty of Biomedical Sciences, Kampala International University-Western Campus (KIU-WC), Ishaka, Bushenyi, Uganda
- Department of Health and Biomedical Sciences, Didia Education and Health Organization (DEHO), P. O. Box 113, Shinyanga, Tanzania
| | - Maximilian A.K. Magulye
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- Department of Health and Biomedical Sciences, Didia Education and Health Organization (DEHO), P. O. Box 113, Shinyanga, Tanzania
- Department of Medical Microbiology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Abdalah Makaranga
- Department of Health and Biomedical Sciences, Didia Education and Health Organization (DEHO), P. O. Box 113, Shinyanga, Tanzania
| | - Gideon Nsubuga
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Simon Sekyanzi
- Department of Medical Microbiology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Edward A. Moto
- Department of Biology, College of Natural and Mathematical Sciences, University of Dodoma, Dodoma, Tanzania
| | - Savannah Mwesigwa
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Eric Katagirya
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
17
|
Andriyanov P, Zhurilov P, Menshikova A, Tutrina A, Yashin I, Kashina D. Large-scale genomic analysis of Elizabethkingia anophelis. BMC Genomics 2024; 25:1015. [PMID: 39472795 PMCID: PMC11523902 DOI: 10.1186/s12864-024-10921-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
The recent emergence of Elizabethkingia anophelis as a human pathogen is a major concern for global public health. This organism has the potential to cause severe infections and has inherent antimicrobial resistance. The potential for widespread outbreaks and rapid global spread highlights the critical importance of understanding the biology and transmission dynamics of this infectious agent. We performed a large-scale analysis of available 540 E. anophelis, including one novel strain isolated from raw milk and sequenced in this study. Pan-genome analysis revealed an open and diverse pan-genome in this species, characterized by the presence of many accessory genes. This suggests that the species has a high level of adaptability and can thrive in a variety of environments. Phylogenetic analysis has also revealed a complex population structure, with limited source-lineage correlation. We identified diverse antimicrobial resistance factors, including core-genome and accessory ones often associated with mobile genetic elements within specific lineages. Mobilome analysis revealed a dynamic landscape primarily composed of genetic islands, integrative and conjugative elements, prophage elements, and small portion of plasmids emphasizing a complex mechanism of horizontal gene transfer. Our study underscores the adaptability of E. anophelis, characterized by a diverse range of antimicrobial resistance genes, putative virulence factors, and genes enhancing fitness. This adaptability is also supported by the organism's ability to acquire genetic material through horizontal gene transfer, primarily facilitated by mobile genetic elements such as integrative and conjugative elements (ICEs). The potential for rapid evolution of this emerging pathogen poses a significant challenge to public health efforts.
Collapse
Affiliation(s)
- Pavel Andriyanov
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia.
| | - Pavel Zhurilov
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Alena Menshikova
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Anastasia Tutrina
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Ivan Yashin
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Daria Kashina
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
18
|
Zhou S, Liu B, Zheng D, Chen L, Yang J. VFDB 2025: an integrated resource for exploring anti-virulence compounds. Nucleic Acids Res 2024:gkae968. [PMID: 39470738 DOI: 10.1093/nar/gkae968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/07/2024] [Accepted: 10/24/2024] [Indexed: 11/01/2024] Open
Abstract
With the escalating crisis of bacterial multidrug resistance, anti-virulence therapeutic strategies have emerged as a highly promising alternative to conventional antibiotic treatments. Anti-virulence compounds are specifically designed to target virulence factors (VFs), disarming pathogens without affecting bacterial growth and thus reduce the selective pressure for resistance development. However, due to the complexity of bacterial pathogenesis, no anti-virulence small molecules have been approved for clinical use thus far, despite the documentation of hundreds of potential candidates. To provide valuable reference resources for drug design, repurposing, and target selection, the virulence factor database (VFDB, http://www.mgc.ac.cn/VFs/) has systematically collected public data on anti-virulence compounds through extensive literature mining, and further integrated this information with its existing knowledge of bacterial VFs. To date, the VFDB has curated a comprehensive dataset of 902 anti-virulence compounds across 17 superclasses reported by 262 studies worldwide. By cross-linking the current knowledge of bacterial VFs with information on relevant compounds (e.g. classification, chemical structure, molecular targets and mechanisms of action), the VFDB aims to bridge the gap between chemists and microbiologists, providing crucial insights for the development of innovative and effective antibacterial therapies to combat bacterial infections and address antibiotic resistance.
Collapse
Affiliation(s)
- Siyu Zhou
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Bo Liu
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Dandan Zheng
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Lihong Chen
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Jian Yang
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| |
Collapse
|
19
|
Wen J, Zhang H, Chu D, Chen X, Feng J, Wang Y, Liu G, Zhang Y, Li Y, Ning K. Deep learning revealed the distribution and evolution patterns for invertible promoters across bacterial lineages. Nucleic Acids Res 2024:gkae966. [PMID: 39460615 DOI: 10.1093/nar/gkae966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/26/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Invertible promoters (invertons) are crucial regulatory elements in bacteria, facilitating gene expression changes under stress. Despite their importance, their prevalence and the range of regulated gene functions are largely unknown. We introduced DeepInverton, a deep learning model that identifies invertons across a broad phylogenetic spectrum without using sequencing reads. By analyzing 68 733 bacterial genomes and 9382 metagenomes, we have uncovered over 200 000 nonredundant invertons and have also highlighted their abundance in pathogens. Additionally, we identified a post-Cambrian Explosion increase of invertons, paralleling species diversification. Furthermore, we revealed that invertons regulate diverse functions, including antimicrobial resistance and biofilm formation, underscoring their role in environmental adaptation. Notably, the majority of inverton identifications by DeepInverton have been confirmed by the in vitro experiments. The comprehensive inverton profiles have deepened our understanding of invertons at pan-genome and pan-metagenome scales, enabling a broad spectrum of applications in microbial ecology and synthetic biology.
Collapse
Affiliation(s)
- Jiejie Wen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Haobo Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Dongliang Chu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xiaoke Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jingru Feng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yucen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Guanxi Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yuhao Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yuxue Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Kang Ning
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
20
|
Vinhal ALDO, de Araújo MRB, Rodrigues EB, Castro DLDC, Pereira CR, Custódio DAC, Dorneles EMS, Aburjaile FF, Brenig B, Azevedo V, Viana MVC. First comparative genomics analysis of Corynebacterium auriscanis. Mem Inst Oswaldo Cruz 2024; 119:e240156. [PMID: 39476150 PMCID: PMC11508509 DOI: 10.1590/0074-02760240156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/05/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Corynebacterium auriscanis is a bacterial species frequently isolated from dogs with external otitis or dermatitis and a zoonotic pathogen transmitted by dog bite. It is considered an opportunistic pathogen, but its pathogenicity mechanisms are poorly studied. Comparative genomics can identify virulence and niche factors that could contribute to understanding its lifestyle. OBJECTIVES The objectives of this project was to compare genomes of C. auriscanis to identify genes related to its virulence and lifestyle. METHODS The genome of strain 32 was sequenced using Illumina HiSeq 2500 (Illumina, CA, USA) and assembled using Unicycler. The two other non-redundant genomes from the same species available in GenBank were included in the analysis. All genomes were annotated and checked for taxonomy, assembly quality, mobile elements, CRISPR-Cas systems, and virulence and antimicrobial resistance genes. The virulence genes in the three genomes were compared to the ones from other pathogens commonly isolated with C. auriscanis. FINDINGS The species has 42 virulence factors that can be classified as niche factors, due to the absence of true virulence factors found in primary pathogens. The gene rbpA could confer basal levels of resistance to rifampin. MAIN CONCLUSIONS The absence of true virulence factors in the three genomes suggests C. auriscanis has an opportunistic pathogen lifestyle.
Collapse
Affiliation(s)
- Ana Lua de Oliveira Vinhal
- Universidade Federal de Minas Gerais, Departamento de Genética, Ecologia e Evolução, Belo Horizonte, MG, Brasil
| | - Max Roberto Batista de Araújo
- Universidade Federal de Minas Gerais, Departamento de Genética, Ecologia e Evolução, Belo Horizonte, MG, Brasil
- Instituto Hermes Pardini-Grupo Fleury, Microbiologia, Núcleo de Operações Técnicas, Vespasiano, MG, Brasil
| | - Evandro Bento Rodrigues
- Universidade Federal de Minas Gerais, Departamento de Genética, Ecologia e Evolução, Belo Horizonte, MG, Brasil
- Instituto Hermes Pardini-Grupo Fleury, Microbiologia, Núcleo de Operações Técnicas, Vespasiano, MG, Brasil
| | | | - Carine Rodrigues Pereira
- Universidade Federal de Lavras, Faculdade de Zootecnia e Medicina Veterinária, Departamento de Medicina Veterinária, Lavras, MG, Brasil
| | - Dircéia Aparecida Costa Custódio
- Universidade Federal de Lavras, Faculdade de Zootecnia e Medicina Veterinária, Departamento de Medicina Veterinária, Lavras, MG, Brasil
| | - Elaine Maria Seles Dorneles
- Universidade Federal de Lavras, Faculdade de Zootecnia e Medicina Veterinária, Departamento de Medicina Veterinária, Lavras, MG, Brasil
| | | | - Bertram Brenig
- University of Göttingen, Institute of Veterinary Medicine, Göttingen, Germany
| | - Vasco Azevedo
- Universidade Federal de Minas Gerais, Departamento de Genética, Ecologia e Evolução, Belo Horizonte, MG, Brasil
| | | |
Collapse
|
21
|
Varshith MR, Ghosh Dastidar R, Shrilaxmi MS, Bhattacharya R, Jha S, Choudhary S, Varny E, Carvalho RA, John L, Sundaramoorthy V, Smith CM, Damerla RR, Herai RH, Biswas SR, Lal PB, Mukhopadhyay C, Ghosh Dastidar S. Virulome and phylogenomic profiling of a novel Burkholderia pseudomallei strain from an Indian clinical isolate. Mol Genet Genomics 2024; 299:98. [PMID: 39441253 DOI: 10.1007/s00438-024-02188-5] [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: 09/02/2023] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Highly pathogenic Burkholderia pseudomallei is the causative agent of melioidosis, a neglected tropical disease endemic in Southeast Asian tropical region. This bacterium encompasses diverse virulence factors which further undergo dynamic gene-expression flux as it transits through distinct environmental niches within the host which may lead to manifestation of differential clinical symptoms. B. pseudomallei, is classified as a Tier 1 select agent in the United States and regarded as a risk group 3 organism in India with the potential to be used as bioweapon. Considering these facts, it is vital to uncover both physiological and genetic heterogeneity of B. pseudomallei, particularly to identify any novel virulence factors that may contribute to pathogenicity. B. pseudomallei strain CM000113 was isolated from a clinical case in India, characterized it for its physiological, biochemical, and prominently genetic traits through WGS. It has a type 2 morphotype with faster doubling time and high biofilm producing capacity as compared to Pseudomonas aeruginosa. The genome size is 7.3 Mbp and it is phylogenetically close to B. pseudomallei strain Mahidol 1106a and Burkholderia mallei Turkey 2. We observed genetic heterogeneity, as key virulence factors that were identified shows sequence dissimilarity with reference strains. Additionally, presence of genomic islands, harbouring two virulence factors, GmhA and GmhB2, associated with pathogenesis indicates possibility of horizontal gene transfer. These results emphasize the need for an extensive study focusing the genome of B. pseudomallei and its associated heterogeneity, to identify molecular biomarkers aiding to develop point-of-care diagnostic kits for early diagnosis of melioidosis.
