1
|
Schellong P, Joean O, Pletz MW, Hagel S, Weis S. Treatment of Complicated Gram-Positive Bacteremia and Infective Endocarditis. Drugs 2024:10.1007/s40265-024-02135-z. [PMID: 39720961 DOI: 10.1007/s40265-024-02135-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2024] [Indexed: 12/26/2024]
Abstract
The Gram-positive cocci Staphylococcus aureus, Streptococcus spp., and Enterococcus spp. are the most frequent causative organisms of bloodstream infections and infective endocarditis. "Complicated bacteremia" is a term used in S. aureus bloodstream infections and originally implied the presence of metastatic infectious foci (i.e. complications of S. aureus bacteremia). These complications demand longer antimicrobial treatment durations and, frequently, interventional source control. Several risk factors for the incidence of bacteremia complications have been identified and are often used for the definition of complicated bacteremia. Here, we discuss management and diagnostic approaches and treatment options for patients with complicated bacteremia, with particular focus on infective endocarditis. We also summarize the available evidence regarding imaging modalities and the choice of antimicrobial mono- or combination therapy according to resistance patterns for these pathogens as well as treatment durations and optimized application routes. Finally, we synopsize current and future areas of research in complicated bacteremia and infective endocarditis.
Collapse
Affiliation(s)
- Paul Schellong
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany.
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany.
| | - Oana Joean
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
| | - Mathias W Pletz
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
| | - Stefan Hagel
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
| | - Sebastian Weis
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
2
|
Willett JLE, Dunny GM. Insights into ecology, pathogenesis, and biofilm formation of Enterococcus faecalis from functional genomics. Microbiol Mol Biol Rev 2024:e0008123. [PMID: 39714182 DOI: 10.1128/mmbr.00081-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
Abstract
SUMMARYEnterococcus faecalis is a significant resident of the gastrointestinal tract of most animals, including humans. Although generally non-pathogenic in healthy hosts, this microbe is adept at the exploitation of compromises in host immune functions, resulting in life-threatening opportunistic infections whose treatments are complicated by a high degree of intrinsic and acquired resistance to antimicrobial chemotherapy. Historically, progress in enterococcal research was limited by a lack of experimental models that replicate natural infection pathways and the relevance of in vitro studies to the natural biology of the organism. In this review, we summarize the history of enterococcal research during the 20th and early 21st centuries and describe more recent genetic and genomic tools and screens developed to address challenges in the field. We also describe how the results of recent studies reveal the importance of previously uncharacterized enterococcal genes, and we provide examples of interesting determinants that have emerged as important contributors to enterococcal biology. These factors may also serve as targets for future vaccines and chemotherapeutic agents to combat life-threatening hospital infections.
Collapse
Affiliation(s)
- Julia L E Willett
- Department of Microbiology & Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Gary M Dunny
- Department of Microbiology & Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Fu Y, Deng Z, Shen Y, Wei W, Xiang Q, Liu Z, Hanf K, Huang S, Lv Z, Cao T, Peng C, Zhang R, Zou X, Shen J, Schwarz S, Wang Y, Liu D, Lv Z, Ke Y. High prevalence and plasmidome diversity of optrA-positive enterococci in a Shenzhen community, China. Front Microbiol 2024; 15:1505107. [PMID: 39760083 PMCID: PMC11695379 DOI: 10.3389/fmicb.2024.1505107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/21/2024] [Indexed: 01/07/2025] Open
Abstract
Background The emergence of optrA, which can confer resistance to phenicols and oxazolidinones in Enterococcus spp., poses a growing public health threat. Methods 102 optrA-positive enterococci (OPEs) including various species were isolated from feces of 719 healthy volunteers in a Shenzhen community, China. Antimicrobial susceptibility of these isolates was tested. Whole-genome sequencing and bioinformatics analysis were performed to characterize molecular epidemiology of OPEs. Results Compared to optrA-negative enterococci (ONEs), antimicrobial resistance (linezolid, florfenicol, doxycycline, erythromycin and ciprofloxacin) and presence of antimicrobial resistance genes (ARGs) (fexA, cat, tet(M), erm(A), erm(B) and etc) were higher in OPEs. Phylogenetic analysis revealed that high similarly (19-338 SNPs) was observed between the optrA-positive E. faecalis from community and the strains from patients, animals, and environment. In 102 OPEs, the optrA gene was detected on the chromosome (n = 36), on plasmids (n = 62), or both (n = 4). A diverse range of optrA-carrying plasmid types was identified. The rep9-plasmid replicons were widely detected in E. faecalis (44/66), whereas repUS1-plasmid replicons were widely identified in other enterococcal species (7/66). Most of all ARGs harbored by isolates were co-existed on optrA-carrying plasmids, suggesting that the acquisition of optrA-carrying plasmids will pose a greater threat to public health. Notably, the pAD1 (rep9 family) + DOp1-type plasmids should receive more attention for the transfer of optrA given their high prevalence (36.36%), high number of co-located ARGs with optrA (83.87% of total ARGs) and presence in multiple sources. Tn6674, IS1216E, ISEnfa1 and ISEnfa5 are related to the transfer of chromosomal and plasmids-derived optrA, respectively. The bcrABDR gene cluster, fexA, and erm(A) were frequently identified surrounding optrA and may be transferred with optrA via IS1216E or ISEnfa1. Conclusion The transfer of optrA gene is related to a variety of mobile elements (including plasmids, insertion sequences, transposons), which will promote the horizontal transfer of optrA. Moreover, many ARGs co-exist with optrA and could co-transfer with optrA. The acquisition of OPEs and optrA-carrying plasmids will pose a greater threat to public health and should be obtained more attention, especially optrA-positive E. faecalis and pAD1 + DOp1-type plasmids.
Collapse
Affiliation(s)
- Yulin Fu
- Shenzhen Centre for Disease Control and Prevention, Shenzhen, China
| | - Zhaoju Deng
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yingbo Shen
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Weizhou Wei
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Qiumei Xiang
- Siming Centre for Disease Control and Prevention, Xiamen, China
| | - Zhiyang Liu
- Beijing University of Chinese Medicine Shenzhen Hospital (Longgang), Shenzhen, China
| | - Kunning Hanf
- Department of Neurology, Shenzhen People's Hospital, Shenzhen, China
| | - Suli Huang
- Shenzhen Centre for Disease Control and Prevention, Shenzhen, China
- School of Public Health, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Zexun Lv
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Tingting Cao
- Shenzhen Centre for Disease Control and Prevention, Shenzhen, China
| | - Changfeng Peng
- Shenzhen Centre for Disease Control and Prevention, Shenzhen, China
| | - Rong Zhang
- Department of Clinical Laboratory, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuan Zou
- Shenzhen Centre for Disease Control and Prevention, Shenzhen, China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Stefan Schwarz
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Institute of Microbiology and Epizootics, Center for Infection Medicine, School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Yang Wang
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dejun Liu
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ziquan Lv
- Shenzhen Centre for Disease Control and Prevention, Shenzhen, China
| | - Yuebin Ke
- Shenzhen Centre for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
4
|
Egan MS, de Macedo R, Zackular JP. Metals in the gut: microbial strategies to overcome nutritional immunity in the intestinal tract. Metallomics 2024; 16:mfae052. [PMID: 39577845 DOI: 10.1093/mtomcs/mfae052] [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: 06/11/2024] [Accepted: 11/20/2024] [Indexed: 11/24/2024]
Abstract
Trace metals are indispensable nutritional factors for all living organisms. During host-pathogen interactions, they serve as crucial resources that dictate infection outcomes. Accordingly, the host uses a defense strategy known as nutritional immunity, which relies on coordinated metal chelation to mitigate bacterial advances. In response, pathogens employ complex strategies to secure these resources at sites of infection. In the gastrointestinal (GI) tract, the microbiota must also acquire metals for survival, making metals a central line of competition in this complex ecosystem. In this minireview, we outline how bacteria secure iron, zinc, and manganese from the host with a focus on the GI tract. We also reflect on how host dietary changes impact disease outcomes and discuss therapeutic opportunities to target bacterial metal uptake systems. Ultimately, we find that recent discoveries on the dynamics of transition metals at the host-pathogen-microbiota interface have reshaped our understanding of enteric infections and provided insights into virulence strategies, microbial cooperation, and antibacterial strategies.
Collapse
Affiliation(s)
- Marisa S Egan
- Division of Protective Immunity, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biology, Swarthmore College, Swarthmore, PA 19081, USA
| | - Raquel de Macedo
- Division of Protective Immunity, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Microbiology, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, SP 01224-001, Brazil
| | - Joseph P Zackular
- Division of Protective Immunity, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Center for Microbial Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Sevillano E, Lafuente I, Peña N, Cintas LM, Muñoz-Atienza E, Hernández PE, Borrero J. Isolation, Genomics-Based and Biochemical Characterization of Bacteriocinogenic Bacteria and Their Bacteriocins, Sourced from the Gastrointestinal Tract of Meat-Producing Pigs. Int J Mol Sci 2024; 25:12210. [PMID: 39596276 PMCID: PMC11594732 DOI: 10.3390/ijms252212210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Antimicrobial resistance (AMR) poses a significant challenge to animal production due to the widespread use of antibiotics. Therefore, there is an urgent need for alternative antimicrobial strategies to effectively manage bacterial infections, protect animal health, and reduce reliance on antibiotics. This study evaluated the use of emerging approaches and procedures for the isolation, identification, and characterization of bacteriocin-producing bacteria and their bacteriocins, sourced from the gastrointestinal tract (GIT) of meat-producing pigs. Out of 2056 isolates screened against Gram-positive and Gram-negative indicator strains, 20 of the most active antimicrobial isolates were subjected to whole genome sequencing (WGS) for the prediction of coding DNA sequences (CDS) and the identification of bacteriocin gene clusters (BGC) and their functions. The use of an in vitro cell-free protein synthesis (IV-CFPS) protocol and the design of an IV-CFPS coupled to a split-intein mediated ligation (IV-CFPS/SIML) procedure made possible the evaluation of the production and antimicrobial activity of described and putatively novel bacteriocins. A colony MALDI-TOF MS procedure assisted in the identification of class I, II, and III lanthipeptides. MALDI-TOF MS and a targeted proteomics, combined with a massive peptide analysis (LC-MS/MS) approach, has proven valuable for the identification and biochemical characterization of previously described and novel bacteriocins encoded by the isolated bacteriocin-producing strains.
Collapse
Affiliation(s)
| | | | | | | | - Estefanía Muñoz-Atienza
- Departamento de Nutrición y Ciencia de los Alimentos (NUTRYCIAL), Sección Departamental de Nutrición y Ciencia de los Alimentos (SD-NUTRYCIAL), Facultad de Veterinaria, Universidad Complutense de Madrid (UCM), Avenida Puerta de Hierro, s/n, 28040 Madrid, Spain; (E.S.); (I.L.); (N.P.); (L.M.C.); (P.E.H.); (J.B.)
| | | | | |
Collapse
|
6
|
Petrilla A, Nemeth P, Fauszt P, Szilagyi-Racz A, Mikolas M, Szilagyi-Tolnai E, David P, Stagel A, Gal F, Gal K, Sohajda R, Pham T, Stundl L, Biro S, Remenyik J, Paholcsek M. Comparative analysis of the postadmission and antemortem oropharyngeal and rectal swab microbiota of ICU patients. Sci Rep 2024; 14:27179. [PMID: 39516251 PMCID: PMC11549221 DOI: 10.1038/s41598-024-78102-1] [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: 04/12/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Shotgun metabarcoding was conducted to examine the microbiota in a total of 48 samples from 12 critically ill patients, analyzing samples from both the oropharynx and rectum. We aimed to compare their postadmission microbiota, characterized as moderately dysbiotic, with the severely dysbiotic antemortem microbiota associated with patients' deaths. We found that, compared with postadmission samples, patient antemortem swab samples presented moderate but not significantly decreased diversity indices. The antemortem oropharyngeal samples presented an increase in biofilm-forming bacteria, including Streptococcus oralis, methicillin-resistant Staphylococcus aureus (MRSA), and Enterococcus faecalis. Although the septic shock rate was 67%, no significant differences were detected in the potential pathogen ratios when the microbiota was analyzed. A notable strain-sharing rate between the oropharynx and intestine was noted. By comparing postadmission and antemortem samples, microbial biomarkers of severe dysbiosis were pinpointed through the analysis of differentially abundant and uniquely emerging species in both oropharyngeal and rectal swabs. Demonstrating strong interconnectivity along the oral-intestinal axis, these biomarkers could serve as indicators of the progression of dysbiosis. Furthermore, the microbial networks of the oropharyngeal microbiota in deceased patients presented the lowest modularity, suggesting a vulnerable community structure. Our data also highlight the critical importance of introducing treatments aimed at enhancing the resilience of the oral cavity microbiome, thereby contributing to better patient outcomes.