Collapse
Affiliation(s)
- M R Varshith
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- Center for Emerging and Tropical Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Ranita Ghosh Dastidar
- Center for Emerging and Tropical Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - M S Shrilaxmi
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- Center for Emerging and Tropical Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Rajarshi Bhattacharya
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- Department of Botany, Visva Bharati University, Santiniketan, India
| | - S Jha
- Center for Emerging and Tropical Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - S Choudhary
- Center for Emerging and Tropical Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - E Varny
- Center for Emerging and Tropical Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - R A Carvalho
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Paraná, Brazil
| | - L John
- School of Medicine, Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geeelong, Australia
| | - V Sundaramoorthy
- School of Medicine, Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geeelong, Australia
| | - C M Smith
- School of Medicine, Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geeelong, Australia
| | - R R Damerla
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - R H Herai
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Paraná, Brazil
| | - S R Biswas
- Department of Botany, Visva Bharati University, Santiniketan, India
| | - P B Lal
- Center for Emerging and Tropical Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Chiranjay Mukhopadhyay
- Center for Emerging and Tropical Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | - Somasish Ghosh Dastidar
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
- Center for Emerging and Tropical Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
22
|
Akinola OT, Dahunsi SO. Whole genome sequencing reveals antibiotic resistance pattern and virulence factors in Klebsiella quasipneumoniae subsp. Similipneumoniae from Hospital wastewater in South-West, Nigeria. Microb Pathog 2024; 197:107040. [PMID: 39427715 DOI: 10.1016/j.micpath.2024.107040] [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: 03/11/2024] [Revised: 09/22/2024] [Accepted: 10/18/2024] [Indexed: 10/22/2024]
Abstract
Klebsiella quasipneumoniae is a distinct species from K. pneumoniae, even though it is sometimes mistaken phenotypically for the latter in clinical situations. K. quasipneumoniae is a pathogen and this study aims at understanding the genomic antibiotic resistance and virulence characteristics of Klebsiella quasipneumoniae subsp. similipneumoniae (B105 strain) isolated from tertiary hospital wastewater and the potential risks associated with its environmental spread. The Illumina platform was used for whole-genome sequencing (WGS), the generated raw reads (de novo) was assembled using RAPT NCBI, while other standardized bioinformatics tools were utilized to validate and examine the landscape of the genome's antibiotic resistance and virulence factors. The K. quasipneumoniae subsp. similipneumoniae (B105 strain), belonged to sequence type 1422 and was resistant to ampicillin, amoxicillin-clavulanic acid, ceftazidime, cefepime, meropenem, tetracycline, but susceptible to gentamicin. The annotated genome acknowledged the presence of blaOKP-B-2, ompK 36, fosA5, oqxAB, virulence genes responsible for capsule formation, lipopolysaccharide, iron uptake aerobactin (iutA), salmochelins (iroE, iroN), enterobactin siderophore, efllux pump (acrA, acrB) adherence, (mrkC, mrkD, and fimD) and two plasmids replicon IncFIB(K) and IncR. The study resonates the inadequacy of conventional microbiological identification methods to distinguish K. pneumoniae and K. quasipneumoniae and at the same time heightens the importance of using a genomic platform to extol the identity of K. quasipneumoniae subsp. similipneumoniae strain. Furthermore, the peculiarities of the acquired antimicrobial resistance and virulence genes, in this strain, are a potential risk to the environment.
Collapse
Affiliation(s)
- Omowumi T Akinola
- Microbiology Programme, College of Agriculture, Engineering and Science, Bowen University, Iwo, Nigeria.
| | - Samuel Olatunde Dahunsi
- Microbiology Programme, College of Agriculture, Engineering and Science, Bowen University, Iwo, Nigeria; The Radcliffe Institute for Advanced Study, Harvard University, Cambridge, Massachusetts, USA.
| |
Collapse
|
23
|
Pu F, Zhang N, Pang J, Zeng N, Baloch FB, Li Z, Li B. Deciphering the Genetic Architecture of Staphylococcus warneri Prophage vB_G30_01: A Comprehensive Molecular Analysis. Viruses 2024; 16:1631. [PMID: 39459963 PMCID: PMC11512304 DOI: 10.3390/v16101631] [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: 08/25/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
The current knowledge of Staphylococcus warneri phages is limited, with few genomes sequenced and characterized. In this study, a prophage, vB_G30_01, isolated from Staphylococcus warneri G30 was characterized and evaluated for its lysogenic host range. The phage was studied using transmission electron microscopy and a host range. The phage genome was sequenced and characterized in depth, including phylogenetic and taxonomic analyses. The linear dsDNA genome of vB_G30_01 contains 67 predicted open reading frames (ORFs), classifying it within Bronfenbrennervirinae. With a total of 10 ORFs involved in DNA replication-related and transcriptional regulator functions, vB_G30_01 may play a role in the genetics and transcription of a host. Additionally, vB_G30_01 possesses a complete set of genes related to host lysogeny and lysis, implying that vB_G30_01 may influence the survival and adaptation of its host. Furthermore, a comparative genomic analysis reveals that vB_G30_01 shares high genomic similarity with other Staphylococcus phages and is relatively closely related to those of Exiguobacterium and Bacillus, which, in combination with the cross-infection assay, suggests possible cross-species infection capabilities. This study enhances the understanding of Staphylococcus warneri prophages, providing insights into phage-host interactions and potential horizontal gene transfer.
Collapse
Affiliation(s)
- Fangxiong Pu
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, China; (F.P.); (J.P.)
| | - Ning Zhang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, China; (F.P.); (J.P.)
| | - Jiahe Pang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, China; (F.P.); (J.P.)
| | - Nan Zeng
- College of Land and Environment, Shenyang Agricultural University, Shenyang 110866, China; (N.Z.); (F.B.B.)
| | - Faryal Babar Baloch
- College of Land and Environment, Shenyang Agricultural University, Shenyang 110866, China; (N.Z.); (F.B.B.)
| | - Zijing Li
- Food Science College, Shenyang Agricultural University, Shenyang 110866, China;
| | - Bingxue Li
- College of Land and Environment, Shenyang Agricultural University, Shenyang 110866, China; (N.Z.); (F.B.B.)
| |
Collapse
|
24
|
Km Rakhi, Bhati R, Jain M, Singh AK, Muthukumaran J. Unveiling MurM inhibitors in Enterococcus faecalis V583: a promising approach to tackle antibiotic resistance. J Biomol Struct Dyn 2024:1-17. [PMID: 39413038 DOI: 10.1080/07391102.2024.2415686] [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: 05/26/2023] [Accepted: 04/12/2024] [Indexed: 10/18/2024]
Abstract
Enterococcus faecalis is commonly found in the GI tract of humans and animals. It causes various infections, especially in hospital environments, and shows growing antibiotic resistance. This study utilized a subtractive proteomics approach to find out the potential drug targets in E. faecalis. Unique metabolic pathways were analysed and compared to the host to minimize adverse effects. Among twenty nine pathogenic specific and seventy three host-pathogen common pathways identified using the KEGG database, sixty seven essential proteins were found through the DEG BLAST search. PSORTB predicted that forty cytoplasmic proteins could be suitable as druggable targets. Further analysis identified fourteen proteins with virulence properties using the VFDB BLAST. Among these, seven proteins with more than ten antigenic sites were subjected to DrugBank BLAST, identifying three novel and four existing drug targets. One of the crucial drug targets, MurM, was selected due to its critical role in peptidoglycan biosynthesis. The reason for selecting MurM is crucial for addressing antibiotic resistance, disrupting bacterial cell wall synthesis, and attaining selective antimicrobial activity. MurM belongs to the mixed αβ class with two functional domains. The possible binding site residues of MurM are Trp31, Lys35, Trp38, Arg215, and Tyr219. Virtual screening identified potential lead candidates for MurM, and four were selected based on their physiochemical, pharmacokinetic, and structural properties. This study provides valuable insights into identifying and analysing a potential drug target, the MurM protein, and its inhibitors in E. faecalis V583.
Collapse
Affiliation(s)
- Km Rakhi
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Rittik Bhati
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Monika Jain
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Amit Kumar Singh
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Jayaraman Muthukumaran
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| |
Collapse
|
25
|
Slavinska A, Kowalczyk M, Kirkliauskienė A, Vizuje G, Siedlecki P, Bikulčienė J, Tamošiūnienė K, Petrutienė A, Kuisiene N. Genetic characterization of Neisseria meningitidis isolates recovered from patients with invasive meningococcal disease in Lithuania. Front Cell Infect Microbiol 2024; 14:1432197. [PMID: 39469455 PMCID: PMC11513629 DOI: 10.3389/fcimb.2024.1432197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction Neisseria meningitidis is a gram-negative bacterium responsible for life-threatening invasive infections known as invasive meningococcal disease and is associated with high fatality rates and serious lifelong disabilities among survivors. Methods This study aimed to characterize N. meningitidis isolates cultured from blood and cerebrospinal fluid collected between 2009 and 2021 in Lithuania, assess their genomic relationships with European strains, and evaluate the possibility of using a cost-effective method for strain characterization, thus improving the national molecular surveillance of invasive meningococcal disease. In total, 321 N. meningitidis isolates were collected and analyzed using multilocus restriction typing (MLRT). Amplification of the penA gene and restriction fragment length polymorphism analysis were performed to identify the modified penA genes. Based on the MLRT genotyping results, we selected 10 strains for additional analysis using whole-genome sequencing. The sequenced genomes were incorporated into a dataset of publicly available N. meningitidis genomes to evaluate genomic diversity and establish phylogenetic relationships within the Lithuanian and European circulating strains. Results We identified 83 different strains using MLRT genotyping. Genomic diversity of N. meningitidis genomes analysed revealed 21 different sequence types (STs) circulating in Lithuania. Among these, ST34 was the most prevalent. Notably, three isolates displayed unique combinations of seven housekeeping genes and were identified as novel STs: ST16969, ST16901, and ST16959. The analyzed strains were found to possess virulence factors not commonly found in N. meningitidis. Six distinct penA profiles were identified, each with different frequencies. In the present study, we also identified N. meningitidis strains with new penA, NEIS0123, NEIS1320, NEIS1525, NEIS1600, and NEIS1753 loci variants. In our study, using the cgMLST scheme, Minimum Spanning Tree (MST) analysis did not identify significant geographic relationships between Lithuanian N. meningitidis isolates and strains from Europe. Discussion Discussion: To our knowledge, this is the first study to employ whole genome sequencing (WGS) method for a comprehensive genetic characterization of invasive N. meningitidis isolates from Lithuania. This approach provides a more detailed and precise analysis of genomic relationships and diversity compared to prior studies relying on traditional molecular typing methods and antigen analysis.
Collapse
Affiliation(s)
- Anželika Slavinska
- Department of Microbiology and Biotechnology, Institute of Biosciences of Vilnius University Life Sciences Centre, Vilnius, Lithuania
| | - Magdalena Kowalczyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Agnė Kirkliauskienė
- Faculty of Medicine, Institute of Biomedical Science, Vilnius University, Vilnius, Lithuania
| | - Greta Vizuje
- Microbiology Laboratory, Republican Vilnius University Hospital, Vilnius, Lithuania
| | - Paweł Siedlecki
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Kristina Tamošiūnienė
- Department of Microbiology and Biotechnology, Institute of Biosciences of Vilnius University Life Sciences Centre, Vilnius, Lithuania
| | - Aurelija Petrutienė
- Department of Clinical Investigations of the National Public Health Surveillance Laboratory, Vilnius, Lithuania
| | - Nomeda Kuisiene
- Department of Microbiology and Biotechnology, Institute of Biosciences of Vilnius University Life Sciences Centre, Vilnius, Lithuania
| |
Collapse
|
26
|
Xuan G, Liu X, Wang Y, Lin H, Jiang X, Wang J. Isolation, characterization, and application of a novel Pseudomonas fluorescens phage vB_PF_Y1-MI in contaminated milk. Mol Genet Genomics 2024; 299:97. [PMID: 39395039 DOI: 10.1007/s00438-024-02179-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 08/25/2024] [Indexed: 10/14/2024]
Abstract
The food industry has incurred substantial losses from contamination by Pseudomonas fluorescens, emphasizing the critical importance of implementing effective control strategies. Phages are potential sterilizers due to their specific killing abilities and the difficulty bacteria face in developing resistance. However, a significant barrier to their development is the lack of diversity among phage types. In this study, we characterized a novel lytic P. fluorescens phage, named vB_PF_Y1-MI. Phage vB_PF_Y1-MI displayed a latent period of nearly 10 min and a high burst size of 1493 PFU/cell. This phage showed good activity over a wide range of temperature (up to 70 °C) and pH (3-12). The genome of phage vB_PF_Y1-MI spans 93,233 bp with a GC content of 45%. It encompasses 174 open-reading frames and 19 tRNA genes, while no lysogeny or virulence-associated genes were detected. Phylogenetic analysis positions it as a novel unassigned evolutionary lineage within the Caudoviricetes class among related dsDNA phages. Our study provides foundational insights into vB_PF_Y1-MI and emphasizes its potential as an effective biological control agent against P. fluorescens. This research offers crucial theoretical groundwork and technical support for subsequent efforts in preventing and controlling P. fluorescens contamination.