Collapse
Affiliation(s)
- Annamaria Petrilla
- Department of Anaesthesiology and Intensive Care, Vas County Markusovszky Teaching Hospital, Szombathely, Hungary
| | - Peter Nemeth
- Department of Anaesthesiology and Intensive Care, Vas County Markusovszky Teaching Hospital, Szombathely, Hungary
| | - Peter Fauszt
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Anna Szilagyi-Racz
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Maja Mikolas
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Emese Szilagyi-Tolnai
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Peter David
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Aniko Stagel
- Hungarian National Blood Transfusion Service Nucleic Acid Testing Laboratory, Budapest, Hungary
| | - Ferenc Gal
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Kristof Gal
- Department of Oncoradiology, University of Debrecen Clinical Centre, Debrecen, Hungary
| | - Reka Sohajda
- Hungarian National Blood Transfusion Service Nucleic Acid Testing Laboratory, Budapest, Hungary
| | - Trinh Pham
- Turku Bioscience Centre, University of Turku and Abo Akademi University, 20520, Turku, Finland
| | - Laszlo Stundl
- Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Debrecen, Hungary
| | - Sandor Biro
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Remenyik
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Melinda Paholcsek
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
7
|
Wei Y, Palacios Araya D, Palmer KL. Enterococcus faecium: evolution, adaptation, pathogenesis and emerging therapeutics. Nat Rev Microbiol 2024; 22:705-721. [PMID: 38890478 DOI: 10.1038/s41579-024-01058-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2024] [Indexed: 06/20/2024]
Abstract
The opportunistic pathogen Enterococcus faecium colonizes humans and a wide range of animals, endures numerous stresses, resists antibiotic treatment and stubbornly persists in clinical environments. The widespread application of antibiotics in hospitals and agriculture has contributed to the emergence of vancomycin-resistant E. faecium, which causes many hospital-acquired infections. In this Review, we explore recent discoveries about the evolutionary history, the environmental adaptation and the colonization and dissemination mechanisms of E. faecium and vancomycin-resistant E. faecium. These studies provide critical insights necessary for developing novel preventive and therapeutic approaches against vancomycin-resistant E. faecium and also reveal the intricate interrelationships between the environment, the microorganism and the host, providing knowledge that is broadly relevant to how antibiotic-resistant pathogens emerge and endure.
Collapse
Affiliation(s)
- Yahan Wei
- School of Podiatric Medicine, The University of Texas Rio Grande Valley, Harlingen, TX, USA
| | - Dennise Palacios Araya
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Kelli L Palmer
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA.
| |
Collapse
|
8
|
Fathima F, Subramaniyan Y, Rai A, Rekha PD. Enterococcus faecalis co-cultured with oral cancer cells exhibits higher virulence and promotes cancer cell survival, proliferation, and migration: an in vitro study. J Med Microbiol 2024; 73. [PMID: 39585322 DOI: 10.1099/jmm.0.001931] [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: 11/26/2024] Open
Abstract
Introduction. Enterococcus faecalis is a common pathogen associated with many oral diseases and is often isolated from oral cancer patients. However, limited information is available on its key virulence gene expression in oral cancer cell microenvironment and cancer cell behaviour in co-culture studies.Hypothesis. E. faecalis overexpresses virulence genes when co-cultured with oral cancer cells and possibly alters the tumour microenvironment, promoting oral cancer proliferation and survival.Aim. To investigate altered virulence gene expression in E. faecalis and oral cancer cell behaviour using in vitro co-culture experiments.Methodology. Cal27 cells were co-cultured with E. faecalis and assessed for their cell proliferation, apoptosis, migration and clonogenicity using standard cell culture assays. The levels of reactive oxygen species (ROS) and inflammatory cytokines, along with proliferative, angiogenic and apoptotic biomarker expressions, were also assessed. E. faecalis adherence to cancer cells was demonstrated by the gentamicin protection assay. Real time-PCR was used to analyse the expression of virulence genes.Results. Co-culture of Cal27 cells with E. faecalis showed significantly higher cell proliferation, migration and clonogenicity compared to the control (P<0.01). A significant increase in the levels of ROS and inflammatory cytokines and overexpression of Ki67, vascular endothelial growth factor, extracellular signal-regulated kinase 1/2, phosphoinositide 3 kinase and Akt was observed in the co-culture group. E. faecalis also downregulated p53 and Bax genes while upregulated Bcl-2. The virulence genes GelE, Asa and Ace were overexpressed in E. faecalis co-cultured with Cal27 cells.Conclusion. The results from this study indicate the possible risks of E. faecalis infection in oral cancer. An effective antibiotic strategy against E. faecalis to prevent complications associated with oral diseases, including cancer, is needed.
Collapse
Affiliation(s)
- Fida Fathima
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Yuvarajan Subramaniyan
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Akshatha Rai
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Punchappady Devasya Rekha
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| |
Collapse
|
9
|
Tedim AP, Almeida-Santos AC, Lanza VF, Novais C, Coque TM, Freitas AR, Peixe L. Bacteriocin distribution patterns in Enterococcus faecium and Enterococcus lactis: bioinformatic analysis using a tailored genomics framework. Appl Environ Microbiol 2024; 90:e0137624. [PMID: 39283104 PMCID: PMC11497781 DOI: 10.1128/aem.01376-24] [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: 07/12/2024] [Accepted: 08/05/2024] [Indexed: 10/25/2024] Open
Abstract
Multidrug-resistant Enterococcus faecium strains represent a major concern due to their ability to thrive in diverse environments and cause life-threatening infections. While antimicrobial resistance and virulence mechanisms have been extensively studied, the contribution of bacteriocins to E. faecium's adaptability remains poorly explored. E. faecium, within the Bacillota phylum, is a prominent bacteriocin producer. Here, we developed a tailored database of 76 Bacillota bacteriocins (217 sequences, including 40 novel bacteriocins) and applied it to uncover bacteriocin distribution patterns in 997 quality-filtered E. faecium and Enterococcus lactis (former E. faecium clade B) genomes. Curated using computational pipelines and literature mining, our database demonstrates superior precision versus leading public tools in identifying diverse bacteriocins. Distinct bacteriocin profiles emerged between E. faecium and E. lactis, highlighting species-specific adaptations. E. faecium strains from hospitalized patients were significantly enriched in bacteriocins as enterocin A and bacteriocins 43 (or T8), AS5, and AS11. These bacteriocin genes were strongly associated with antibiotic resistance, particularly vancomycin and ampicillin, and Inc18 rep2_pRE25-derivative plasmids, classically associated with vancomycin resistance transposons. Such bacteriocin arsenal likely enhances the adaptability and competitive fitness of E. faecium in the nosocomial environment. By combining a novel tailored database, whole-genome sequencing, and epidemiological data, our work elucidates meaningful connections between bacteriocin determinants, antimicrobial resistance, mobile genetic elements, and ecological origins in E. faecium and provides a framework for elucidating bacteriocin landscapes in other organisms. Characterizing species- and strain-level differences in bacteriocin profiles may reveal determinants of ecological adaptation, and translating these discoveries could further inform strategies to exploit bacteriocins against high-risk clones. IMPORTANCE This work significantly expands the knowledge on the understudied bacteriocin diversity in opportunistic enterococci, revealing their contribution in the adaptation to different environments. It underscores the importance of placing increased emphasis on genetic platforms carrying bacteriocins as well as on cryptic plasmids that often exclusively harbor bacteriocins since bacteriocin production can significantly contribute to plasmid maintenance, potentially facilitating their stable transmission across generations. Further characterization of strain-level bacteriocin landscapes could inform strategies to combat high-risk clones. Overall, these insights provide a framework for unraveling the therapeutic and biotechnological potential of bacteriocins.
Collapse
Affiliation(s)
- Ana P. Tedim
- Group for Biomedical Research in Sepsis (BioSepsis), Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
- Grupo de Investigación Biomédica en Sepsis-BioSepsis, Hospital Universitario Río Hortega, Instituto de Investigación Biomédica de Salamanca (IBSAL), Valladollid, Spain
| | - Ana C. Almeida-Santos
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Val F. Lanza
- Department of Microbiology, Ramón y Cajal University Hospital and Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
| | - Carla Novais
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Teresa M. Coque
- Department of Microbiology, Ramón y Cajal University Hospital and Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- Network Research Centre for Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana R. Freitas
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
- One Health Toxicology Research Unit (1H-TOXRUN), University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
| | - Luísa Peixe
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - from the ESCMID Study Group on Food- and Water-borne Infections (EFWISG)
- Group for Biomedical Research in Sepsis (BioSepsis), Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
- Grupo de Investigación Biomédica en Sepsis-BioSepsis, Hospital Universitario Río Hortega, Instituto de Investigación Biomédica de Salamanca (IBSAL), Valladollid, Spain
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Department of Microbiology, Ramón y Cajal University Hospital and Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- Network Research Centre for Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- One Health Toxicology Research Unit (1H-TOXRUN), University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
| |
Collapse
|
10
|
Ashraf D, Shaaban MI, Hassan R, El-Aziz AMA. Polidocanol inhibits Enterococcus faecalis virulence factors by targeting fsr quorum sensing system. BMC Microbiol 2024; 24:411. [PMID: 39415105 PMCID: PMC11481293 DOI: 10.1186/s12866-024-03548-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 09/25/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND The wide spread of antimicrobial resistance in Enterococcus faecalis is a critical global concern, leading to increasingly limited treatment options. The fsr quorum sensing (QS) plays a critical role in the pathogenicity of E. faecalis, allowing bacteria to coordinate gene expression and regulate many virulence factors. Therefore, fsr QS of E. faecalis represents a potential therapeutic target that provides an effective strategy to treat antibiotic-resistant infections induced by E. faecalis. METHODS In this study, distribution of different virulence factors including, gelatinase, protease, cell surface hydrophobicity and biofilm formation in sixty clinical isolates of Enterococcus faecalis was investigated. Sixty-six compounds were tested for their activity against fsr QS. The minimal inhibitory concentration of the tested compounds was evaluated using the microbroth dilution method. The effect of sub-inhibitory concentrations of the tested compounds on fsr QS was investigated using the gelatinase assay method. Additionally, the effect of potential QS inhibitor on the virulence factors was estimated. Quantitative real-time PCR was used to investigate the effect of the potential inhibitor on fsr QS related genes (fsrB-fsrC) and (gelE-sprE) and virulence associated genes including, asa1 and epbA. RESULTS The assessment of polidocanol activity against the fsr QS system was demonstrated by studying its effect on gelatinase production in E. faecalis clinical isolates. Sub-lethal concentrations of polidocanol showed a significant reduction in gelatinase and protease production by 54% to 70% and 64% to 85%, respectively. Additionally, it significantly reduced biofilm formation (P < 0.01) and interrupted mature biofilm at concentrations of ½, 1 × and 2 × MIC. Furthermore, polidocanol significantly decreased cell surface hydrophobicity (P < 0.01). Polidocanol at ½ MIC showed a significant reduction in the expression of QS genes including fsrB, fsrC, gelE and sprE by 57% to 97% without affecting bacterial viability. Moreover, it reduced the expression of virulence associated genes (asa1 and epbA) (P < 0.01). CONCLUSION Polidocanol appears to be a promising option for treating of E. faecalis infections by targeting the fsr QS system and exhibiting anti-biofilm activity.