Collapse
Affiliation(s)
- Guanhua Xuan
- Food Safety Laboratory, College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Xianjun Liu
- Food Safety Laboratory, College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Yinfeng Wang
- Food Safety Laboratory, College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Hong Lin
- Food Safety Laboratory, College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Xiuping Jiang
- Department of Food, Nutrition and Packaging Sciences, Clemson University, Clemson, SC, 29631, USA
| | - Jingxue Wang
- Food Safety Laboratory, College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
27
|
Soro O, Kigen C, Nyerere A, Gachoya M, Wachira J, Onyonyi V, Georges M, Wataka A, Ondolo S, Cherono K, Odoyo E, Musila L. Complete genome sequences of 12 lytic phages against multidrug-resistant Enterococcus faecalis. Microbiol Resour Announc 2024; 13:e0068724. [PMID: 39254335 PMCID: PMC11465766 DOI: 10.1128/mra.00687-24] [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: 06/22/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
We report the genome sequences of 12 Enterococcus faecalis phages isolated in Kenya, belonging to the genus Copernicusvirus, Efquatrovirus, Saphexavirus, and Kochikohdavirus. They have double-stranded DNA with lengths varying from 17,979 to 147,374 bp and G+C content from 33.14% to 40.05%. The genomes contain 28-250 coding sequences.
Collapse
Affiliation(s)
- Oumarou Soro
- Department of Molecular Biology and Biotechnology, Pan African University Institute for Basic Sciences, Technology, and Innovation, Nairobi, Kenya
| | - Collins Kigen
- Department of Emerging Infectious Diseases, Walter Reed Army Institute of Research-Africa, Kericho, Kenya
| | - Andrew Nyerere
- Department of Medical Microbiology, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Moses Gachoya
- Department of Emerging Infectious Diseases, Walter Reed Army Institute of Research-Africa, Kericho, Kenya
| | - James Wachira
- Department of Emerging Infectious Diseases, Walter Reed Army Institute of Research-Africa, Kericho, Kenya
| | - Vanessa Onyonyi
- Department of Emerging Infectious Diseases, Walter Reed Army Institute of Research-Africa, Kericho, Kenya
| | - Martin Georges
- Department of Emerging Infectious Diseases, Walter Reed Army Institute of Research-Africa, Kericho, Kenya
| | - Allan Wataka
- Department of Emerging Infectious Diseases, Walter Reed Army Institute of Research-Africa, Kericho, Kenya
| | - Stephen Ondolo
- Department of Emerging Infectious Diseases, Walter Reed Army Institute of Research-Africa, Kericho, Kenya
| | - Karen Cherono
- Department of Emerging Infectious Diseases, Walter Reed Army Institute of Research-Africa, Kericho, Kenya
| | - Erick Odoyo
- Department of Emerging Infectious Diseases, Walter Reed Army Institute of Research-Africa, Kericho, Kenya
| | - Lillian Musila
- Department of Emerging Infectious Diseases, Walter Reed Army Institute of Research-Africa, Kericho, Kenya
| |
Collapse
|
28
|
Thänert R, Schwartz DJ, Keen EC, Hall-Moore C, Wang B, Shaikh N, Ning J, Rouggly-Nickless LC, Thänert A, Ferreiro A, Fishbein SRS, Sullivan JE, Radmacher P, Escobedo M, Warner BB, Tarr PI, Dantas G. Clinical sequelae of gut microbiome development and disruption in hospitalized preterm infants. Cell Host Microbe 2024; 32:1822-1837.e5. [PMID: 39197454 PMCID: PMC11466706 DOI: 10.1016/j.chom.2024.07.027] [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: 05/10/2024] [Revised: 06/24/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Aberrant preterm infant gut microbiota assembly predisposes to early-life disorders and persistent health problems. Here, we characterize gut microbiome dynamics over the first 3 months of life in 236 preterm infants hospitalized in three neonatal intensive care units using shotgun metagenomics of 2,512 stools and metatranscriptomics of 1,381 stools. Strain tracking, taxonomic and functional profiling, and comprehensive clinical metadata identify Enterobacteriaceae, enterococci, and staphylococci as primarily exploiting available niches to populate the gut microbiome. Clostridioides difficile lineages persist between individuals in single centers, and Staphylococcus epidermidis lineages persist within and, unexpectedly, between centers. Collectively, antibiotic and non-antibiotic medications influence gut microbiome composition to greater extents than maternal or baseline variables. Finally, we identify a persistent low-diversity gut microbiome in neonates who develop necrotizing enterocolitis after day of life 40. Overall, we comprehensively describe gut microbiome dynamics in response to medical interventions in preterm, hospitalized neonates.
Collapse
Affiliation(s)
- Robert Thänert
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Drew J Schwartz
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Women's Infectious Diseases Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric C Keen
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bin Wang
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie Ning
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Anna Thänert
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aura Ferreiro
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Skye R S Fishbein
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Janice E Sullivan
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY 40202, USA
| | - Paula Radmacher
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY 40202, USA
| | - Marilyn Escobedo
- Department of Pediatrics, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
29
|
Wu G, Xu T, Zhao N, Lam YY, Ding X, Wei D, Fan J, Shi Y, Li X, Li M, Ji S, Wang X, Fu H, Zhang F, Shi Y, Zhang C, Peng Y, Zhao L. A core microbiome signature as an indicator of health. Cell 2024:S0092-8674(24)01038-9. [PMID: 39378879 DOI: 10.1016/j.cell.2024.09.019] [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: 12/09/2023] [Revised: 07/29/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
The gut microbiota is crucial for human health, functioning as a complex adaptive system akin to a vital organ. To identify core health-relevant gut microbes, we followed the systems biology tenet that stable relationships signify core components. By analyzing metagenomic datasets from a high-fiber dietary intervention in type 2 diabetes and 26 case-control studies across 15 diseases, we identified a set of stably correlated genome pairs within co-abundance networks perturbed by dietary interventions and diseases. These genomes formed a "two competing guilds" (TCGs) model, with one guild specialized in fiber fermentation and butyrate production and the other characterized by virulence and antibiotic resistance. Our random forest models successfully distinguished cases from controls across multiple diseases and predicted immunotherapy outcomes through the use of these genomes. Our guild-based approach, which is genome specific, database independent, and interaction focused, identifies a core microbiome signature that serves as a holistic health indicator and a potential common target for health enhancement.
Collapse
Affiliation(s)
- Guojun Wu
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences and Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; Rutgers-Jiaotong Joint Laboratory for Microbiome and Human Health, New Brunswick, NJ, USA
| | - Ting Xu
- State Key Laboratory of Microbial Metabolism and Ministry of Education Key Laboratory of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Naisi Zhao
- Department of Public Health and Community Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA
| | - Yan Y Lam
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiaoying Ding
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Dongqin Wei
- Department of Endocrinology and Metabolism, Qidong People's Hospital, Qidong, Jiangsu 226200, China
| | - Jian Fan
- Department of Endocrinology and Metabolism, Qidong People's Hospital, Qidong, Jiangsu 226200, China
| | - Yajuan Shi
- Department of Endocrinology and Metabolism, Qidong People's Hospital, Qidong, Jiangsu 226200, China
| | - Xiaofeng Li
- Department of Endocrinology and Metabolism, Qidong People's Hospital, Qidong, Jiangsu 226200, China
| | - Mi Li
- Department of Endocrinology and Metabolism, Qidong People's Hospital, Qidong, Jiangsu 226200, China
| | - Shenjie Ji
- Department of Endocrinology and Metabolism, Qidong People's Hospital, Qidong, Jiangsu 226200, China
| | - Xuejiao Wang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Huaqing Fu
- State Key Laboratory of Microbial Metabolism and Ministry of Education Key Laboratory of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Feng Zhang
- Nutrition Department (Clinical Study Center of Functional Food), The Affiliated Hospital of Jiangnan University Wuxi, Wuxi, Jiangsu 214122, China
| | - Yu Shi
- Department of Endocrinology and Metabolism, Qidong People's Hospital, Qidong, Jiangsu 226200, China.
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism and Ministry of Education Key Laboratory of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yongde Peng
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Liping Zhao
- State Key Laboratory of Microbial Metabolism and Ministry of Education Key Laboratory of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences and Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; Rutgers-Jiaotong Joint Laboratory for Microbiome and Human Health, New Brunswick, NJ, USA.
| |
Collapse
|
30
|
Bachelle SV, Bah SY, Addo RT, Bediako-Bowan AAA, Egyir B, Tsatsu SE, Dzudzor B, Amarh V. Genomic analysis of Enterobacteriaceae from colorectal cancer patients at a tertiary hospital in Ghana: a case-control study. Sci Rep 2024; 14:23195. [PMID: 39369124 PMCID: PMC11455924 DOI: 10.1038/s41598-024-74299-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/25/2024] [Indexed: 10/07/2024] Open
Abstract
Colorectal cancer (CRC) is a severe gastrointestinal cancer and a leading cause of cancer-related deaths in Ghana. The potential role of gut Enterobacteriaceae in the increasing incidence of CRC in Ghana is yet to be thoroughly investigated. In this study, Enterobacteriaceae from CRC patients and healthy control participants were analyzed by whole genome sequencing to identify genomic features that are associated with CRC. Socio-demographic data showed a significant association between age and alcohol consumption and CRC. Escherichia coli was the most abundant Enterobacteriaceae isolated from the study participants and they were predominantly intestinal commensals. Escherichia coli isolates belonging to phylogroup D encoded the highest number of virulence genes. The agn43 and int genes were widespread in Escherichia coli isolates from the CRC patients. Multilocus sequence types of potentially pathogenic Escherichia coli from the CRC patients also encoded genes involved in aggregation, adherence and biofilm formation. The ampC2 and ampH antimicrobial resistance genes were also widespread in the genome of the Escherichia coli isolates. This study highlights the virulence tendencies of Escherichia coli from CRC patients and their ability to transfer virulence determinants to other Enterobacteriaceae residing in the gut.
Collapse
Affiliation(s)
- Sarah V Bachelle
- Department of Medical Biochemistry, University of Ghana Medical School, Korle-Bu, Accra, Ghana
| | - Saikou Y Bah
- School of Infection & Immunity, University of Glasgow, Glasgow, UK
| | - Richmond T Addo
- Central Laboratory, Korle-Bu Teaching Hospital, Korle-Bu, Accra, Ghana
| | - Antoinette A A Bediako-Bowan
- Department of Surgery, University of Ghana Medical School, Korle-Bu, Accra, Ghana
- Department of Surgery, Korle-Bu Teaching Hospital, Korle-Bu, Accra, Ghana
| | - Beverly Egyir
- Bacteriology Department, Noguchi Memorial Institute for Medical Research, Accra, Ghana
| | - Sandra E Tsatsu
- Department of Surgery, University of Ghana Medical School, Korle-Bu, Accra, Ghana
- Department of Surgery, Korle-Bu Teaching Hospital, Korle-Bu, Accra, Ghana
| | - Bartholomew Dzudzor
- Department of Medical Biochemistry, University of Ghana Medical School, Korle-Bu, Accra, Ghana.
| | - Vincent Amarh
- Department of Medical Biochemistry, University of Ghana Medical School, Korle-Bu, Accra, Ghana.
| |
Collapse
|
31
|
Romão FT, Santos ACM, Puño-Sarmiento JJ, Sperandio V, Hernandes RT, Gomes TAT. Expression of the locus of enterocyte effacement genes during the invasion process of the atypical enteropathogenic Escherichia coli 1711-4 strain of serotype O51:H40. Microbiol Spectr 2024; 12:e0030424. [PMID: 39189752 PMCID: PMC11448038 DOI: 10.1128/spectrum.00304-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 07/01/2024] [Indexed: 08/28/2024] Open
Abstract
Atypical enteropathogenic Escherichia coli (aEPEC) is a significant cause of diarrhea in low- and middle-income countries. Certain aEPEC strains, including the Brazilian representative strain of serotype O51:H40 called aEPEC 1711-4, can use flagella to attach to, invade, and persist in T84 and Caco-2 intestinal cells. It can also translocate from the gut to extraintestinal sites in a rat model. Although various aspects of the virulence of this strain were studied and the requirement of a type III secretion system for the efficiency of the invasion process was demonstrated, the expression of the locus of enterocyte effacement (LEE) genes during the invasion and intracellular persistence remains unclear. To address this question, the expression of flagella and the different LEE operons was evaluated during kinetic experiments of the interaction of aEPEC 1711-4 with enterocytes in vitro. The genome of the strain was also sequenced. The results showed that flagella expression remained unchanged, but the expression of eae and escJ increased during the early interaction and invasion of aEPEC 1711-4 into Caco-2 cells, and there was no change 24 h post-infection during the persistence period. The number of actin accumulation foci formed on HeLa cells also increased during the 6-h analysis. No known gene related to the invasion process was identified in the genome of aEPEC 1711-4, which was shown to belong to the global EPEC lineage 10. These findings suggest that the LEE components and the intimate adherence promoted by intimin are necessary for the invasion and persistence of aEPEC 1711-4, but the detailed mechanism needs further study.IMPORTANCEAtypical enteropathogenic Escherichia coli (aEPEC) is a major cause of diarrhea, especially in low- and middle-income countries, like Brazil. However, due to the genome heterogeneity of each clonal group, it is difficult to comprehend the pathogenicity of this strain fully. Among aEPEC strains, 1711-4 can invade eukaryotic cells in vitro, cross the gut barrier, and reach extraintestinal sites in animal models. By studying how different known aEPEC virulence factors are expressed during the invasion process, we can gain insight into the commonalities of this phenotype among other aEPEC strains. This will help in developing preventive measures to control infections caused by invasive strains. No known virulence-encoding genes linked to the invasion process were found. Nevertheless, additional studies are still necessary to evaluate the role of other factors in this phenotype.
Collapse
Affiliation(s)
- Fabiano T. Romão
- Disciplina de Microbiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Department of Microbiology, UT Southwestern Medical Center, Dallas, Texas, USA
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ana C. M. Santos
- Disciplina de Microbiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Juan J. Puño-Sarmiento
- Disciplina de Microbiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Vanessa Sperandio
- Department of Microbiology, UT Southwestern Medical Center, Dallas, Texas, USA
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Rodrigo T. Hernandes
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Tânia A. T. Gomes
- Disciplina de Microbiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
32
|
Zhao Y, Zhao Q, Liu D, Xie H, Zhang J, Zheng Y, Xu X, Wu H, Hu Z. Antibiotic resistomes and ecological risk elimination in field-scale constructed wetland revealed by integrated metagenomics and metatranscriptomics. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136045. [PMID: 39368357 DOI: 10.1016/j.jhazmat.2024.136045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/05/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Constructed wetlands (CWs) are identified as significant ecological systems for the potential control of antibiotic resistance genes (ARGs) in the environment. However, the precise mechanisms governing removal, persistence, expression, and associated risks of ARGs during wetland treatment remain poorly understood. In this study, the distribution, mobility, expression, and hosts of ARGs in water, sediments, and plants of a field-scale CW and its parallel natural river were systematically investigated through metagenomic and metatranscriptomic approaches. Results showed that both the abundance and diversity of ARGs in water gradually decreased along the way of CW, reaching a final abundance removal rate of 72.28 % in the effluent. Source tracking analysis indicted that the reduction of ARGs in water was mainly achieved by the dynamic accumulation of ARGs in sediments and plants of the CW. Proteobacteria were identified as primary hosts for ARGs, particularly in sediments and plants during CW treatment. Moreover, although ESKAPE pathogens carrying multiple ARGs persisted in all media throughout the CW treatment, ARG expression levels and risk of water were also significantly decreased after CW treatment. Collectively, our comprehensive multi-omics study would enhance the understanding of ARG removal by CWs, offering insights for controlling antimicrobial resistance in wastewater treatment system.
Collapse
Affiliation(s)
- Yanhui Zhao
- School of Environmental Science & Engineering, Shandong Key Laboratory of Water Pollution Control and Resource Reuse, Shandong University, Qingdao 266237, PR China
| | - Qian Zhao
- School of Environmental Science & Engineering, Shandong Key Laboratory of Water Pollution Control and Resource Reuse, Shandong University, Qingdao 266237, PR China
| | - Daoxing Liu
- Shandong Innovation and Entrepreneurship Community of Green Industry and Environmental Security, Jinan 250199, PR China; Shandong Academy of Environmental Science Co., LTD., Jinan 250199, PR China
| | - Huijun Xie
- Environmental Research Institute, Shandong University, Qingdao 266237, PR China
| | - Jian Zhang
- School of Environmental Science & Engineering, Shandong Key Laboratory of Water Pollution Control and Resource Reuse, Shandong University, Qingdao 266237, PR China; Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, PR China.
| | - Yu Zheng
- RIKEN Center for Sustainable Resource Science, Saitama, 351-0198, Japan
| | - Xinyi Xu
- School of Environmental Science & Engineering, Shandong Key Laboratory of Water Pollution Control and Resource Reuse, Shandong University, Qingdao 266237, PR China
| | - Haiming Wu
- School of Environmental Science & Engineering, Shandong Key Laboratory of Water Pollution Control and Resource Reuse, Shandong University, Qingdao 266237, PR China
| | - Zhen Hu
- School of Environmental Science & Engineering, Shandong Key Laboratory of Water Pollution Control and Resource Reuse, Shandong University, Qingdao 266237, PR China.
| |
Collapse
|
33
|
Cabal A, Hörtenhuber A, Salaheddin Y, Stöger A, Springer B, Bletz S, Mellmann A, Hyden P, Hartl R, Weinberger J, Conzemius R, Hell M, Daza-Prieto B, Lippert K, Steindl G, Köberl-Jelovcan S, Ruppitsch W. Three prolonged outbreaks of metallo-β-lactamase-producing Pseudomonas aeruginosa in an Upper Austrian hospital, 2017-2023. Microbiol Spectr 2024; 12:e0074024. [PMID: 39162508 PMCID: PMC11448029 DOI: 10.1128/spectrum.00740-24] [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: 03/22/2024] [Accepted: 07/01/2024] [Indexed: 08/21/2024] Open
Abstract
In spring 2022, an increase in metallo-β-lactamase-producing Pseudomonas aeruginosa (MBL-Pa) infections was detected in a hospital in Upper Austria. To identify the source of infection and to stop further transmissions, an epidemiological outbreak investigation including whole-genome sequencing (WGS)-based typing was conducted. The final case definition included cases admitted to the hospital between 2020 and 2023 with an MBL-Pa in one of the three genomic clusters identified. In addition, the investigation was extended to include historical cases from 2017. Core genome multilocus sequence typing was performed to assess the genetic relatedness between the isolates. Fifty-four clinical P. aeruginosa isolates and eight P. aeruginosa isolates from the hospital environment were obtained. All but nine isolates grouped into one of three genomic clusters (ST235/blaVIM-1, ST111/blaVIM-2, or ST621/blaIMP-13), which were considered to be distinct, prolonged outbreaks involving 47 out of 52 cases. The most likely source of infection for cluster 1 (ST111/blaVIM-2) and cluster 2 (ST235/blaVIM-1) was sinks in the intensive care unit (ICU) washroom. Cluster 3 clone (ST621/blaIMP-13) could have originated in the urology ward in 2020 and then spread to the ICU years later. However, the nosocomial origin of this clone could not be proven. In March 2023, following the implementation of control measures (gowning, patient isolation, screening, and daily disinfection), no further MLB-Pa was detected, and the outbreaks were considered to be over. As ICUs play an important role in the transmission of P. aeruginosa, emphasis should be placed on genomic surveillance, infection prevention, and control in such wards. IMPORTANCE The significance of our work lies in the successful resolution of three prolonged outbreaks of MBL-Pa infections in a hospital in Upper Austria. Through a comprehensive epidemiological investigation coupled with WGS-based typing of P. aeruginosa isolates, the study identified three distinct genomic clusters responsible for prolonged outbreaks involving 47 cases. The investigation pinpointed sinks in the ICU washroom as the likely source of infection for two of the clusters. The study demonstrates the effectiveness of control measures such as hand hygiene, gowning, patient isolation, screening, and disinfection in stopping further transmission and bringing the outbreaks to a close. This underscores the critical role of genomic surveillance and control measures, particularly in high-risk settings like ICUs, in reducing nosocomial transmission of MBL-Pa infections.
Collapse
Affiliation(s)
- Adriana Cabal
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Anna Hörtenhuber
- Institute of Pathology, Upper Austrian Health Holding GmbH, Pyhrn-Eisenwurzen Clinical Centre Kirchdorf Steyr, Steyr, Austria
| | - Yarub Salaheddin
- Institute of Pathology, Upper Austrian Health Holding GmbH, Pyhrn-Eisenwurzen Clinical Centre Kirchdorf Steyr, Steyr, Austria
| | - Anna Stöger
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Burkhard Springer
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Graz, Austria
| | - Stefan Bletz
- Institute of Hygiene, University Hospital Muenster and University of Muenster, Muenster, Germany
| | - Alexander Mellmann
- Institute of Hygiene, University Hospital Muenster and University of Muenster, Muenster, Germany
| | - Patrick Hyden
- Department of Statistics and Analytical Epidemiology, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Rainer Hartl
- National Reference Center for Antimicrobial Resistance, Institute for Hygiene, Microbiology and Tropical Medicine, Ordensklinikum Linz Elisabethinen, Linz, Austria
- Johannes Kepler University Linz, Medical Faculty, Linz, Austria
| | | | | | - Markus Hell
- MEDILAB, Teaching Laboratory of the Paracelsus Medical University, Salzburg, Austria
| | - Beatriz Daza-Prieto
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Kathrin Lippert
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| | - Georg Steindl
- Institute for Hospital Hygiene and Microbiology (IKM), Graz, Austria
| | - Sandra Köberl-Jelovcan
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Graz, Austria
| | - Werner Ruppitsch
- Division for Public Health, Institute for Medical Microbiology and Hygiene, Austrian Agency for Health and Food Safety, Vienna, Austria
| |
Collapse
|
34
|
Wang L, Chen S, Xing M, Dong L, Zhu H, Lin Y, Li J, Sun T, Zhu X, Wang X. Genome characterization of Shewanella algae in Hainan Province, China. Front Microbiol 2024; 15:1474871. [PMID: 39417074 PMCID: PMC11480045 DOI: 10.3389/fmicb.2024.1474871] [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: 08/05/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Shewanella algae is an emerging marine zoonotic pathogen. In this study, we first reported the Shewanella algae infections in patients and animals in Hainan Province, China. Currently, there is still relatively little known about the whole-genome characteristics of Shewanella algae in most tropical regions, including in southern China. Here, we sequenced the 62 Shewanella algae strains isolated from Hainan Province and combined with the whole genomes sequences of 144 Shewanella algae genomes from public databases to analyze genomic features. Phylogenetic analysis revealed that Shewanella algae is widely distributed in the marine environments of both temperate and tropical countries, exhibiting close phylogenetic relationships with genomes isolated from patients, animals, and plants. Thereby confirming that exposure to marine environments is a risk factor for Shewanella algae infections. Average nucleotide identity analysis indicated that the clonally identical genomes could be isolated from patients with different sample types at different times. Pan-genome analysis identified a total of 21,909 genes, including 1,563 core genes, 8,292 strain-specific genes, and 12,054 accessory genes. Multiple putative virulence-associated genes were identified, encompassing 14 categories and 16 subcategories, with 171 distinct virulence factors. Three different plasmid replicon types were detected in 33 genomes. Eleven classes of antibiotic resistance genes and 352 integrons were identified. Antimicrobial susceptibility testing revealed a high resistance rate to imipenem and colistin among the strains studied, with 5 strains exhibiting multidrug resistance. However, they were all sensitive to amikacin, minocycline, and tigecycline. Our findings clarify the genomic characteristics and population structure of Shewanella algae in Hainan Province. The results offer insights into the genetic basis of pathogenicity in Shewanella algae and enhance our understanding of its global phylogeography.