Collapse
Affiliation(s)
- Dina Ashraf
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Mona I Shaaban
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Ramadan Hassan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Abeer M Abd El-Aziz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
11
|
Oumeddour DZ, Al-Dalali S, Zhao L, Zhao L, Wang C. Recent advances on cyanidin-3-O-glucoside in preventing obesity-related metabolic disorders: A comprehensive review. Biochem Biophys Res Commun 2024; 729:150344. [PMID: 38976946 DOI: 10.1016/j.bbrc.2024.150344] [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: 05/16/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Anthocyanins, found in various pigmented plants as secondary metabolites, represent a class of dietary polyphenols known for their bioactive properties, demonstrating health-promoting effects against several chronic diseases. Among these, cyanidin-3-O-glucoside (C3G) is one of the most prevalent types of anthocyanins. Upon consumption, C3G undergoes phases I and II metabolism by oral epithelial cells, absorption in the gastric epithelium, and gut transformation (phase II & microbial metabolism), with limited amounts reaching the bloodstream. Obesity, characterized by excessive body fat accumulation, is a global health concern associated with heightened risks of disability, illness, and mortality. This comprehensive review delves into the biodegradation and absorption dynamics of C3G within the gastrointestinal tract. It meticulously examines the latest research findings, drawn from in vitro and in vivo models, presenting evidence underlining C3G's bioactivity. Notably, C3G has demonstrated significant efficacy in combating obesity, by regulating lipid metabolism, specifically decreasing lipid synthesis, increasing fatty acid oxidation, and reducing lipid accumulation. Additionally, C3G enhances energy homeostasis by boosting energy expenditure, promoting the activity of brown adipose tissue, and stimulating mitochondrial biogenesis. Furthermore, C3G shows potential in managing various prevalent obesity-related conditions. These include cardiovascular diseases (CVD) and hypertension through the suppression of reactive oxygen species (ROS) production, enhancement of endogenous antioxidant enzyme levels, and inhibition of the nuclear factor-kappa B (NF-κB) signaling pathway and by exercising its cardioprotective and vascular effects by decreasing pulmonary artery thickness and systolic pressure which enhances vascular relaxation and angiogenesis. Type 2 diabetes mellitus (T2DM) and insulin resistance (IR) are also managed by reducing gluconeogenesis via AMPK pathway activation, promoting autophagy, protecting pancreatic β-cells from oxidative stress and enhancing glucose-stimulated insulin secretion. Additionally, C3G improves insulin sensitivity by upregulating GLUT-1 and GLUT-4 expression and regulating the PI3K/Akt pathway. C3G exhibits anti-inflammatory properties by inhibiting the NF-κB pathway, reducing pro-inflammatory cytokines, and shifting macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. C3G demonstrates antioxidative effects by enhancing the expression of antioxidant enzymes, reducing ROS production, and activating the Nrf2/AMPK signaling pathway. Moreover, these mechanisms also contribute to attenuating inflammatory bowel disease and regulating gut microbiota by decreasing Firmicutes and increasing Bacteroidetes abundance, restoring colon length, and reducing levels of inflammatory cytokines. The therapeutic potential of C3G extends beyond metabolic disorders; it has also been found effective in managing specific cancer types and neurodegenerative disorders. The findings of this research can provide an important reference for future investigations that seek to improve human health through the use of naturally occurring bioactive compounds.
Collapse
Affiliation(s)
- Dounya Zad Oumeddour
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, 100048, China; Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing, 100048, China.
| | - Sam Al-Dalali
- School of Food and Health, Guilin Tourism University, Guilin, 541006, China; Department of Food Science and Technology, Faculty of Agriculture and Food Science, Ibb University, Ibb, 70270, Yemen.
| | - Liang Zhao
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, 100048, China; Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing, 100048, China.
| | - Lei Zhao
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, 100048, China; Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing, 100048, China.
| | - Chengtao Wang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, 100048, China; Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing, 100048, China.
| |
Collapse
|
12
|
王 宏, 张 明, 林 瑶, 刘 杨, 薛 冠, 石 琳, 袁 静, 李 晓. [Characteristics of intestinal microbiota in the acute phase of Kawasaki disease in infants and children]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:1101-1107. [PMID: 39467681 PMCID: PMC11527410 DOI: 10.7499/j.issn.1008-8830.2405111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/02/2024] [Indexed: 10/30/2024]
Abstract
OBJECTIVES To study the composition, abundance, and functional profiles of the intestinal microbiota in infants and young children with Kawasaki disease (KD) during the acute phase, and to explore the potential role of intestinal microbiota in the pathogenesis of KD. METHODS Six children aged 0-3 years with acute KD admitted to the Department of Cardiology, Children's Hospital Affiliated to Capital Institute of Pediatrics from July to October 2021 were prospectively included as the KD group. Six age- and sex-matched healthy children who underwent physical examinations at the hospital during the same period were selected as the healthy control group. Metagenomics sequencing was used to detect and compare the differences in the microflora structure and functional profiles of fecal samples between the two groups. RESULTS There were significant differences in the structural composition and diversity of intestinal microbiota between the two groups (P<0.05). Compared with the healthy control group, the abundance of Listeria_monocytogenes (family Listeriaceae and genus Listeria), Bifidobacterium_rousetti, Enterococcus_avium, and Enterococcus_hirae was significantly higher in the intestinal microbiota in the KD group (|LDA|>2.0, P<0.05). The steroid degradation and apoptosis pathways were significantly upregulated in the KD group compared with the healthy control group, while the Bacterial_secretion_system, Sulfur_metabolism, Butanoate_metabolism, Benzoate_degradation, β-alanine metabolism, and α-linolenic acid pathways were significantly downregulated (|LDA|>2, P<0.05). CONCLUSIONS There are significant differences in the structure and diversity of intestinal microbiota between children aged 0-3 years with acute KD and healthy children, suggesting that disturbances in intestinal microbiota occur during the acute phase of KD. In particular, Listeria_monocytogenes, Enterococcus_avium, and Enterococcus_hirae may be involved in the pathogenesis of KD through steroid degradation and apoptosis pathways.
Collapse
Affiliation(s)
| | | | | | | | - 冠华 薛
- 首都儿科研究所附属儿童医院,细菌研究室北京100020
| | | | - 静 袁
- 首都儿科研究所附属儿童医院,细菌研究室北京100020
| | | |
Collapse
|
13
|
Koul K, Jawanda IK, Soni T, Madaan K, Bhatt S, Singh P, Sharma D, Bhardwaj SB, Kumari S. Antibacterial and antibiofilm potential of Thuja orientalis L. extract targeting cariogenic Enterococcus faecalis ATCC 29212: A combined in-vitro, in-silico study, and cytotoxicity screening. Arch Oral Biol 2024; 171:106107. [PMID: 39647458 DOI: 10.1016/j.archoralbio.2024.106107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/10/2024]
Abstract
OBJECTIVES In this study, we explored the efficacy of methanolic extract of Thuja orientalis (TOME) as a novel antibacterial and antibiofilm agent against a cariogenic bacterium, Enterococcus faecalis ATCC 29212. DESIGN Antibacterial susceptibility studies were conducted and surface morphology analysis was performed using field emission scanning electron microscopy (FESEM). Antibiofilm activity was evaluated through both qualitative and quantitative biofilm inhibition assays and validated by microscopic analysis. In-silico molecular docking studies were conducted using the EDock server. The effectiveness of TOME was substantiated by biofilm model on dentin discs and cytotoxicity towards the HaCaT cell line was assessed using the MTT assay. RESULTS TOME exhibited significant bactericidal activity with minimum inhibitory concentration of 12.5 mg/mL and additionally, it effectively compromised bacterial cell wall integrity. Qualitative, quantitative and microscopic studies depicted the inhibition of biofilm formation. TOME significantly impacted the production of extracellular polymeric substance and extracellular DNA. Molecular docking studies identified beta-caryophyllene as a potent inhibitor of the Enterococcal surface protein (Esp). Biofilm model depicted the reduction of bacterial load on dentin discs. Additionally, TOME showed reduced cytotoxicity on HaCaT cells, indicating its potential as a safe therapeutic agent. CONCLUSION These findings highlight TOME's promise for developing novel treatments for dental infections and biofilm-associated diseases. Further research should focus on isolating and characterizing the active compounds within TOME, particularly beta-caryophyllene, to elucidate their precise mechanisms of action.
Collapse
Affiliation(s)
- Khyati Koul
- Department of Microbiology, Panjab University, Chandigarh 160014, India
| | | | - Thomson Soni
- Department of Microbiology, Panjab University, Chandigarh 160014, India
| | - Kashish Madaan
- Department of Microbiology, Panjab University, Chandigarh 160014, India
| | - Sunidhi Bhatt
- Bioremediation and Metabolomics Research Group, Department of Environmental Sciences, Central University of Himachal Pradesh, Kangra 176206, India
| | - Pranjali Singh
- Department of Microbiology, Panjab University, Chandigarh 160014, India
| | - Divyani Sharma
- Department of Microbiology, Panjab University, Chandigarh 160014, India
| | - Sonia Bhonchal Bhardwaj
- Department of Microbiology, Dr. Harvansh Singh Judge Institute of Dental Science and Hospital, Panjab University, Chandigarh 160014, India
| | - Seema Kumari
- Department of Microbiology, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
14
|
Lam LN, Sedra A, Kajfasz J, Berges A, Saengpet IS, Adams G, Fairman J, Lemos JA. Trivalent immunization with metal-binding proteins confers protection against enterococci in a mouse infection model. FEMS MICROBES 2024; 5:xtae031. [PMID: 39524556 PMCID: PMC11549557 DOI: 10.1093/femsmc/xtae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/13/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
Enterococcus faecalis is ranked among the top five bacterial pathogens responsible for catheter-associated urinary tract infections, wound infections, secondary root canal infections, and infective endocarditis. Previously, we showed that inactivation of either the manganese- and iron-binding (EfaA) or zinc-binding (AdcA and AdcAII) lipoproteins significantly reduced E. faecalis virulence. Here, we explored whether immunization using a multi-valent approach induces protective immunity against systemic enterococcal infections. We found that multi-antigen antisera raised against EfaA, AdcA, and AdcAII displayed similar capacities to initiate neutrophil-mediated opsonization, like their single-antigen counterparts. Further, these antigen-specific antibodies worked synergistically with calprotectin, a divalent host metal chelator, to inhibit the growth of E. faecalis in laboratory media as well as in human sera. Using the Galleria mellonella invertebrate model and mouse peritonitis model, we showed that passive immunization with multi-antigen antisera conferred robust protection against E. faecalis infection, while the protective effects of single antigen antisera were negligible in G. mellonella, and negligible-to-moderate in the mouse model. Lastly, active immunization with the 3-antigen (trivalent) cocktail significantly protected mice against either lethal or non-lethal E. faecalis infections, with this protection appearing to be far-reaching based on immunization results obtained with contemporary strains of E. faecalis and closely related Enterococcus faecium.