Collapse
Affiliation(s)
- Licheng Wang
- Clinical & Central Laboratory of Sanya People’s Hospital, Sanya, China
| | - Shaojin Chen
- Clinical & Central Laboratory of Sanya People’s Hospital, Sanya, China
| | - Mei Xing
- Wenchang People’s Hospital, Wenchang, China
| | - Lingzhi Dong
- Clinical & Central Laboratory of Sanya People’s Hospital, Sanya, China
| | - Huaxiong Zhu
- Clinical & Central Laboratory of Sanya People’s Hospital, Sanya, China
| | - Yujin Lin
- Clinical & Central Laboratory of Sanya People’s Hospital, Sanya, China
| | - Jinyi Li
- Clinical & Central Laboratory of Sanya People’s Hospital, Sanya, China
| | - Tuo Sun
- Clinical & Central Laboratory of Sanya People’s Hospital, Sanya, China
| | - Xiong Zhu
- Clinical & Central Laboratory of Sanya People’s Hospital, Sanya, China
| | - Xiaoxia Wang
- Clinical & Central Laboratory of Sanya People’s Hospital, Sanya, China
| |
Collapse
|
35
|
Wei Q, Yin Y, Tong Q, Gong Z, Shi Y. Multi-omics analysis of excessive nitrogen fertilizer application: Assessing environmental damage and solutions in potato farming. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116916. [PMID: 39181078 DOI: 10.1016/j.ecoenv.2024.116916] [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: 03/04/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Potatoes (Solanum tuberosum L.) are the third largest food crop globally and are pivotal for global food security. Widespread N fertilizer waste in potato cultivation has caused diverse environmental issues. This study employed microbial metagenomic sequencing to analyze the causes behind the declining N use efficiency (NUE) and escalating greenhouse gas emissions resulting from excessive N fertilizer application. Addressing N fertilizer inefficiency through breeding has emerged as a viable solution for mitigating overuse in potato cultivation. In this study, transcriptome and metabolome analyses were applied to identify N fertilizer-responsive genes. Metagenomic sequencing revealed that excessive N fertilizer application triggered alterations in the population dynamics of 11 major bacterial phyla, consequently affecting soil microbial functions, particularly N metabolism pathways and bacterial secretion systems. Notably, the enzyme levels associated with NO3- increased, and those associated with NO and N2O increased. Furthermore, excessive N fertilizer application enhanced soil virulence factors and increased potato susceptibility to diseases. Transcriptome and metabolome sequencing revealed significant impacts of excessive N fertilizer use on lipid and amino acid metabolism pathways. Weighted gene co‑expression network analysis (WGCNA) was adopted to identify two genes associated with N fertilizer response: PGSC0003DMG400021157 and PGSC0003DMG400009544.
Collapse
Affiliation(s)
- Qiaorong Wei
- College of Agriculture, Northeast Agricultural University, Harbin, China; National Key Laboratory of Smart Farm Technologies and Systems, Harbin, China; Key Laboratory of Germplasm Enhancement, Physiology and Ecology of Food Crops in Cold Region, Ministry of Education, Harbin, China
| | - Yanbin Yin
- College of Agriculture, Northeast Agricultural University, Harbin, China; National Key Laboratory of Smart Farm Technologies and Systems, Harbin, China
| | - Qingsong Tong
- College of Agriculture, Northeast Agricultural University, Harbin, China
| | - Zhenping Gong
- College of Agriculture, Northeast Agricultural University, Harbin, China.
| | - Ying Shi
- College of Agriculture, Northeast Agricultural University, Harbin, China; Key Laboratory of Germplasm Enhancement, Physiology and Ecology of Food Crops in Cold Region, Ministry of Education, Harbin, China.
| |
Collapse
|
36
|
Méndez Acevedo M, Rolon ML, Johnson BB, Burns LH, Stacy J, Aurand-Cravens A, LaBorde L, Kovac J. Sanitizer Resistance and Persistence of Listeria monocytogenes Isolates in Tree Fruit Packing Facilities. J Food Prot 2024; 87:100354. [PMID: 39218076 DOI: 10.1016/j.jfp.2024.100354] [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: 03/27/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The foodborne pathogen Listeria monocytogenes can persist in produce processing environments, which increases the risk for food contamination. Increased resistance to antimicrobials commonly used in cleaning and sanitizing procedures may contribute to L. monocytogenes' persistence in these environments. This study aimed to evaluate sanitizer resistance in L. monocytogenes isolates collected from three tree fruit packing facilities (F1, F2, and F3) during packing seasons 2020-2021 (Y1) and 2021-2022 (Y2), and to assess evidence of persistence based on the genomic similarity of isolates to historical isolates collected in previous years. L. monocytogenes isolates collected in 2020-2022 (n = 44) were tested for resistance to peroxyacetic acid (PAA) and a proprietary biofilm-removing agent using a broth microdilution assay. Further, L. monocytogenes isolates were whole genome sequenced and screened for the presence of antimicrobial resistance and virulence genes, as well as to assess the genomic similarity of isolates using the CFSAN SNP bioinformatic pipeline. Over half (57%) of the tested isolates had a PAA minimum inhibitory concentration (MIC) of 250 ppm, which was similar to the applied concentration of the PAA sanitizer in the three facilities (230 ppm). In contrast, 80% of tested isolates had a biofilm remover MIC of 0.13 ppm, which was substantially below the concentration applied in the facilities (137 ppm). Genomes of all tested isolates carried antimicrobial resistance (fosX, lin, mdrL, mprF, and norB) and virulence (inlA, inlB, plcA, plcB, prfA, hly, mpl, and iap) genes. L. monocytogenes isolates collected between 2020 and 2022 belonged to three distinct lineages, with 22 multilocus sequence types (MLSTs) belonging to 22 different clonal complexes. Genomic similarity analysis with historical isolates collected from the same facilities in 2016-2017 demonstrated a 5-year persistence of the genotypes ST 1003 and ST 554 in F2, which were no longer detected in 2022. Overall, our results highlight the need to re-evaluate sanitizer concentrations to effectively control persistent L. monocytogenes strains in tree fruit packing facilities.
Collapse
Affiliation(s)
- Marysabel Méndez Acevedo
- Department of Food Science, The Pennsylvania State University, University Park, PA 16802, United States.
| | - M Laura Rolon
- Department of Food Science, The Pennsylvania State University, University Park, PA 16802, United States.
| | - Beth B Johnson
- Division of Laboratory Services, Kentucky Department of Public Health, Frankfort, KY 40601, United States.
| | - Logan H Burns
- Division of Laboratory Services, Kentucky Department of Public Health, Frankfort, KY 40601, United States.
| | - Joshua Stacy
- Division of Laboratory Services, Kentucky Department of Public Health, Frankfort, KY 40601, United States.
| | - Ashley Aurand-Cravens
- Division of Laboratory Services, Kentucky Department of Public Health, Frankfort, KY 40601, United States.
| | - Luke LaBorde
- Department of Food Science, The Pennsylvania State University, University Park, PA 16802, United States.
| | - Jasna Kovac
- Department of Food Science, The Pennsylvania State University, University Park, PA 16802, United States.
| |
Collapse
|
37
|
Jiao P, Zhou Y, Zhang X, Jian H, Zhang XX, Ma L. Mechanisms of horizontal gene transfer and viral contribution to the fate of intracellular and extracellular antibiotic resistance genes in anaerobic digestion supplemented with conductive materials under ammonia stress. WATER RESEARCH 2024; 267:122549. [PMID: 39368190 DOI: 10.1016/j.watres.2024.122549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/07/2024]
Abstract
The addition of conductive materials (CMs) is an effective strategy for mitigating ammonia inhibition during anaerobic digestion (AD). However, the introduction of CMs can result in increased antibiotic resistance genes (ARGs) pollution, potentially facilitated by enhanced horizontal gene transfer (HGT). The complex dynamics of intracellular and extracellular ARGs (iARGs/eARGs) and the mechanisms underlying their transfer, mediated by CMs, in ammonia-stressed AD systems remain unclear. In this study, we investigated the effects of three commonly used CMs-nano magnetite (Mag), nano zero-valent iron (nZVI), and granular activated carbon (GAC)-on the fate of iARGs and eARGs during the AD of waste activated sludge under ammonia stress. The results revealed an unexpected enrichment of iARGs by 1.5 %-10.9 % and a reduction of eARGs by 14.1 %-25.2 % in CM-supplemented AD. This discrepancy in the dynamics of iARGs and eARGs may be attributed to changes in microbial hosts and the horizontal transfer of ARGs. Notably, CMs activated prophages within antibiotic-resistant bacteria (ARB) and their symbiotic partners involved in vitamin B12 provision, leading to the lysis of ARB and the subsequent release of eARGs for transformation. Additionally, the abundance of potentially mobile ARGs, which co-occurred with mobile genetic elements, increased by 56.6 %-134.5 % with CM addition, highlighting an enhanced potential for the HGT of ARGs. Specifically, Mag appeared to promote both transformation and conjugation processes, while nZVI only promoted conjugation. Moreover, none of the three CMs had any discernible impact on transduction. GAC proved superior to both nano Mag and nZVI in controlling the enrichment of iARGs, reducing eARGs, and limiting HGTs simultaneously. Overall, these findings provide novel insights into the role of viruses and the mechanisms of ARG spread in CM-assisted AD, offering valuable information for developing strategies to mitigate ARG pollution in practical applications.