Collapse
Affiliation(s)
- Ling Ning Lam
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, United States
| | - Angie Sedra
- Vaxcyte, Inc., San Carlos, CA 94070, United States
| | - Jessica Kajfasz
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, United States
| | - Aym Berges
- Vaxcyte, Inc., San Carlos, CA 94070, United States
| | - Irene S Saengpet
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, United States
| | - Grace Adams
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, United States
| | | | - José A Lemos
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, United States
| |
Collapse
|
15
|
Aydın B, Alkuyruk SB, Tekman E, Yuca H, Karadayı M, Gülşahin Y, Çeçen Ö, Bona GE, Demirci B, Bona M, Karakaya S. Investigating the phytochemical profile, biological potentials, morphological, and anatomical characteristics of Cyclotrichium origanifolium (Labill.) Manden. & Scheng. (Lamiaceae) from Turkey. PROTOPLASMA 2024:10.1007/s00709-024-01992-5. [PMID: 39358643 DOI: 10.1007/s00709-024-01992-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/08/2024] [Indexed: 10/04/2024]
Abstract
Cyclotrichium origanifolium, a plant widely used in Eastern and Southern Anatolia for culinary purposes, was subject of this study, which aimed to comprehensively evaluate its potential therapeutic applications. This research stands out due to its holistic approach, combining morpho-anatomical studies, chemical, and biological analyses to explore antioxidant, antidiabetic, anticholinesterase, genotoxic, and anti-genotoxic effects of methanolic and aqueous extracts, as well as flowering aerial part essential oil. It is a perennial plant, typically ranging from 10 to 40 cm in height, with a suffrutescent and highly branched growth habit. Essential oils are produced within glandular trichomes. Oil, analyzed via GC-MS/MS, revealed 24 compounds accounting for 96.4% of oil, with isomenthone (52.4%), pulegone (23.4%), and β-pinene (9.5%) as predominant components. These findings are significant as they provide new insights into chemical composition of oils, particularly highlighting pharmacologically active compounds. Methanol extract exhibited superior antioxidant activity, correlated with high phenol and tannin content. Essential oil showed moderate inhibition of α-amylase (49.54%) and mild inhibition of acetylcholinesterase (11.84%) and butyrylcholinesterase (16.93%), suggesting potential in managing oxidative stress and neurodegenerative diseases. Study also conducted biosafety evaluations using Ames/Salmonella and Allium tests, essential for assessing genotoxic and antigenotoxic potential of natural products. Notably, significant antimicrobial effects were identified, particularly against Pseudomonas aeruginosa and Enterococcus faecalis. Comprehensive analysis and discovery of significant bioactivities position this research as a valuable contribution to field, distinguishing it from previous studies on similar species. This study provides a foundational understanding of morpho-anatomical, pharmacological, biological properties of plant, opening avenues for future research.
Collapse
Affiliation(s)
- Bilge Aydın
- Department of Pharmacognosy, Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Satuk Buğra Alkuyruk
- Department of Pharmaceutical Microbiology, Ankara University, Ankara, Türkiye
- Department of Pharmaceutical Microbiology, Ataturk University, Erzurum, Türkiye
| | - Enes Tekman
- Department of Pharmaceutical Botany, Ankara University, Ankara, Türkiye
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Ataturk University, Erzurum, Türkiye
| | - Hafize Yuca
- Department of Pharmacognosy, Ataturk University, Erzurum, Türkiye
| | | | - Yusuf Gülşahin
- Department of Biology, Ataturk University, Erzurum, Türkiye
| | - Ömer Çeçen
- Department of Plant and Animal Production, Karamanoglu Mehmetbey University, Karaman, Türkiye
| | - Gülnur Ekşi Bona
- Department of Pharmaceutical Botany, İstanbul Cerrahpasa University, Istanbul, Türkiye
| | - Betül Demirci
- Department of Pharmacognosy, Anadolu University, Eskişehir, Türkiye
| | - Mehmet Bona
- Department of Biology, İstanbul University, Istanbul, Türkiye
| | - Songül Karakaya
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Ataturk University, Erzurum, Türkiye.
| |
Collapse
|
16
|
Kim YH, Lee DH, Seo HS, Eun SH, Lee DS, Choi YK, Lee SH, Kim TY. Genome-based taxonomic identification and safety assessment of an Enterococcus strain isolated from a homemade dairy product. Int Microbiol 2024; 27:1513-1525. [PMID: 38466360 DOI: 10.1007/s10123-024-00496-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/24/2024] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
The aim of this study was to explore the taxonomic identification and evaluate the safety of a bacterium, Enterococcus lactis IDCC 2105, isolated from homemade cheese in Korea, using whole genome sequence (WGS) analysis. It sought to identify the species level of this Enterococcus spp., assess its antibiotic resistance, and evaluate its virulence potential. WGS analysis confirmed the bacterial strain IDCC 2105 as E. lactis and identified genes responsible for resistance to erythromycin and clindamycin, specifically msrC, and eatAv, which are chromosomally located, indicating a minimal risk for horizontal gene transfer. The absence of plasmids in E. lactis IDCC 2105 further diminishes the likelihood of resistance gene dissemination. Additionally, our investigation into seven virulence factors, including hemolysis, platelet aggregation, biofilm formation, hyaluronidase, gelatinase, ammonia production, and β-glucuronidase activity, revealed no detectable virulence traits. Although bioinformatic analysis suggested the presence of collagen adhesion genes acm and scm, these were not corroborated by phenotypic virulence assays. Based on these findings, E. lactis IDCC 2105 presents as a safe strain for potential applications, contributing valuable information on its taxonomy, antibiotic resistance profile, and lack of virulence factors, supporting its use in food products.
Collapse
Affiliation(s)
- Young-Hoo Kim
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, South Korea
| | | | - Han Sol Seo
- Yunovia Co., Ltd, Hwaseong, 18449, South Korea
| | | | - Do Sup Lee
- Yunovia Co., Ltd, Hwaseong, 18449, South Korea
| | | | - Sang Hyun Lee
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, South Korea
| | - Tae-Yoon Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, 13488, Republic of Korea.
| |
Collapse
|
17
|
Shahapur PR, Shahapur RP, Veggalam S, Kandi V. A Report of a Rare Case of Vagococcus fluvialis Isolated From Urine: Clinical Significance of Vagococcus Species With an Update of the Available Literature. Cureus 2024; 16:e71620. [PMID: 39553042 PMCID: PMC11566384 DOI: 10.7759/cureus.71620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024] Open
Abstract
Vagococcusspecies (spp.) are gram-positive cocci that are rarely reported in humans. These bacteria share physiological, cultural, and biochemical properties with Enterococcus spp. Recently, they have garnered attention as potential opportunistic pathogens capable of causing infections in individuals with predisposing factors and comorbidities. Human infections are sporadic, with only a few cases reported worldwide. A multidisciplinary microbiological approach is essential for the successful identification of these organisms. Antibiotic sensitivity is critical for effective treatment, especially considering that Vagococcus spp. not only were recently discovered but are also developing resistance to several antibiotics, as confirmed by the available literature, including the present case where the organism was found to be pan-drug resistant. Clinicians should be aware of these bacteria and consider them as emerging opportunistic pathogens. We report the case of a 56-year-old male with grade III hydronephrosis, urolithiasis, and a UTI, in whichVagococcus fluvialis was isolated from the patient's urine specimen.
Collapse
Affiliation(s)
- Praveen R Shahapur
- Microbiology, Shri B M Patil Medical College Hospital and Research Centre, Bijapur Lingayat District Educational (BLDE) (Deemed to be University), Vijayapura, IND
| | - Roopa P Shahapur
- Dentistry, Shri B M Patil Medical College Hospital and Research Centre, Bijapur Lingayat District Educational (BLDE) (Deemed to be University), Vijayapura, IND
| | - Shreya Veggalam
- Medicine, Prathima Institute of Medical Sciences, Karimnagar, IND
| | - Venkataramana Kandi
- Clinical Microbiology, Prathima Institute of Medical Sciences, Karimnagar, IND
| |
Collapse
|
18
|
Liyanarachi KV, Mohus RM, Rogne T, Gustad LT, Åsvold BO, Romundstad S, Solligård E, Hallan S, Damås JK. Chronic kidney disease and risk of bloodstream infections and sepsis: a 17-year follow-up of the population-based Trøndelag Health Study in Norway. Infection 2024; 52:1983-1993. [PMID: 38679665 PMCID: PMC11499395 DOI: 10.1007/s15010-024-02265-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/08/2024] [Indexed: 05/01/2024]
Abstract
PURPOSE Bloodstream infections (BSI) and sepsis are important causes of hospitalization, loss of health, and death globally. Targetable risk factors need to be identified to improve prevention and treatment. In this study, we aimed to evaluate the association of chronic kidney disease (CKD) and risk of and mortality from BSI and sepsis in the general population during a 22-year period. METHODS We conducted a prospective cohort study among participants in the population-based Norwegian HUNT Study, where 68,438 participated. The median follow-up time was 17.4 years. The exposures were estimated glomerular filtration rate (eGFR) and albumin-creatinine ratio (ACR) in urine. The outcomes were hazard ratios (HR) of hospital admission or death due to BSI or sepsis. The associations were adjusted for age, sex, diabetes, obesity, systolic blood pressure, smoking status, and cardiovascular disease. RESULTS Participants with eGFR < 30 ml/min/1.732 had HR 3.35 for BSI (95% confidence intervals (CI) 2.12-5.3) and HR 2.94 for sepsis (95% CI 1.82-4.8) compared to normal eGFR (≥ 90 ml/min/1.732). HRs of death from BSI and sepsis were 4.2 (95% CI 1.71-10.4) and 4.1 (95% CI 1.88-8.9), respectively. Participants with severely increased albuminuria (ACR > 30 mg/mmol) had HR 3.60 for BSI (95% CI 2.30-5.6) and 3.14 for sepsis (95% CI 1.94-5.1) compared to normal albumin excretion (ACR < 3 mg/mmol). HRs of death were 2.67 (95% CI 0.82-8.7) and 2.16 (95% CI 0.78-6.0), respectively. CONCLUSION In this large population-based cohort study, CKD was clearly associated with an increased risk of BSI and sepsis and related death.
Collapse
Affiliation(s)
- Kristin Vardheim Liyanarachi
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.
- Department of Infectious Diseases, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| | - Randi Marie Mohus
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Anaesthesia and Intensive Care, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tormod Rogne
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Yale Department of Chronic Disease Epidemiology and Center for Perinatal, Pediatric and Environmental Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Lise Tuset Gustad
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Faculty of Nursing and Health Sciences, Nord University, Levanger, Norway
- Department of Internal Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Bjørn Olav Åsvold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Center, NTNU, Norwegian University of Science and Technology, Levanger, Norway
| | - Solfrid Romundstad
- Department of Internal Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Erik Solligård
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Helse Møre Og Romsdal Hospital Trust, Ålesund, Norway
| | - Stein Hallan
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Nephrology, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Jan Kristian Damås
- Mid-Norway Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Infectious Diseases, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
19
|
Archambaud C, Nunez N, da Silva RAG, Kline KA, Serror P. Enterococcus faecalis: an overlooked cell invader. Microbiol Mol Biol Rev 2024; 88:e0006924. [PMID: 39239986 PMCID: PMC11426025 DOI: 10.1128/mmbr.00069-24] [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] [Indexed: 09/07/2024] Open
Abstract
SUMMARYEnterococcus faecalis and Enterococcus faecium are human pathobionts that exhibit a dual lifestyle as commensal and pathogenic bacteria. The pathogenic lifestyle is associated with specific conditions involving host susceptibility and intestinal overgrowth or the use of a medical device. Although the virulence of E. faecium appears to benefit from its antimicrobial resistance, E. faecalis is recognized for its higher pathogenic potential. E. faecalis has long been considered a predominantly extracellular pathogen; it adheres to and is taken up by a wide range of mammalian cells, albeit with less efficiency than classical intracellular enteropathogens. Carbohydrate structures, rather than proteinaceous moieties, are likely to be primarily involved in the adhesion of E. faecalis to epithelial cells. Consistently, few adhesins have been implicated in the adhesion of E. faecalis to epithelial cells. On the host side, very little is known about cognate receptors, except for the role of glycosaminoglycans during macrophage infection. Several lines of evidence indicate that E. faecalis internalization may involve a zipper-like mechanism as well as a macropinocytosis pathway. Conversely, E. faecalis can use several strategies to prevent engulfment in phagocytes. However, the bacterial and host mechanisms underlying cell infection by E. faecalis are still in their infancy. The most recent striking finding is the existence of an intracellular lifestyle where E. faecalis can replicate within a variety of host cells. In this review, we summarize and discuss the current knowledge of E. faecalis-host cell interactions and argue on the need for further mechanistic studies to prevent or reduce infections.
Collapse
Affiliation(s)
- Cristel Archambaud
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Natalia Nunez
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Ronni A G da Silva
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Drug Resistance Interdisciplinary Research Group, Singapore, Singapore
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Kimberly A Kline
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Pascale Serror
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| |
Collapse
|
20
|
Szczuka E, Rolnicka D, Wesołowska M. Cytotoxic Activity of Vancomycin-Resistant Enterococci Isolated from Hospitalised Patients. Pathogens 2024; 13:827. [PMID: 39452699 PMCID: PMC11509928 DOI: 10.3390/pathogens13100827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024] Open
Abstract
Vancomycin-resistant enterococci (VRE) are considered one of the main nosocomial pathogens due to their increasing antibiotic resistance and ability to cause life-threatening infections in humans. This study included VRE isolates obtained from various specimens including urine, blood, faeces, wounds, sputum, and oral cavity wash. Of the 37 strains, 30 (81.1%) and 7 (18.9%) were identified by MALDI TOF as Enterococcus faecium and Enterococcus faecalis, respectively. The clinical vancomycin-resistant enterococci exhibited multi-drug resistance (MDR). Apart from vancomycin, the enterococci exhibited resistance to penicillins (89.1 to 100%), fluoroquinolones (100%), rifampicin (86.5%), tetracycline (27%), aminoglycosides (56.8 to 86.5%), quinupristin-dalfopristin (35.1%), and chloramphenicol (10.8%). Moreover, resistance to linezolid and tigecycline emerged among the tested vancomycin-resistant enterococci. The analysis of aminoglycoside modifying enzyme (AME) genes showed the presence of bifunctional aac(6')-Ie-aph(2″)-Ia genes contributed to high-level aminoglycoside resistance (HLAR) in the E. faecalis and E. faecium isolates. The other AME gene, i.e., aph(3')-IIIa, was also found in the VRE isolates. All strains carried the vanA gene. Enterococci from colonised gastrointestinal tracts (1/2.7%) and from infection (6/16.2%) showed cytotoxic activity against the human epithelial cell line HEp-2.
Collapse
Affiliation(s)
- Ewa Szczuka
- Department of Microbiology, Faculty of Biology, Institute of Experimental Biology, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland; (D.R.); (M.W.)
| | - Dominika Rolnicka
- Department of Microbiology, Faculty of Biology, Institute of Experimental Biology, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland; (D.R.); (M.W.)
| | - Maria Wesołowska
- Department of Microbiology, Faculty of Biology, Institute of Experimental Biology, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland; (D.R.); (M.W.)
- Microbiology Laboratory, University Clinical Hospital in Poznań, ul. Przybyszewskiego 49, 60-355 Poznań, Poland
| |
Collapse
|
21
|
Guevara-Ramírez P, Cadena-Ullauri S, Paz-Cruz E, Ruiz-Pozo VA, Tamayo-Trujillo R, Cabrera-Andrade A, Zambrano AK. Gut Microbiota Disruption in Hematologic Cancer Therapy: Molecular Insights and Implications for Treatment Efficacy. Int J Mol Sci 2024; 25:10255. [PMID: 39408584 PMCID: PMC11476909 DOI: 10.3390/ijms251910255] [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: 07/29/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Hematologic malignancies (HMs), including leukemia, lymphoma, and multiple myeloma, involve the uncontrolled proliferation of abnormal blood cells, posing significant clinical challenges due to their heterogeneity and varied treatment responses. Despite recent advancements in therapies that have improved survival rates, particularly in chronic lymphocytic leukemia and acute lymphoblastic leukemia, treatments like chemotherapy and stem cell transplantation often disrupt gut microbiota, which can negatively impact treatment outcomes and increase infection risks. This review explores the complex, bidirectional interactions between gut microbiota and cancer treatments in patients with HMs. Gut microbiota can influence drug metabolism through mechanisms such as the production of enzymes like bacterial β-glucuronidases, which can alter drug efficacy and toxicity. Moreover, microbial metabolites like short-chain fatty acids can modulate the host immune response, enhancing treatment effectiveness. However, therapy often reduces the diversity of beneficial bacteria, such as Bifidobacterium and Faecalibacterium, while increasing pathogenic bacteria like Enterococcus and Escherichia coli. These findings highlight the critical need to preserve microbiota diversity during treatment. Future research should focus on personalized microbiome-based therapies, including probiotics, prebiotics, and fecal microbiota transplantation, to improve outcomes and quality of life for patients with hematologic malignancies.
Collapse
Affiliation(s)
- Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Santiago Cadena-Ullauri
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Elius Paz-Cruz
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Viviana A. Ruiz-Pozo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Rafael Tamayo-Trujillo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| | - Alejandro Cabrera-Andrade
- Escuela de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito 170124, Ecuador
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170124, Ecuador
| | - Ana Karina Zambrano
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
| |
Collapse
|
22
|
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
|
23
|
Madani WAM, Ramos Y, Cubillos-Ruiz JR, Morales DK. Enterococcal-host interactions in the gastrointestinal tract and beyond. FEMS MICROBES 2024; 5:xtae027. [PMID: 39391373 PMCID: PMC11466040 DOI: 10.1093/femsmc/xtae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/05/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
The gastrointestinal tract (GIT) is typically considered the natural niche of enterococci. However, these bacteria also inhabit extraintestinal tissues, where they can disrupt organ physiology and cause life-threatening infections. Here, we discuss how enterococci, primarily Enterococcus faecalis, interact with the intestine and other host anatomical locations such as the oral cavity, heart, liver, kidney, and vaginal tract. The metabolic flexibility of these bacteria allows them to quickly adapt to new environments, promoting their persistence in diverse tissues. In transitioning from commensals to pathogens, enterococci must overcome harsh conditions such as nutrient competition, exposure to antimicrobials, and immune pressure. Therefore, enterococci have evolved multiple mechanisms to adhere, colonize, persist, and endure these challenges in the host. This review provides a comprehensive overview of how enterococci interact with diverse host cells and tissues across multiple organ systems, highlighting the key molecular pathways that mediate enterococcal adaptation, persistence, and pathogenic behavior.
Collapse
Affiliation(s)
- Wiam Abdalla Mo Madani
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, NY 10065, United States
| | - Yusibeska Ramos
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, NY 10065, United States
| | - Juan R Cubillos-Ruiz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, NY 10065, United States
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, NY 10065, United States
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, NY 10065, United States
| | - Diana K Morales
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, NY 10065, United States
| |
Collapse
|
24
|
Seong YJ, Song JE, Lee E, Kim EJ, Heo JY, Choi YH, Kim YC. Clinical outcome of ampicillin or ampicillin/sulbactam versus glycopeptides in ampicillin-susceptible Enterococcus faecalis/faecium bacteremia: a 10-year retrospective cohort study. BMC Infect Dis 2024; 24:906. [PMID: 39223521 PMCID: PMC11368023 DOI: 10.1186/s12879-024-09824-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Glycopeptides for ampicillin-susceptible Enterococcus faecalis/faecium bacteremia are readily prescribed depending on the severity of the condition. However, there is limited data on the outcomes of glycopeptide use compared to ampicillin-containing regimens for ampicillin-susceptible E. faecalis/faecium bacteremia. From an antibiotic stewardship perspective, it is important to determine whether the use of glycopeptides is associated with improved clinical outcomes in patients with ampicillin-susceptible E. faecalis/faecium bacteremia. METHODS This retrospective cohort study was conducted at a university-affiliated hospital between January 2010 and September 2019. We collected data from patients with positive blood cultures for Enterococcus species isolates. The clinical data of patients who received ampicillin-containing regimens or glycopeptides as definitive therapy for ampicillin-susceptible E. faecalis/faecium bacteremia were reviewed. Multivariate logistic regression analysis was performed to identify risk factors for 28-day mortality. RESULTS Ampicillin-susceptible E. faecalis/faecium accounted for 41.2% (557/1,353) of enterococcal bacteremia cases during the study period. A total of 127 patients who received ampicillin-containing regimens (N = 56) or glycopeptides (N = 71) as definitive therapy were included in the analysis. The 28-day mortality rate was higher in patients treated with glycopeptides (19.7%) than in those treated with ampicillin-containing regimens (3.6%) (p = 0.006). However, in the multivariate model, antibiotic choice was not an independent predictor of 28-day mortality (adjusted OR, 3.7; 95% CI, 0.6-23.6). CONCLUSIONS Glycopeptide use was not associated with improved mortality in patients with ampicillin-susceptible E. faecalis/faecium bacteremia. This study provides insights to reduce the inappropriate use of glycopeptides in ampicillin-susceptible E. faecalis/faecium bacteremia treatment and promote antimicrobial stewardship.
Collapse
Affiliation(s)
- Yeol Jung Seong
- Department of Internal Medicine, Busan Medical Center, Busan, Korea
| | - Je Eun Song
- Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang, Korea
| | - Eugene Lee
- Department of Biology, Duke University, Durham, NC, USA
| | - Eun Jin Kim
- Department of Infectious Diseases, Ajou University School of Medicine, 164 World cup-ro, Yeongtong- gu, Suwon, 16499, Republic of Korea
| | - Jung Yeon Heo
- Department of Infectious Diseases, Ajou University School of Medicine, 164 World cup-ro, Yeongtong- gu, Suwon, 16499, Republic of Korea
| | - Young Hwa Choi
- Department of Infectious Diseases, Ajou University School of Medicine, 164 World cup-ro, Yeongtong- gu, Suwon, 16499, Republic of Korea.
| | - Yong Chan Kim
- Division of Infectious Diseases, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, 363 Dongbaekjukjeon-daero, Giheung-gu, Yongin-si, Gyeonggi-do, 16995, Republic of Korea.
| |
Collapse
|
25
|
Alrafaie AM, Pyrzanowska K, Smith EM, Partridge DG, Rafferty J, Mesnage S, Shepherd J, Stafford GP. A diverse set of Enterococcus-infecting phage provides insight into phage host-range determinants. Virus Res 2024; 347:199426. [PMID: 38960003 PMCID: PMC11269942 DOI: 10.1016/j.virusres.2024.199426] [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: 04/01/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024]
Abstract
Enterococci are robust Gram-positive bacteria that pose a significant threat in healthcare settings due to antibiotic resistance, with vancomycin-resistant enterococci (VRE) most prominent. To tackle this issue, bacteriophages (bacterial viruses) can be exploited as they specifically and efficiently target bacteria. Here, we successfully isolated and characterised a set of novel phages: SHEF10, SHEF11, SHEF13, SHEF14, and SHEF16 which target E. faecalis (SHEF10,11,13), or E. faecium (SHEF13, SHEF14 & SHEF16) strains including a range of clinical and VRE isolates. Genomic analysis shows that all phages are strictly lytic and diverse in terms of genome size and content, quickly and effectively lysing strains at different multiplicity of infections. Detailed analysis of the broad host-range SHEF13 phage revealed the crucial role of the enterococcal polysaccharide antigen (EPA) variable region in its infection of E. faecalis V583. In parallel, the discovery of a carbohydrate-targeting domain (CBM22) found conserved within the three phage genomes indicates a role in cell surface interactions that may be important in phage-bacterial interactons. These findings advance our comprehension of phage-host interactions and pave the way for targeted therapeutic strategies against antibiotic-resistant enterococcal infections.