Collapse
Affiliation(s)
- Pengbo Jiao
- Shanghai Key Lab for Urban Ecological Processes and Eco-Restoration, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China
| | - Ying Zhou
- Shanghai Key Lab for Urban Ecological Processes and Eco-Restoration, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China
| | - Xingxing Zhang
- Shanghai Key Lab for Urban Ecological Processes and Eco-Restoration, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China
| | - Huahua Jian
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Development Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xu-Xiang Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China
| | - Liping Ma
- Shanghai Key Lab for Urban Ecological Processes and Eco-Restoration, Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University, Shanghai, 200241, China; Technology Innovation Center for Land Spatial Eco-restoration in Metropolitan Area, Ministry of Natural Resources, Shanghai, 200062, China.
| |
Collapse
|
38
|
González A, Fullaondo A, Odriozola A. Host genetics and microbiota data analysis in colorectal cancer research. ADVANCES IN GENETICS 2024; 112:31-81. [PMID: 39396840 DOI: 10.1016/bs.adgen.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Colorectal cancer (CRC) is a heterogeneous disease with a complex aetiology influenced by a myriad of genetic and environmental factors. Despite advances in CRC research, it is a major burden of disease, with the second highest incidence and third leading cause of cancer deaths worldwide. To individualise diagnosis, prognosis, and treatment of CRC, developing new strategies combining precision medicine and bioinformatic procedures is promising. Precision medicine is based on omics technologies and aims to individualise the management of CRC based on patient host genetic characteristics and microbiota. Bioinformatics is central to the application of personalised medicine because it enables the analysis of large datasets generated by these technologies. At the level of host genetics, bioinformatics allows the identification of mutations, genes, molecular pathways, biomarkers and drugs relevant to colorectal carcinogenesis. At the microbiota level, bioinformatics is fundamental to analysing microbial communities' composition and functionality and developing biomarkers and personalised microbiota-based therapies. This paper explores the host and microbiota genetic data analysis in CRC research.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| |
Collapse
|
39
|
Cheng M, Xu Y, Cui X, Wei X, Chang Y, Xu J, Lei C, Xue L, Zheng Y, Wang Z, Huang L, Zheng M, Luo H, Leng Y, Jiang C. Deep longitudinal lower respiratory tract microbiome profiling reveals genome-resolved functional and evolutionary dynamics in critical illness. Nat Commun 2024; 15:8361. [PMID: 39333527 PMCID: PMC11436904 DOI: 10.1038/s41467-024-52713-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
The lower respiratory tract (LRT) microbiome impacts human health, especially among critically ill patients. However, comprehensive characterizations of the LRT microbiome remain challenging due to low microbial mass and host contamination. We develop a chelex100-based low-biomass microbial-enrichment method (CMEM) that enables deep metagenomic profiling of LRT samples to recover near-complete microbial genomes. We apply the method to 453 longitudinal LRT samples from 157 intensive care unit (ICU) patients in three geographically distant hospitals. We recover 120 high-quality metagenome-assembled genomes (MAGs) and associated plasmids without culturing. We detect divergent longitudinal microbiome dynamics and hospital-specific dominant opportunistic pathogens and resistomes in pneumonia patients. Diagnosed pneumonia and the ICU stay duration were associated with the abundance of specific antibiotic-resistance genes (ARGs). Moreover, CMEM can serve as a robust tool for genome-resolved analyses. MAG-based analyses reveal strain-specific resistome and virulome among opportunistic pathogen strains. Evolutionary analyses discover increased mobilome in prevailing opportunistic pathogens, highly conserved plasmids, and new recombination hotspots associated with conjugative elements and prophages. Integrative analysis with epidemiological data reveals frequent putative inter-patient strain transmissions in ICUs. In summary, we present a genome-resolved functional, transmission, and evolutionary landscape of the LRT microbiota in critically ill patients.
Collapse
Affiliation(s)
- Minghui Cheng
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310030, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Yingjie Xu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xiao Cui
- Department of Intensive Care Unit, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Xin Wei
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310030, China
| | - Yundi Chang
- Department of Intensive Care Unit, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Jun Xu
- Department of Critical Care Medicine, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cheng Lei
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Lei Xue
- Department of Intensive Care Unit, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Yifan Zheng
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310030, China
| | - Zhang Wang
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Lingtong Huang
- Department of Critical Care Medicine, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Hong Luo
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Yuxin Leng
- Department of Intensive Care Unit, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China.
| | - Chao Jiang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310030, China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, 321000, China.
| |
Collapse
|
40
|
de Oliveira VF, de Britto-Costa LF, de Aragão GL, Scaccia N, Mamana AC, Côrtes MF, de Oliveira MS, de Melo Tavares B, Manuli ER, Leal FE, de Oliveira Xavier GT, Grespan RMZ, Sequeira CCR, Nunes FLS, Dropa M, Martone-Rocha S, Razzolini MTP, Sabino EC, Padoveze MC, Holmes A, Costa SF, Levin AS. Colonisation by multidrug-resistant organisms in health workers in primary care: narrow spectrum oral antimicrobials are a risk factor. Eur J Clin Microbiol Infect Dis 2024:10.1007/s10096-024-04953-1. [PMID: 39320520 DOI: 10.1007/s10096-024-04953-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Limited information exists on carriage of multidrug-resistant organisms (MDRO) by health workers (HWs) in primary care settings. This study aims to determine the prevalence of MDRO carriage among HWs in primary care and to identify associated risk factors. METHODS A cross-sectional study was conducted across all 12 primary care units in São Caetano do Sul-SP, Brazil, from October to December 2023. Self-collected samples (nasal, oropharyngeal, and inguinal) were obtained. Environment cultures (potable water, sewage and stream water) were evaluated. Stenotrophomonas maltophilia isolates (human and environmental) were typed. RESULTS The study included 265/288 (92%) of HWs in primary care teams, mostly women with a median age of 47 years (IQR 38-57); 78% had no comorbidities. MDRO colonisation was found in 8.7% (23 HWs). The following bacteria were found: S. maltophilia (n = 9; 3.4%) in inguinal swabs; methicillin-resistant Staphylococcus aureus (n = 8; 3%) from all sites; extended-spectrum ß-lactamase-producing bacteria (n = 5; 2%) in inguinal swabs; and vancomycin-resistant enterococci in an inguinal swab (n = 1; 0.4%). Previous antibiotic use was significantly associated with MDRO colonisation (OR 2.91, 95% CI 1.19-7.09, p = 0.018), mainly narrow spectrum oral beta-lactams and macrolides. S. malthophilia was polyclonal and human and environmental isolates differed. CONCLUSION Colonisation by MRSA, VRE, and ESBL-producing bacteria was low; however, 4% were surprisingly colonized by polyclonal S. maltophilia. This pathogen may also suggest using narrow-spectrum rather than the expected broad-spectrum antimicrobials. Antibiotic use was the only risk factor found, mainly with oral narrow-spectrum drugs.
Collapse
Affiliation(s)
- Vítor Falcão de Oliveira
- Division of Infectious Diseases, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil.
| | | | | | - Nazareno Scaccia
- Institute of Tropical Medicine, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ana Carolina Mamana
- Institute of Tropical Medicine, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Marina Farrel Côrtes
- Institute of Tropical Medicine, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Maura Salaroli de Oliveira
- Division of Infectious Diseases, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruno de Melo Tavares
- Division of Infectious Diseases, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Erika Regina Manuli
- Division of Infectious Diseases, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
- Institute of Tropical Medicine, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
- Departamento de Pesquisa Clínica E Inovação Em Saúde, Universidade Municipal de São Caetano Do Sul, Sao Paulo, Brazil
| | - Fábio Eudes Leal
- Departamento de Pesquisa Clínica E Inovação Em Saúde, Universidade Municipal de São Caetano Do Sul, Sao Paulo, Brazil
- Divisão de Pesquisa Clínica (DIPETEC), Instituto Nacional Do Câncer, Rio de Janeiro, Brazil
| | | | - Regina Maura Zetone Grespan
- Departamento de Pesquisa Clínica E Inovação Em Saúde, Universidade Municipal de São Caetano Do Sul, Sao Paulo, Brazil
| | - Cibele Cristine Remondes Sequeira
- Municipal Health Department, Primary Health System, Sao Caetano Do Sul, Sao Paulo, Brazil
- Departamento de Pesquisa Clínica E Inovação Em Saúde, Universidade Municipal de São Caetano Do Sul, Sao Paulo, Brazil
| | - Fatima L S Nunes
- Laboratory for Informatics Applications in Health, School of Arts, Humanities and Science, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Milena Dropa
- School of Public Health, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | | | - Ester Cerdeira Sabino
- Division of Infectious Diseases, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
- Institute of Tropical Medicine, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
- Departamento de Pesquisa Clínica E Inovação Em Saúde, Universidade Municipal de São Caetano Do Sul, Sao Paulo, Brazil
| | | | - Alison Holmes
- University of Liverpool and Imperial College London, London, UK
| | - Silvia F Costa
- Division of Infectious Diseases, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
- Institute of Tropical Medicine, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Anna S Levin
- Division of Infectious Diseases, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
- Institute of Tropical Medicine, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
41
|
Maciel-Guerra A, Babaarslan K, Baker M, Rahman A, Hossain M, Sadique A, Alam J, Uzzaman S, Ferdous Rahman Sarker M, Sultana N, Islam Khan A, Ara Begum Y, Hassan Afrad M, Senin N, Hossain Habib Z, Shirin T, Qadri F, Dottorini T. Core and accessory genomic traits of Vibrio cholerae O1 drive lineage transmission and disease severity. Nat Commun 2024; 15:8231. [PMID: 39313510 PMCID: PMC11420230 DOI: 10.1038/s41467-024-52238-0] [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: 03/28/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024] Open
Abstract
In Bangladesh, Vibrio cholerae lineages are undergoing genomic evolution, with increased virulence and spreading ability. However, our understanding of the genomic determinants influencing lineage transmission and disease severity remains incomplete. Here, we developed a computational framework using machine-learning, genome scale metabolic modelling (GSSM) and 3D structural analysis, to identify V. cholerae genomic traits linked to lineage transmission and disease severity. We analysed in-patients isolates from six Bangladeshi regions (2015-2021), and uncovered accessory genes and core SNPs unique to the most recent dominant lineage, with virulence, motility and bacteriophage resistance functions. We also found a strong correlation between V. cholerae genomic traits and disease severity, with some traits overlapping those driving lineage transmission. GSMM and 3D structure analysis unveiled a complex interplay between transcription regulation, protein interaction and stability, and metabolic networks, associated to lifestyle adaptation, intestinal colonization, acid tolerance and symptom severity. Our findings support advancing therapeutics and targeted interventions to mitigate cholera spread.
Collapse
Affiliation(s)
- Alexandre Maciel-Guerra
- School of Veterinary Medicine and Science, University of Nottingham, College Road, Sutton Bonington, Loughborough, Leicestershire, LE12 5RD, UK
| | - Kubra Babaarslan
- School of Veterinary Medicine and Science, University of Nottingham, College Road, Sutton Bonington, Loughborough, Leicestershire, LE12 5RD, UK
| | - Michelle Baker
- School of Veterinary Medicine and Science, University of Nottingham, College Road, Sutton Bonington, Loughborough, Leicestershire, LE12 5RD, UK
| | - Aura Rahman
- NSU Genome Research Institute (NGRI), North South University, Baridhara, Bashundhara, Dhaka, 1229, Bangladesh
| | - Maqsud Hossain
- School of Veterinary Medicine and Science, University of Nottingham, College Road, Sutton Bonington, Loughborough, Leicestershire, LE12 5RD, UK
- NSU Genome Research Institute (NGRI), North South University, Baridhara, Bashundhara, Dhaka, 1229, Bangladesh
| | - Abdus Sadique
- NSU Genome Research Institute (NGRI), North South University, Baridhara, Bashundhara, Dhaka, 1229, Bangladesh
| | - Jahidul Alam
- NSU Genome Research Institute (NGRI), North South University, Baridhara, Bashundhara, Dhaka, 1229, Bangladesh
| | - Salim Uzzaman
- Institute of Epidemiology, Disease Control and Research (IEDCR), 44, Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka, 1212, Bangladesh
| | - Mohammad Ferdous Rahman Sarker
- Institute of Epidemiology, Disease Control and Research (IEDCR), 44, Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka, 1212, Bangladesh
| | - Nasrin Sultana
- Institute of Epidemiology, Disease Control and Research (IEDCR), 44, Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka, 1212, Bangladesh
| | - Ashraful Islam Khan
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr, b), 68, Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka, 1212, Bangladesh
| | - Yasmin Ara Begum
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr, b), 68, Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka, 1212, Bangladesh
| | - Mokibul Hassan Afrad
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr, b), 68, Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka, 1212, Bangladesh
| | - Nicola Senin
- Department of Engineering, University of Perugia, 06125, Perugia, Italy
| | - Zakir Hossain Habib
- Institute of Epidemiology, Disease Control and Research (IEDCR), 44, Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka, 1212, Bangladesh
| | - Tahmina Shirin
- Institute of Epidemiology, Disease Control and Research (IEDCR), 44, Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka, 1212, Bangladesh
| | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr, b), 68, Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka, 1212, Bangladesh
| | - Tania Dottorini
- School of Veterinary Medicine and Science, University of Nottingham, College Road, Sutton Bonington, Loughborough, Leicestershire, LE12 5RD, UK.
- Centre for Smart Food Research, Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100, P. R. China.
| |
Collapse
|
42
|
Lim YC, Ong KH, Khor WC, Chua FYX, Lim JQ, Tan LK, Chen SL, Wong WK, Maiwald M, Barkham T, Koh TH, Khoo J, Chan JSH, Aung KT. Sequence Types and Antimicrobial Resistance Profiles of Salmonella Typhimurium in the Food Chain in Singapore. Microorganisms 2024; 12:1912. [PMID: 39338586 PMCID: PMC11434088 DOI: 10.3390/microorganisms12091912] [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: 09/02/2024] [Revised: 09/09/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Salmonella remains a significant foodborne pathogen globally with S. Typhimurium presenting as a frequently occurring serovar. This study aimed to characterize 67 S. Typhimurium isolates from humans, food, farms, and slaughterhouses collected in Singapore from 2016 to 2017. Using whole-genome sequencing analysis, the isolates were found to belong to either ST19 (n = 33) or ST36 (n = 34). ST36 predominated in human intestinal and chicken isolates, while human extra-intestinal and non-chicken food isolates belonged to ST19. Plasmids were predicted in 88.1% (n = 59) of the isolates with the most common incompatibility group profiles being IncFIB(S), IncFII(S) and IncQ1. IncFIB(S) (adjusted p-value < 0.05) and IncFII(S) (adjusted p-value < 0.05) were significantly more prevalent in ST19 isolates, while Col156 (adjusted p-value < 0.05) was more significantly found in ST36 isolates. ST36 isolates exhibited higher resistance to multiple antibiotic classes such as penicillins, phenicols, folate pathway inhibitors, aminoglycosides, β-lactam/β-lactamase inhibitor combinations, tetracyclines, and fluoroquinolones. Phylogenetics analysis suggested potential shared routes of transmission among human, chicken, farm and slaughterhouse environments. Taken together, this study offers a cross-sectional epidemiological insight into the genomic epidemiology and antimicrobial landscape of S. Typhimurium isolates in Singapore, informing strategies for future public health and food safety surveillance.
Collapse
Affiliation(s)
- Yen Ching Lim
- National Centre for Food Science, Singapore Food Agency, 7 International Business Park, Singapore 609919, Singapore
| | - Kar Hui Ong
- National Centre for Food Science, Singapore Food Agency, 7 International Business Park, Singapore 609919, Singapore
| | - Wei Ching Khor
- National Centre for Food Science, Singapore Food Agency, 7 International Business Park, Singapore 609919, Singapore
| | - Favian Yue Xuan Chua
- National Centre for Food Science, Singapore Food Agency, 7 International Business Park, Singapore 609919, Singapore
| | - Jia Qi Lim
- National Centre for Food Science, Singapore Food Agency, 7 International Business Park, Singapore 609919, Singapore
| | - Li Kiang Tan
- National Centre for Food Science, Singapore Food Agency, 7 International Business Park, Singapore 609919, Singapore
| | - Swaine L. Chen
- Infectious Diseases Translational Research Programme, Department of Medicine, Division of Infectious Diseases, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Singapore 119228, Singapore
- Laboratory of Bacterial Genomics, Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore
| | - Wai Kwan Wong
- Centre for Animal & Veterinary Service, National Parks Board, Singapore 718827, Singapore
| | - Matthias Maiwald
- Department of Pathology and Laboratory Medicine, KK Women’s and Children’s Hospital, Singapore 229899, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
| | - Timothy Barkham
- Department of Laboratory Medicine, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Tse Hsien Koh
- Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
- Department of Microbiology, Singapore General Hospital, Singapore 169856, Singapore
| | - Joanna Khoo
- National Centre for Food Science, Singapore Food Agency, 7 International Business Park, Singapore 609919, Singapore
| | - Joanne Sheot Harn Chan
- National Centre for Food Science, Singapore Food Agency, 7 International Business Park, Singapore 609919, Singapore
- Department of Food Science & Technology, National University of Singapore, Science Drive 2, Singapore 117542, Singapore
| | - Kyaw Thu Aung
- National Centre for Food Science, Singapore Food Agency, 7 International Business Park, Singapore 609919, Singapore
- Department of Food Science & Technology, National University of Singapore, Science Drive 2, Singapore 117542, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Dr, Singapore 637551, Singapore
| |
Collapse
|
43
|
Shi J, Xiao Y, Shen L, Wan C, Wang B, Zhou P, Zhang J, Han W, Yu F. Phenotypic and genomic analysis of the hypervirulent methicillin-resistant Staphylococcus aureus ST630 clone in China. mSystems 2024; 9:e0066424. [PMID: 39158330 PMCID: PMC11406941 DOI: 10.1128/msystems.00664-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/11/2024] [Indexed: 08/20/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) sequence type 630 (ST630) is a rarely reported lineage worldwide. This study aimed to trace the dissemination of the emerging MRSA ST630 clones in China and investigate their virulence potential. We collected 22 ST630-MRSA isolates from across China and performed whole-genome sequencing analysis and virulence characterization on these isolates. Epidemiological results showed that MRSA ST630 isolates were primarily isolated from pus/wound secretions, mainly originating from Jiangxi province, and carried diverse virulence and drug resistance genes. Staphylococcal cassette chromosome mec type V (SCCmec V) predominated (11/22, 50.0%) among the MRSA ST630 isolates. Interestingly, nearly half (45.5%) of the 22 ST630-MRSA isolates tested lacked intact SCCmec elements. Phylogenetic analysis demonstrated that ST630-MRSA could be divided into two distinct clades, with widespread dissemination mainly in Chinese regions. Five representative isolates were selected for phenotypic assays, including hemolysin activity, real-time fluorescence quantitative PCR, western blot analysis, hydrogen peroxide killing assay, blood killing assay, cell adhesion and invasion assay, and mouse skin abscess model. The results showed that, compared to the USA300-LAC strain, ST630 isolates exhibited particularly strong invasiveness and virulence in the aforementioned phenotypic assays. This study described the emergence of a highly virulent ST630-MRSA lineage and improved our insight into the molecular epidemiology of ST630 clones in China.IMPORTANCEMethicillin-resistant Staphylococcus aureus (MRSA) sequence type 630 (ST630) is an emerging clone with an increasing isolation rate in China. This study raises awareness of the hypervirulent MRSA ST630 clones in China and alerts people to their widespread dissemination. ST630-staphylococcal cassette chromosome mec V is a noteworthy clone in China, and we present the first comprehensive genetic and phenotypic analysis of this lineage. Our findings provide valuable insights for the prevention and control of infections caused by this emerging MRSA clone.
Collapse
Affiliation(s)
- Junhong Shi
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cailing Wan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peiyao Zhou
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weihua Han
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
44
|
Liu Y, Wang Y, Wei F, Chai L, Wang H. Gut microbiota-bile acid crosstalk contributes to intestinal damage after nitrate exposure in Bufo gargarizans tadpoles. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 943:173795. [PMID: 38851338 DOI: 10.1016/j.scitotenv.2024.173795] [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: 01/10/2024] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Bile acids (BAs) are amphipathic steroid acids whose production and diversity depend on both host and microbial metabolism. Nitrate (NO3-) is a widespread pollutant in aquatic ecosystems, which can cause rapid changes in microbial community structure and function. However, the effect of gut microbiota reshaped by nitrate‑nitrogen (NO3-N) on BAs profiles remains unclarified. To test this, intestinal targeted BAs metabolomics and fecal metagenomic sequencing were performed on Bufo gargarizans tadpoles treated with different concentrations of NO3-N. NO3-N exposure induced a reduction in the abundance of microbiota with bile acid-inducible enzymes (BAIs) and/or hydroxysteroid dehydrogenases (HSDHs), thus inhibiting the conversion of primary BAs to secondary BAs. Inhibition of BAs biotransformation decreased protective hydrophilic BAs (UDCA) and increased toxic hydrophobic BAs (CA and CDCA), which may contribute to intestinal histopathological damage. Moreover, we found that NO3-N treatment increased microbial virulence factors and decreased Glycoside hydrolases, further highlighting the deleterious risk of NO3-N. Overall, this study shed light on the complex interactions of NO3-N, gut microbiota, and BAs, and emphasized the hazardous effects of NO3-N pollution on the health of amphibians.
Collapse
Affiliation(s)
- Ying Liu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yaxi Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Fei Wei
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Lihong Chai
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Chang'an University, Xi'an 710054, China
| | - Hongyuan Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
45
|
Dalby MJ, Kiu R, Serghiou IR, Miyazaki A, Acford-Palmer H, Tung R, Caim S, Phillips S, Kujawska M, Matsui M, Iwamoto A, Taking B, Cox SE, Hall LJ. Faecal microbiota and cytokine profiles of rural Cambodian infants linked to diet and diarrhoeal episodes. NPJ Biofilms Microbiomes 2024; 10:85. [PMID: 39277573 PMCID: PMC11401897 DOI: 10.1038/s41522-024-00562-0] [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: 01/19/2024] [Accepted: 08/28/2024] [Indexed: 09/17/2024] Open
Abstract
The gut microbiota of infants in low- to middle-income countries is underrepresented in microbiome research. This study explored the faecal microbiota composition and faecal cytokine profiles in a cohort of infants in a rural province of Cambodia and investigated the impact of sample storage conditions and infant environment on microbiota composition. Faecal samples collected at three time points from 32 infants were analysed for microbiota composition using 16S rRNA amplicon sequencing and concentrations of faecal cytokines. Faecal bacterial isolates were subjected to whole genome sequencing and genomic analysis. We compared the effects of two sample collection methods due to the challenges of faecal sample collection in a rural location. Storage of faecal samples in a DNA preservation solution preserved Bacteroides abundance. Microbiota analysis of preserved samples showed that Bifidobacterium was the most abundant genus with Bifidobacterium longum the most abundant species, with higher abundance in breast-fed infants. Most infants had detectable pathogenic taxa, with Shigella and Klebsiella more abundant in infants with recent diarrhoeal illness. Neither antibiotics nor infant growth were associated with gut microbiota composition. Genomic analysis of isolates showed gene clusters encoding the ability to digest human milk oligosaccharides in B. longum and B. breve isolates. Antibiotic-resistant genes were present in both potentially pathogenic species and in Bifidobacterium. Faecal concentrations of Interlukin-1alpha and vascular endothelial growth factor were higher in breast-fed infants. This study provides insights into an underrepresented population of rural Cambodian infants, showing pathogen exposure and breastfeeding impact gut microbiota composition and faecal immune profiles.
Collapse
Affiliation(s)
- Matthew J Dalby
- Microbes, Infection and Microbiomes, College of Medicine and Health, University of Birmingham, Birmingham, B15 2TT, UK
- Food, Microbiome & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Raymond Kiu
- Microbes, Infection and Microbiomes, College of Medicine and Health, University of Birmingham, Birmingham, B15 2TT, UK
- Food, Microbiome & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Iliana R Serghiou
- Food, Microbiome & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Asuka Miyazaki
- School of Tropical Medicine & Global Health, Nagasaki University, Nagasaki, Japan
| | - Holly Acford-Palmer
- Food, Microbiome & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Rathavy Tung
- National Maternal and Child Health Centre, Ministry of Health, Phnom Penh, Cambodia
| | - Shabhonam Caim
- Food, Microbiome & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Sarah Phillips
- Food, Microbiome & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Magdalena Kujawska
- Microbes, Infection and Microbiomes, College of Medicine and Health, University of Birmingham, Birmingham, B15 2TT, UK
- Food, Microbiome & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food & Health, Technical University of Munich, Freising, 80333, Germany
| | - Mitsuaki Matsui
- School of Tropical Medicine & Global Health, Nagasaki University, Nagasaki, Japan
| | - Azusa Iwamoto
- Bureau of International Health Cooperation, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Bunsreng Taking
- Kampong Cham Provincial Health Department, Ministry of Health, Kampong Cham, Cambodia
| | - Sharon E Cox
- School of Tropical Medicine & Global Health, Nagasaki University, Nagasaki, Japan
- London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
- Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Lindsay J Hall
- Microbes, Infection and Microbiomes, College of Medicine and Health, University of Birmingham, Birmingham, B15 2TT, UK.