Collapse
Affiliation(s)
- Alhassan M Alrafaie
- Department of Medical Laboratory, College of Applied Medical Sciences in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Karolina Pyrzanowska
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom, S10 2TA, UK
| | - Elspeth M Smith
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom, S10 2TA, UK
| | - David G Partridge
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield S10 2JF, UK
| | - John Rafferty
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
| | - Stephane Mesnage
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
| | - Joanna Shepherd
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom, S10 2TA, UK
| | - Graham P Stafford
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom, S10 2TA, UK; School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK.
| |
Collapse
|
26
|
Shaker AA, Samir A, Zaher HM, Abdel-Moein KA. Emergence of Virulent Extensively Drug-Resistant Vancomycin-Resistant Enterococci Among Diarrheic Pet Animals: A Possible Public Health Threat on the Move. Vector Borne Zoonotic Dis 2024; 24:600-606. [PMID: 38800841 DOI: 10.1089/vbz.2023.0167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024] Open
Abstract
Background: Vancomycin-resistant enterococci (VRE) have become an increasing public health concern in the past few decades, being associated with serious multidrug-resistant (MDR) infections. This study was conducted to investigate the role of diarrheic pet animals as potential reservoirs for virulent extensively drug-resistant (XDR) VRE and their threat on human health. Materials and Methods: Rectal swabs were collected from 153 diarrheic pet animals (80 dogs and 73 cats). The collected swabs were cultured on CHROMagarTMVRE for the isolation of vancomycin-resistant Enterococcus faecalis and Enterococcus faecium, and then suspected colonies were identified as enterococci after Gram staining, conventional biochemical tests, and molecular techniques. VRE were basically identified using the disk diffusion method; however, molecular identification of vanA and vanB genes was carried out among confirmed VRE isolates. Moreover, three virulence genes (cytolysin A, cylA; enterococcal surface protein, esp; and hyaluronidase, hyl) were investigated in VRE isolates. Thereafter, VRE strains that harbored virulence genes were tested for antimicrobial susceptibility. Results: Eighteen out of 153 animals (11.8%) were positive for VRE, which were obtained from 15% and 8.2% of the examined dogs and cats, respectively. None of the obtained isolates carried the vanA gene, whereas the vanB gene was detected in E. faecalis (4/10) with a prevalence rate (40%). Of the obtained VRE isolates, five possessed esp and/or cylA, while all strains were negative for the hyl gene. Furthermore, four virulent VRE isolates exhibited an XDR pattern, and one isolate was MDR. Conclusion: Diarrheic pet animals could represent a potential zoonotic reservoir for virulent XDR vancomycin-resistant E. faecalis, which may have serious public health implications.
Collapse
Affiliation(s)
- Alaa A Shaker
- Department of Zoonoses, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Ahmed Samir
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Hala M Zaher
- Department of Zoonoses, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Khaled A Abdel-Moein
- Department of Zoonoses, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
27
|
Santiago GS, Dropa M, Martone-Rocha S, Dos Santos TP, de Moura Gomes VT, Barbosa MRF, Razzolini MTP. Antimicrobial resistance characterization of Enterococcus faecium, Enterococcus faecalis and Enterococcus hirae isolated from marine coastal recreational waters in the State of São Paulo, Brazil. JOURNAL OF WATER AND HEALTH 2024; 22:1628-1640. [PMID: 39340376 DOI: 10.2166/wh.2024.098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/31/2024] [Indexed: 09/30/2024]
Abstract
Coastal water quality is facing increasing threats due to human activities. Their contamination by sewage discharges poses significant risks to the environment and public health. We aimed to investigate the presence of antibiotic-resistant Enterococcus in beach waters. Over a 10-month period, samples were collected from four beaches in the State of São Paulo (Brazil). Enterococcus isolates underwent matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF/MS) and molecular analysis for accurate genus and species identification. The antimicrobial susceptibility for 14 antibiotics was evaluated using the disc diffusion method followed by a multidrug-resistance (MDR) classification. PCR amplification method was used to detect antimicrobial resistance genes (ARGs). Our findings revealed the prevalence of Enterococcus faecalis, E. faecium and E. hirae. Out of 130 isolates, 118 were resistant to multiple antibiotics. The detection of resistance genes provided evidence of the potential transfer of antibiotic resistance within the environment. Our findings underscore the necessity for continuous research and surveillance to enhance understanding of the pathogenicity and antimicrobial resistance mechanisms of Enterococcus, which is crucial to implement effective measures to preserve the integrity of coastal ecosystems.
Collapse
Affiliation(s)
- Giovanna Silva Santiago
- School of Public Health, Environmental Health Department, University of São Paulo, Av. Dr Arnaldo 715, São Paulo, SP 01246-904, Brazil
| | - Milena Dropa
- School of Public Health, Environmental Health Department, University of São Paulo, Av. Dr Arnaldo 715, São Paulo, SP 01246-904, Brazil
| | - Solange Martone-Rocha
- School of Public Health, Environmental Health Department, University of São Paulo, Av. Dr Arnaldo 715, São Paulo, SP 01246-904, Brazil
| | - Talita Pereira Dos Santos
- School of Public Health, Environmental Health Department, University of São Paulo, Av. Dr Arnaldo 715, São Paulo, SP 01246-904, Brazil
| | - Vasco Tulio de Moura Gomes
- CETESB - Environmental Company of São Paulo State, Av. Professor Frederico Hermann Jr, 345, São Paulo, SP 05459-900, Brazil
| | - Mikaela Renata Funada Barbosa
- CETESB - Environmental Company of São Paulo State, Av. Professor Frederico Hermann Jr, 345, São Paulo, SP 05459-900, Brazil; Environmental Health Department, NARA - Center for Research in Environmental Risk Assessment, Av. Dr Arnaldo 715, São Paulo, SP 01246-904, Brazil
| | - Maria Tereza Pepe Razzolini
- School of Public Health, Environmental Health Department, University of São Paulo, Av. Dr Arnaldo 715, São Paulo, SP 01246-904, Brazil; Environmental Health Department, NARA - Center for Research in Environmental Risk Assessment, Av. Dr Arnaldo 715, São Paulo, SP 01246-904, Brazil E-mail:
| |
Collapse
|
28
|
Szymanski M, Skiba MM, Piasecka M, Olender A. A rare case of invasive Enterococcus cecorum infection and related diagnostic difficulties. Clin Case Rep 2024; 12:e9386. [PMID: 39210929 PMCID: PMC11358030 DOI: 10.1002/ccr3.9386] [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: 05/23/2024] [Revised: 08/03/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Key Clinical Message This report presents a rare case of invasive infection caused by Enterococcus cecorum. There are no specific guidelines regarding antibiotic therapy for this infection. Based on this case, it can be concluded that linezolid demonstrates in vivo activity against Enterococcus cecorum and can be successfully used in therapy. Abstract Enterococcus cecorum is an extremely rare pathogen in humans. Since 1984, when the microorganism was first described, only a dozen cases of invasive infections in humans have been reported in the literature. The diagnostic pathway may involve difficulties in correctly identifying this microorganism. Based on the case described, it can be thought that Enterococcus cecorum is a more challenging bacterium than the much more common Enterococcus faecium or Enterococcus faecalis. The described case underscores the importance of medical vigilance in clinical practice. It seems that due to increasingly advanced techniques in molecular biology, we will more frequently detect pathogens that were previously encountered only sporadically. Since not every center has access to modern and advanced microbiological diagnostic methods, publications that practically combine classical microbiological diagnostic methods with those less accessible but more modern are exceptionally valuable. In the case described, it is also worth noting that classical methods still play a significant and crucial role in conducting microbiological diagnostics. In the era of rapid diagnostic tool development, it is important to emphasize the necessity of combining different methods rather than replacing one with another.
Collapse
Affiliation(s)
- Mateusz Szymanski
- Human Anatomy DepartmentMedical UniversityLublinPoland
- Intensive Care UnitStefan Cardinal Wyszyński District Specialist HospitalLublinPoland
| | - Małgorzata M. Skiba
- Intensive Care UnitStefan Cardinal Wyszyński District Specialist HospitalLublinPoland
| | - Małgorzata Piasecka
- Intensive Care UnitStefan Cardinal Wyszyński District Specialist HospitalLublinPoland
| | - Alina Olender
- Chair and Department of Medical MicrobiologyMedical UniversityLublinPoland
| |
Collapse
|
29
|
Koc F, Magner C, Murphy K, Kelleher ST, Tan MH, O'Toole M, Jenkins D, Boyle J, Lavelle M, Maguire N, Ross PR, Stanton C, McMahon CJ. Gut Microbiome in Children with Congenital Heart Disease After Cardiopulmonary Bypass Surgery (GuMiBear Study). Pediatr Cardiol 2024:10.1007/s00246-024-03634-2. [PMID: 39174731 DOI: 10.1007/s00246-024-03634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024]
Abstract
The gut microbiome of infants with congenital heart disease (CHD) undergoing cardiopulmonary bypass surgery (CPB) is at risk of profound alteration. The aim of this study was to examine the gut microbiome pre- and post-bypass surgery to explore potential implications of altered gut biodiversity. A prospective cohort study involving infants with CHD who underwent CPB was performed. Faecal samples were collected from infants alongside the collection of demographic and clinical data in order to examine gut microbiome changes before and after surgery. 16S rRNA sequencing analysis was performed on DNA isolated from stool samples to determine changes in gut microbiome composition. Thirty-three patients were recruited, with samples from thirteen of these available for final analysis. Compared with healthy, matched controls, at a genus level, pre-operative samples for infants with CHD demonstrated a higher relative abundance of Escherichia-Shigella (31% vs 2-6%) and a lower relative abundance of Bifidobacterium (13% vs 40-60%). In post-operative samples, the relative abundance of Escherichia-Shigella (35%), Enterococcus (11%), Akkermansia (6%), and Staphylococcus (5%) were higher than pre-op samples. One infant developed post-operative necrotising-enterocolitis (NEC). They displayed a marked abundance of the Enterococcus (93%) genus pre-operatively. This study demonstrates that infants with CHD have an altered gut microbiome when compared with healthy controls and there might be a possible link between an abundance of virulent species and NEC.
Collapse
Affiliation(s)
- Fatma Koc
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Claire Magner
- School of Nursing, Midwifery and Health Systems, University College Dublin, Dublin, Ireland
| | - Kiera Murphy
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Ireland
| | - Sean T Kelleher
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Mong H Tan
- Paediatric Intensive Care Unit, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Molly O'Toole
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Dominic Jenkins
- Laboratory, Children's Health Ireland at Crumlin, Crumlin, Ireland
| | - Jordan Boyle
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Marie Lavelle
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Niamh Maguire
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland
| | - Paul R Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Colin J McMahon
- Department Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin 12, Ireland.
- School of Medicine, University College Dublin, Dublin, Ireland.
- School of Health Professions Education (SHE), Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
30
|
Cao X, Li W, Xu Z, Li G, Wen Z, Meng Q, Li P, Yu Z, Chen Z, Zheng J. Loratadine Derivative Lo-7: A Weapon against Drug-Resistant Enterococcus and Streptococcal Infections. ACS Infect Dis 2024; 10:2961-2977. [PMID: 39066703 DOI: 10.1021/acsinfecdis.4c00293] [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: 07/30/2024]
Abstract
The primary obstacles in the management of Enterococcus and Streptococcal infections are drug resistance and biofilm formation. Our study revealed that loratadine at a concentration of ≥25 μM exhibited significant inhibitory effects on biofilm formation in 167 clinical strains of Enterococcus faecalis and 15 clinical isolates of Streptococcus agalactiae, Streptococcus pyogenes, and Streptococcus pneumoniae. Additionally, the antibiofilm activity against E. faecalis and Streptococcal was demonstrated by several loratadine derivatives with altered side-chain carbamate moieties. This study investigated the antibacterial activity of the loratadine derivative Lo-7 against clinical strains of S. agalactiae and S. pyogenes, with minimum inhibitory concentrations ranging from 12.5 to 25 μM. The findings revealed that a low concentration of loratadine derivative Lo-7 (3.125 μM) significantly augmented the bactericidal efficacy of vancomycin against multidrug-resistant (MDR) S. agalactiae, both in vitro and in vivo. The loratadine derivative Lo-7, even at low concentrations, demonstrated significant efficacy in eliminating intracellular MDR S. agalactiae within macrophages, potentially indicating a unique advantage over vancomycin, linezolid, and loratadine. Mechanistically, exposure to the loratadine derivative Lo-7 resulted in membrane depolarization without affecting membrane permeability in S. agalactiae. The potential targeting of the SecG subunit of the SecYEG membrane-embedded channel by the loratadine derivative Lo-7 in S. agalactiae was identified through quantitative proteomics, a drug affinity responsive target stability assay, and molecular docking.
Collapse
Affiliation(s)
- Xinyi Cao
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University Medical School, Shenzhen 518052, China
- Department of Microbiology, The First Affiliated Hospital of Jiamusi University, Jiamusi 154003, China
| | - Wei Li
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Zhichao Xu
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Guiqiu Li
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Zewen Wen
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Qingyin Meng
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Peiyu Li
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Zhijian Yu
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Zhong Chen
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| | - Jinxin Zheng
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University Medical School, Shenzhen 518052, China
| |
Collapse
|
31
|
Gao W, Liu X, Zhang S, Wang J, Qiu B, Shao J, Huang W, Huang Y, Yao M, Tang LL. Alterations in gut microbiota and inflammatory cytokines after administration of antibiotics in mice. Microbiol Spectr 2024; 12:e0309523. [PMID: 38899904 PMCID: PMC11302321 DOI: 10.1128/spectrum.03095-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/13/2024] [Indexed: 06/21/2024] Open
Abstract
Antibiotics are widely used to treat bacterial infection and reduce the mortality rate, while antibiotic overuse can cause gut microbiota dysbiosis. The impact of antibiotics on gut microbiota is not fully understood. In our study, four commonly used antibiotics (ceftazidime, cefoperazone-sulbactam, imipenem-cilastatin, and moxifloxacin) were given subcutaneously to mice, and their impacts on the gut microbiota composition and serum cytokine levels were evaluated through 16S rRNA analysis and a multiplex immunoassay. Antibiotic treatment markedly reduced gut microbiota diversity and changed gut microbiota composition. Antibiotic treatment significantly increased and decreased the abundance of Firmicutes and Bacteroidota, respectively. The antibiotic treatments increased the abundance of opportunistic pathogens such as Enterococcus and decreased that of Lachnospiraceae and Muribaculaceae. For moxifloxacin, the significantly high abundance of Enterococcus and Klebsiella was observed after 14 and 21 days of treatment. However, a relatively low abundance of opportunistic pathogens was found after 14 days of imipenem-cilastatin treatment. Additionally, the serum levels of various pro-inflammatory cytokines, such as IL-1β, IL-12 (p70), and IL-17, significantly increased after 21 days of antibiotic treatments. Overall, these results provide a guide for rational use of antibiotics in clinical settings: short-term use of moxifloxacin is recommended with regard to gut microbiota health, and the 14-day use of imipenem-cilastatin may have a less severe impact than other antibiotics.IMPORTANCEAntibiotic treatments are directly associated with changes in gut microbiota and are effective against both pathogens and beneficial bacteria. Gut microbiota dysbiosis induced by antibiotic treatment could increase the risk of some diseases. Therefore, an adequate understanding of gut microbiota changes after antibiotic use is crucial. In this study, we investigated the effects of continuous treatment with antibiotics on gut microbiota, serum cytokines, and intestinal inflammatory response. Our results suggest that short-term use of moxifloxacin is recommended, and the 14-day use of imipenem-cilastatin may have a less severe effect on gut microbiota health than cefoperazone-sulbactam. These results provide useful guidance on the rational use of antibiotics with regard to gut microbiota health.
Collapse
Affiliation(s)
- Wang Gao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xingyu Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuobo Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingxia Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junhua Shao
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University, Shulan International Medical College, Hangzhou, China
| | - Weixin Huang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Shaoxing Tongchuang Biotechnology Co., Ltd, Shaoxing, China
| | - Yilun Huang
- Alberta Institute, Wenzhou Medical University, Wenzhou, China
| | - Mingfei Yao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ling-Ling Tang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University, Shulan International Medical College, Hangzhou, China
| |
Collapse
|
32
|
Maier L, Stein-Thoeringer C, Ley RE, Brötz-Oesterhelt H, Link H, Ziemert N, Wagner S, Peschel A. Integrating research on bacterial pathogens and commensals to fight infections-an ecological perspective. THE LANCET. MICROBE 2024; 5:100843. [PMID: 38608681 DOI: 10.1016/s2666-5247(24)00049-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 04/14/2024]
Abstract
The incidence of antibiotic-resistant bacterial infections is increasing, and development of new antibiotics has been deprioritised by the pharmaceutical industry. Interdisciplinary research approaches, based on the ecological principles of bacterial fitness, competition, and transmission, could open new avenues to combat antibiotic-resistant infections. Many facultative bacterial pathogens use human mucosal surfaces as their major reservoirs and induce infectious diseases to aid their lateral transmission to new host organisms under some pathological states of the microbiome and host. Beneficial bacterial commensals can outcompete specific pathogens, thereby lowering the capacity of the pathogens to spread and cause serious infections. Despite the clinical relevance, however, the understanding of commensal-pathogen interactions in their natural habitats remains poor. In this Personal View, we highlight directions to intensify research on the interactions between bacterial pathogens and commensals in the context of human microbiomes and host biology that can lead to the development of innovative and sustainable ways of preventing and treating infectious diseases.
Collapse
Affiliation(s)
- Lisa Maier
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany
| | - Christoph Stein-Thoeringer
- Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany; Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Ruth E Ley
- Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; Max Planck Institute for Biology, Tübingen, Germany
| | - Heike Brötz-Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany
| | - Hannes Link
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany
| | - Nadine Ziemert
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany
| | - Samuel Wagner
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany.
| |
Collapse
|
33
|
Andriani L, Rega M, Bonilauri P, Pupillo G, De Lorenzi G, Bonardi S, Conter M, Bacci C. Vancomycin resistance and virulence genes evaluation in Enterococci isolated from pork and wild boar meat. Heliyon 2024; 10:e34543. [PMID: 39104496 PMCID: PMC11298925 DOI: 10.1016/j.heliyon.2024.e34543] [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: 04/22/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 08/07/2024] Open
Abstract
Enterococci are considered valuable sentinel Gram-positive bacteria for monitoring vancomycin antibiotic resistance due to their widespread presence and characteristics. The use of antimicrobials in farming animals has a role in the increasing of Antimicrobial Resistance (AMR) and the anthropogenic transformation of the landscape has forced wildlife into greater contact with humans and their livestock. The transmission of resistant bacteria by their meat products is a significant contributor to AMR development. The present study aimed to assess the prevalence of vancomycin resistant Enterococci spp. In antimicrobial-treated farmed pigs meat and in antimicrobial-free wild boars meat. A total of 341 Enterococci were isolated from 598 pork meat samples (57 %) and 173 Enterococci were isolated from 404 wild boar meat samples (42.8 %). Data found showed that low-resistance was detected more in wild boars meat Enterococci (52.6 %) than in pork meat once (48.4 %). However, the prevalence of resistance genes was at low level (33.9 % in pork meat Enterococci and 4.4 % in wild boar meat ones) and the only gene found was vanC1/C2, related to intrinsic AMR. Normally, Enterococci persist in the normal intestinal flora of animals including humans. However, the presence of resistance genes was frequently linked to the detection of pathogenic genes, mostly gelE in pork meat isolates and asa1 in wild boars meat isolates. Pathogenic bacteria can cause severe infections in human that can become more risky if associated to the presence of AMR. Pathogenic bacteria were characterized and a high presence of E. gallinarum and E. casseliflavus was found. Given the growing interest in wild game meat consumption the monitoring of AMR in these matrices is essential. Further surveillance studies are needed to fully evaluate the emergence and spread of vancomycin-resistant Enterococci (VRE) and pathogenic Enterococci from animal-derived food to humans, including the role of wildlife in this phenomenon. Giving the higher interest in wild animals meat consumption, it is important to better evaluate the spread of AMR phenomenon in the future and intensify hygienic control of wild animals derived food.
Collapse
Affiliation(s)
- Laura Andriani
- Food Hygiene and Inspection Unit, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126, Parma, Italy
| | - Martina Rega
- Food Hygiene and Inspection Unit, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126, Parma, Italy
| | - Paolo Bonilauri
- Istituto Zooprofilattico Sperimentale della Lombardia ed Emilia Romagna, via Pitagora, 2, 42124, Reggio Emilia, Italy
| | - Giovanni Pupillo
- Istituto Zooprofilattico Sperimentale della Lombardia ed Emilia Romagna, via Pitagora, 2, 42124, Reggio Emilia, Italy
| | - Giorgia De Lorenzi
- Istituto Zooprofilattico Sperimentale della Lombardia ed Emilia Romagna, via Pitagora, 2, 42124, Reggio Emilia, Italy
| | - Silvia Bonardi
- Food Hygiene and Inspection Unit, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126, Parma, Italy
| | - Mauro Conter
- Food Hygiene and Inspection Unit, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126, Parma, Italy
| | - Cristina Bacci
- Food Hygiene and Inspection Unit, Veterinary Science Department, University of Parma, Strada del Taglio, 10, 43126, Parma, Italy
| |
Collapse
|
34
|
Vivenzio VM, Esposito D, Monti SM, De Simone G. Bacterial α-CAs: a biochemical and structural overview. Enzymes 2024; 55:31-63. [PMID: 39222995 DOI: 10.1016/bs.enz.2024.07.001] [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: 09/04/2024]
Abstract
Carbonic anhydrases belonging to the α-class are widely distributed in bacterial species. These enzymes have been isolated from bacteria with completely different characteristics including both Gram-negative and Gram-positive strains. α-CAs show a considerable similarity when comparing the biochemical, kinetic and structural features, with only small differences which reflect the diverse role these enzymes play in Nature. In this chapter, we provide a comprehensive overview on bacterial α-CA data, with a highlight to their potential biomedical and biotechnological applications.
Collapse
|
35
|
Costache C, Colosi I, Toc DA, Daian K, Damacus D, Botan A, Toc A, Pana AG, Panaitescu P, Neculicioiu V, Schiopu P, Iordache D, Butiuc-Keul A. CRISPR-Cas System, Antimicrobial Resistance, and Enterococcus Genus-A Complicated Relationship. Biomedicines 2024; 12:1625. [PMID: 39062198 PMCID: PMC11274382 DOI: 10.3390/biomedicines12071625] [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: 06/13/2024] [Revised: 07/07/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
(1) Background: The rise in antibiotic resistant bacteria poses a significant threat to public health worldwide, necessitating innovative solutions. This study explores the role of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) in the context of antibiotic resistance among different species from the Enterococcus genus. (2) Methods: The genomes of Enterococcus included in the study were analyzed using CRISPRCasFinder to distinguish between CRISPR-positive (level 4 CRISPR) and CRISPR-negative genomes. Antibiotic resistance genes were identified, and a comparative analysis explored potential associations between CRISPR presence and antibiotic resistance profiles in Enterococcus species. (3) Results: Out of ten antibiotic resistance genes found in Enterococcus species, only one, the efmA gene, showed a strong association with CRISPR-negative isolates, while the others did not significantly differ between CRISPR-positive and CRISPR-negative Enterococcus genomes. (4) Conclusion: These findings indicate that the efmA gene may be more prevalent in CRISPR-negative Enterococcus genomes, and they may contribute to a better understanding of the molecular mechanisms underlying the acquisition of antibiotic resistance genes in Enterococcus species.