- Food, Microbiome & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK.
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food & Health, Technical University of Munich, Freising, 80333, Germany.
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
46
|
Varney AM, Smitten KL, Southam HM, Fairbanks SD, Robertson CC, Thomas JA, McLean S. In Vitro and In Vivo Studies on a Mononuclear Ruthenium Complex Reveals It is a Highly Effective, Fast-Acting, Broad-Spectrum Antimicrobial in Physiologically Relevant Conditions. ACS Infect Dis 2024; 10:3346-3357. [PMID: 39106475 PMCID: PMC11406528 DOI: 10.1021/acsinfecdis.4c00447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
Abstract
The crystal structure of a previously reported antimicrobial RuII complex that targets bacterial DNA is presented. Studies utilizing clinical isolates of Gram-negative bacteria that cause catheter-associated urinary tract infection, (CA)UTI, in media that model urine and plasma reveal that good antimicrobial activity is maintained in all conditions tested. Experiments with a series of Staphylococcus aureus clinical isolates show that, unlike the majority of previously reported RuII-based antimicrobial leads, the compound retains its potent activity even in MRSA strains. Furthermore, experiments using bacteria in early exponential growth and at different pHs reveal that the compound also retains its activity across a range of conditions that are relevant to those encountered in clinical settings. Combinatorial studies involving cotreatment with conventional antibiotics or a previously reported analogous dinuclear RuII complex showed no antagonistic effects. In fact, although all combinations show distinct additive antibacterial activity, in one case, this effect approaches synergy. It was found that the Galleria Mellonella model organism infected with a multidrug resistant strain of the ESKAPE pathogen Acinetobacter baumannii could be successfully treated and totally cleared within 48 h after a single dose of the lead complex with no detectable deleterious effect to the host.
Collapse
Affiliation(s)
- Adam M Varney
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, U.K
- Medical Technologies Innovation Facility (MTIF), Clifton Lane, Nottingham NG11 8NS, U.K
| | - Kirsty L Smitten
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
- School of Bioscience, The University of Sheffield, Western Bank, Sheffield S10 2TN, U.K
| | - Hannah M Southam
- School of Bioscience, The University of Sheffield, Western Bank, Sheffield S10 2TN, U.K
| | - Simon D Fairbanks
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Craig C Robertson
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Jim A Thomas
- Department of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Samantha McLean
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, U.K
| |
Collapse
|
47
|
Salamzade R, Tran PQ, Martin C, Manson AL, Gilmore MS, Earl AM, Anantharaman K, Kalan LR. zol & fai: large-scale targeted detection and evolutionary investigation of gene clusters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.07.544063. [PMID: 37333121 PMCID: PMC10274777 DOI: 10.1101/2023.06.07.544063] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Many universally and conditionally important genes are genomically aggregated within clusters. Here, we introduce fai and zol, which together enable large-scale comparative analysis of different types of gene clusters and mobile-genetic elements (MGEs), such as biosynthetic gene clusters (BGCs) or viruses. Fundamentally, they overcome a current bottleneck to reliably perform comprehensive orthology inference at large scale across broad taxonomic contexts and thousands of genomes. First, fai allows the identification of orthologous instances of a query gene cluster of interest amongst a database of target genomes. Subsequently, zol enables reliable, context-specific inference of ortholog groups for individual protein-encoding genes across gene cluster instances. In addition, zol performs functional annotation and computes a variety of evolutionary statistics for each inferred ortholog group. Importantly, in comparison to tools for visual exploration of homologous relationships between gene clusters, zol can scale to thousands of gene cluster instances and produce detailed reports that are easy to digest. To showcase fai and zol, we apply them for: (i) longitudinal tracking of a virus in metagenomes, (ii) discovering novel population-level genetic insights of two common BGCs in the fungal species Aspergillus flavus, and (iii) uncovering large-scale evolutionary trends of a virulence-associated gene cluster across thousands of genomes from a diverse bacterial genus.
Collapse
Affiliation(s)
- Rauf Salamzade
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Patricia Q. Tran
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Freshwater and Marine Science Doctoral Program, University of Wisconsin-Madison, WI, USA
| | - Cody Martin
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Abigail L. Manson
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Michael S. Gilmore
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Ophthalmology, Harvard Medical School and Mass Eye and Ear, Boston, Massachusetts, USA
- Department of Microbiology, Harvard Medical School and Mass Eye and Ear, Boston, Massachusetts, USA
| | - Ashlee M. Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Lindsay R. Kalan
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, Division of Infectious Disease, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
48
|
Kim AR, Jeon SG, Kim HR, Hong H, Yoon YW, Lee BM, Yoon CH, Choi SJ, Jang MH, Yang BG. Preventive and Therapeutic Effects of Lactiplantibacillus plantarum HD02 and MD159 through Mast Cell Degranulation Inhibition in Mouse Models of Atopic Dermatitis. Nutrients 2024; 16:3021. [PMID: 39275335 PMCID: PMC11396792 DOI: 10.3390/nu16173021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/16/2024] Open
Abstract
As the relationship between the gut microbiome and allergies becomes better understood, targeted strategies to prevent and treat allergies through gut microbiome modulation are being increasingly developed. In the study presented herein, we screened various probiotics for their ability to inhibit mast cell degranulation and identified Lactiplatibacillus plantarum HD02 and MD159 as effective candidates. The two strains significantly attenuated vascular permeability induced by mast cell degranulation in a passive cutaneous anaphylaxis (PCA) model and, in the MC903-induced murine atopic dermatitis (AD) model, demonstrated comparable preventive effects against allergies, reducing blood levels of MCPT-1 (mast cell protease-1) and total IgE. In the house dust mite (HDM)-induced murine AD model, both L. plantarum HD02 and MD159 showed therapeutic effects, with L. plantarum HD02 demonstrating superior efficacy. Nevertheless, L. plantarum MD159 better suppressed transepidermal water loss (TEWL). Furthermore, L. plantarum HD02 and MD159 significantly increased the number of splenic Foxp3+ regulatory T cells, with L. plantarum MD159 having a more pronounced effect. However, only L. plantarum HD02 achieved a reduction in immune cells in the draining lymph nodes. Our findings highlight L. plantarum HD02 and MD159 as promising candidates for the prevention and treatment of allergies, demonstrating significant efficacy in suppressing mast cell degranulation, reducing the number of allergy biomarkers, and modulating immune responses in experimental models of AD. Their distinct mechanisms of action suggest potential complementary roles in addressing allergic diseases, underscoring their therapeutic promise in clinical applications.
Collapse
Affiliation(s)
- A-Ram Kim
- Research Institute, GI Biome Inc., Seongnam 13201, Republic of Korea
| | - Seong-Gak Jeon
- Research Institute, GI Biome Inc., Seongnam 13201, Republic of Korea
| | - Hyung-Ran Kim
- Research Institute, GI Biome Inc., Seongnam 13201, Republic of Korea
| | - Heeji Hong
- Research Institute, GI Biome Inc., Seongnam 13201, Republic of Korea
| | - Yong Won Yoon
- Maeil Innovation Center, Maeil Dairies Co., Ltd., Pyeongtaek 17714, Republic of Korea
| | - Byung-Min Lee
- Maeil Innovation Center, Maeil Dairies Co., Ltd., Pyeongtaek 17714, Republic of Korea
| | - Chung Hoo Yoon
- Maeil Innovation Center, Maeil Dairies Co., Ltd., Pyeongtaek 17714, Republic of Korea
| | - Soo Jin Choi
- Maeil Innovation Center, Maeil Dairies Co., Ltd., Pyeongtaek 17714, Republic of Korea
| | - Myoung Ho Jang
- Research Institute, GI Innovation Inc., Seoul 05855, Republic of Korea
| | - Bo-Gie Yang
- Research Institute, GI Biome Inc., Seongnam 13201, Republic of Korea
| |
Collapse
|
49
|
Sun J, Yin H, Ju C, Wang Y, Yang Z. DTVF: A User-Friendly Tool for Virulence Factor Prediction Based on ProtT5 and Deep Transfer Learning Models. Genes (Basel) 2024; 15:1170. [PMID: 39336761 PMCID: PMC11430887 DOI: 10.3390/genes15091170] [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: 08/07/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Virulencefactors (VFs) are key molecules that enable pathogens to evade the immune systems of the host. These factors are crucial for revealing the pathogenic processes of microbes and drug discovery. Identification of virulence factors in microbes become an important problem in the field of bioinformatics. To address this problem, this study proposes a novel model DTVF (Deep Transfer Learning for Virulence Factor Prediction), which integrates the ProtT5 protein sequence extraction model with a dual-channel deep learning model. In the dual-channel deep learning model, we innovatively integrate long short-term memory (LSTM) with convolutional neural networks (CNNs), creating a novel integrated architecture. Furthermore, by incorporating the attention mechanism, the accuracy of VF detection was significantly enhanced. We evaluated the DTVF model against other excellent-performing models in the field. DTVF demonstrates superior performance, achieving an accuracy rate of 84.55% and an AUROC of 92.08% on the benchmark dataset. DTVF shows state-of-the-art performance in this field, surpassing the existing models in nearly all metrics. To facilitate the use of biologists, we have also developed an interactive web-based user interface version of DTVF based on Gradio.
Collapse
Affiliation(s)
- Jiawei Sun
- School of Artificial Intelligence, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Hongbo Yin
- School of Geography, University of Leeds, Leeds LS2 9JT, UK
| | - Chenxiao Ju
- School of Electrical and Computer Engineering, University of Sydney, Camperdown, NSW 2006, Australia
| | - Yongheng Wang
- School of Artificial Intelligence, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Zhiyuan Yang
- School of Artificial Intelligence, Hangzhou Dianzi University, Hangzhou 310018, China
| |
Collapse
|
50
|
Xie ST, Zhu D, Song YQ, Zhu YG, Ding LJ. Unveiling potential roles of earthworms in mitigating the presence of virulence factor genes in terrestrial ecosystems. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135133. [PMID: 38986408 DOI: 10.1016/j.jhazmat.2024.135133] [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: 03/28/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
Earthworms can redistribute soil microbiota, and thus might affect the profile of virulence factor genes (VFGs) which are carried by pathogens in soils. Nevertheless, the knowledge of VFG profile in the earthworm guts and its interaction with earthworm gut microbiome is still lacking. Herein, we characterized earthworm gut and soil microbiome and VFG profiles in natural and agricultural ecosystems at a national scale using metagenomics. VFG profiles in the earthworm guts significantly differed from those in the surrounding soils, which was mainly driven by variations of bacterial communities. Furthermore, the total abundance of different types of VFGs in the earthworm guts was about 20-fold lower than that in the soils due to the dramatic decline (also by approximately 20-fold) of VFG-carrying bacterial pathogens in the earthworm guts. Additionally, five VFGs related to nutritional/metabolic factors and stress survival were identified as keystones merely in the microbe-VFG network in the earthworm guts, implying their pivotal roles in facilitating pathogen colonization in earthworm gut microhabitats. These findings suggest the potential roles of earthworms in reducing risks related to the presence of VFGs in soils, providing novel insights into earthworm-based bioremediation of VFG contamination in terrestrial ecosystems.
Collapse
Affiliation(s)
- Shu-Ting Xie
- State Key Laboratory of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Dong Zhu
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China; Key Laboratory of Urban Environment and Health, Ningbo Urban Environment Observation and Research Station, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China; Zhejiang Key Laboratory of Urban Environmental Processes and Pollution Control, CAS Haixi Industrial Technology Innovation Center in Beilun, Ningbo 315830, China
| | - Ya-Qiong Song
- State Key Laboratory of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Sino-Danish College of University of Chinese Academy of Sciences, Beijing 101400, China; Sino-Danish Centre for Education and Research, Beijing 100049, China; Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg, Denmark
| | - Yong-Guan Zhu
- State Key Laboratory of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China; Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Jimei District, Xiamen 361021, China
| | - Long-Jun Ding
- State Key Laboratory of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China.
| |
Collapse
|