Collapse
Affiliation(s)
- Carmen Costache
- Department of Microbiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania (I.C.)
- Cluj County Emergency Hospital, 400000 Cluj-Napoca, Romania
| | - Ioana Colosi
- Department of Microbiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania (I.C.)
| | - Dan-Alexandru Toc
- Department of Microbiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania (I.C.)
- Cluj County Emergency Hospital, 400000 Cluj-Napoca, Romania
| | - Karla Daian
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - David Damacus
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Alexandru Botan
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Adelina Toc
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Adrian Gabriel Pana
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Paul Panaitescu
- Department of Microbiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania (I.C.)
| | - Vlad Neculicioiu
- Department of Microbiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania (I.C.)
| | - Pavel Schiopu
- Department of Microbiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania (I.C.)
| | - Dumitrana Iordache
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babeş-Bolyai University, 400084 Cluj-Napoca, Romania
- Centre for Systems Biology, Biodiversity and Bioresources, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Anca Butiuc-Keul
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babeş-Bolyai University, 400084 Cluj-Napoca, Romania
- Centre for Systems Biology, Biodiversity and Bioresources, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| |
Collapse
|
36
|
Hanzelová Z, Dudriková E, Lovayová V, Výrostková J, Regecová I, Zigo F, Bartáková K. Occurrence of Enterococci in the Process of Artisanal Cheesemaking and Their Antimicrobial Resistance. Life (Basel) 2024; 14:890. [PMID: 39063643 PMCID: PMC11277685 DOI: 10.3390/life14070890] [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: 06/17/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Enterococci are a group of microorganisms that have a controversial position from some scientific points of view. The species of the greatest clinical importance are E. faecalis and E. faecium, which are common agents of nosocomial infections. However, enterococci also have important applications in the dairy industry, as they are used as non-starter lactic acid bacteria (NSLAB) in a variety of cheeses, especially artisanal cheeses. The aim of this study was to determine the presence of representatives from the Enterococcus genus using PCR and MALDI-TOF MS methods on samples of raw milk, processing environment swabs, and cheese from four different artisanal dairy plants in Slovakia. Among the 136 isolates of enterococci, 9 species of genus Enterococci (E. faecalis, E. faecium, E. durans, E. devriesi, E. hirae, E. italicus, E. casseliflavus, E. malodoratus, and E. gallinarum) were identified and were tested for their antimicrobial resistance (AMR) to 8 antibiotics (amoxicillin, penicillin, ampicillin, erythromycin, levofloxacin, vancomycin, rifampicin, and tetracycline); most of them were resistant to rifampicin (35.3%), ampicillin (22.8%), and tetracycline (19.9%). A PCR analysis of vanA (4.41%) and tetM (14.71%) revealed that antimicrobial resistance genes were present in not only phenotypic resistant isolates of enterococci but also susceptible isolates. The investigation of antimicrobial resistance in enterococci during the cheesemaking process can be a source of valuable information for public health in the concept of "One Health".
Collapse
Affiliation(s)
- Zuzana Hanzelová
- Department of Food Hygiene, Technology and Safety, The University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia; (Z.H.); (J.V.); (I.R.)
| | - Eva Dudriková
- Department of Food Hygiene, Technology and Safety, The University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia; (Z.H.); (J.V.); (I.R.)
| | - Viera Lovayová
- Department of Medical and Clinical Microbiology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 040 11 Kosice, Slovakia;
| | - Jana Výrostková
- Department of Food Hygiene, Technology and Safety, The University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia; (Z.H.); (J.V.); (I.R.)
| | - Ivana Regecová
- Department of Food Hygiene, Technology and Safety, The University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia; (Z.H.); (J.V.); (I.R.)
| | - František Zigo
- Department of Animal Nutrition and Husbandry, The University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia;
| | - Klára Bartáková
- Department of Animal Origin Food & Gastronomic Sciences, University of Veterinary Sciences Brno, 612 42 Brno, Czech Republic;
| |
Collapse
|
37
|
Wang M, Wu M, Han M, Niu X, Fan A, Zhu S, Tong Y. Mining the Biosynthetic Landscape of Lactic Acid Bacteria Unearths a New Family of RiPPs Assembled by a Novel Type of ThiF-like Adenylyltransferases. ACS OMEGA 2024; 9:30891-30903. [PMID: 39035879 PMCID: PMC11256085 DOI: 10.1021/acsomega.4c03760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024]
Abstract
Ribosomally synthesized and post-translationally modified peptides (RiPPs) are chemically diverse natural products of ribosomal origin. These peptides, which frequently act as signals or antimicrobials, are biosynthesized by conserved enzymatic machinery, making genome mining a powerful strategy for unearthing previously uncharacterized members of their class. Herein, we investigate the untapped biosynthetic potential of Lactobacillales (i.e., lactic acid bacteria), an order of Gram-positive bacteria closely associated with human life, including pathogenic species and industrially relevant fermenters of dairy products. Through genome mining methods, we systematically explored the distribution and diversity of ThiF-like adenylyltransferase-utilizing RiPP systems in lactic acid bacteria and identified a number of unprecedented biosynthetic gene clusters. In one of these clusters, we found a previously undescribed group of macrocyclic imide biosynthetic pathways containing multiple transporters that may be involved in a potential quorum sensing (QS) system. Through in vitro assays, we determined that one such adenylyltransferase specifically catalyzes the intracyclization of its precursor peptide through macrocyclic imide formation. Incubating the enzyme with various primary amines revealed that it could effectively amidate the C-terminus of the precursor peptide. This new transformation adds to the growing list of Nature's peptide macrocyclization strategies and expands the impressive catalytic repertoire of the adenylyltransferase family. The diverse RiPP systems identified herein represent a vast, unexploited landscape for the discovery of a novel class of natural products and QS systems.
Collapse
Affiliation(s)
- Mengjiao Wang
- College
of Life Science and Technology, Beijing
University of Chemical Technology, Beijing 100029, People’s Republic of China
| | - Mengyue Wu
- State
Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, People’s
Republic of China
| | - Meng Han
- MOE
Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Xiaogang Niu
- Beijing
Nuclear Magnetic Resonance Center, College of Chemistry and Molecular
Engineering, Peking University, Beijing 100871, People’s Republic of China
| | - Aili Fan
- State
Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, People’s
Republic of China
| | - Shaozhou Zhu
- National
Institutes for Food and Drug Control, Beijing 102629, People’s Republic of China
| | - Yigang Tong
- College
of Life Science and Technology, Beijing
University of Chemical Technology, Beijing 100029, People’s Republic of China
| |
Collapse
|
38
|
Pan P, Sun L, Shi X, Huang X, Yin Y, Pan B, Hu L, Shen Q. Analysis of molecular epidemiological characteristics and antimicrobial susceptibility of vancomycin-resistant and linezolid-resistant Enterococcus in China. BMC Med Genomics 2024; 17:174. [PMID: 38951840 PMCID: PMC11218351 DOI: 10.1186/s12920-024-01948-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/21/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND This study investigates the distribution and characteristics of linezolid and vancomycin susceptibilities among Enterococcus faecalis (E. faecalis) and Enterococcus faecium (E. faecium) and explores the underlying resistance mechanisms. METHODS A total of 2842 Enterococcus clinical isolates from patients were retrospectively collected, and their clinical data were further analyzed. The minimum inhibitory concentrations (MICs) of vancomycin and linezolid were validated by broth dilution method. The resistance genes optrA, cfr, vanA, vanB and vanM were investigated using polymerase chain reaction (PCR). Housekeeping genes and resistance genes were obtianed through whole-genome sequencing (WGS). RESULTS Of the 2842 Enterococcus isolates, 88.5% (2516) originated from urine, with E. faecium accounted for 60.1% of these. The vanA gene was identified in 27/28 vancomycin resistant Enterococcus (VRE) isolates, 4 of which carried both vanA and vanM genes. The remaining strain was vanM positive. The optrA gene was identified in all E. faecalis isolates among linezolid resistant Enterococcus (LRE). E. faecium showed a higher multiple antibiotic resistance index (MAR index) compared to E. faecalis. The multi-locus sequence typing (MLST) showed the sequence type of E. faecium mainly belongs to clonal complex (CC) 17, nearly E. faecalis isolates analyzed were differentiated into 7 characteristics of sequence types (STs), among which ST16 of CC16 were the major lineage. CONCLUSION Urine was the primary source of VRE and LRE isolates in this study. E. faecium showed higher levels of resistance compared to E. faecalis. OptrA gene was detected in 91.6% of LRE, which could explain linezolid resistance, and van genes were detected in all vancomycin resistant Enterococcus strains, while vanA was a key resistance mechanism in VRE identified in this study.
Collapse
Affiliation(s)
- Ping Pan
- Department of Clinical Laboratory, Hangzhou Women's Hospital (Hangzhou Matenal and Child Health Care Hospital), Hangzhou, Zhejiang, 310000, China
| | - Long Sun
- Department of Clinical Laboratory, Hangzhou Women's Hospital (Hangzhou Matenal and Child Health Care Hospital), Hangzhou, Zhejiang, 310000, China
| | - Xinyan Shi
- Department of Clinical Laboratory, Hangzhou Women's Hospital (Hangzhou Matenal and Child Health Care Hospital), Hangzhou, Zhejiang, 310000, China
| | - Xian Huang
- Respiratory department, Zhejiang Provincial General Hospital of Chinese People's Armed Police CN, Hangzhou, Zhejiang, 310051, China
| | - Yiping Yin
- Department of Hospital-acquired infection control, Zhejiang Provincial General Hospital of Chinese People's Armed Police CN, Hangzhou, Zhejiang, 310051, China
| | - Beilei Pan
- Department of Clinical Laboratory, Hangzhou Women's Hospital (Hangzhou Matenal and Child Health Care Hospital), Hangzhou, Zhejiang, 310000, China
| | - Lihua Hu
- Department of Critical Care Medicine, Zhejiang Provincial General Hospital of Chinese People's Armed Police CN, Hangzhou, Zhejiang, 310051, China.
| | - Qiang Shen
- Hangzhou xixi Hospital, Hangzhou, Zhejiang, 310023, China.
| |
Collapse
|
39
|
AlJindan R, Mahmoud N, AlEraky DM, Almandil NB, AbdulAzeez S, Borgio JF. Phenomics and genomic features of Enterococcus avium IRMC1622a isolated from a clinical sample of hospitalized patient. J Infect Public Health 2024; 17:102463. [PMID: 38833914 DOI: 10.1016/j.jiph.2024.05.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 05/16/2024] [Accepted: 05/26/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Enterococcus avium (E. avium) is a Gram-positive nosocomial pathogen that is commonly isolated from the alimentary tract. The objective of this functional genomics study was to identify the resistant genes by analyzing the genome of E. avium IRMC1622a, a type of bacteria found in feces collected from a patient at a Saudi Arabian tertiary hospital. METHODS The bacterial strain IRMC1622a was identified by 16 S rRNA sequencing as Enterococcus sp. The resistance phenomics were performed using VITEK® 2, and morphological analysis was achieved using a scanning electron microscope (SEM). Finally, the whole bacterial genome of the bacterial strain IRMC1622a was subjected to sequencing during October 2023 using Oxford Nanopore long-read sequencing technology, and mining for resistant genes. RESULTS The results of antimicrobial resistant phenomics indicated that the IRMC1622a strain was sensitive to all tested antimicrobial agents except for erythromycin, and the same result was confirmed by genomic analysis in addition to other classes of antibiotics. SEM showed E. avium IRMC1622a is ovoid shape, in single cells (L 1.2797 ± 0.1490 µm), in pairs (L 1.7333 ± 0.1054 µm), and in chains (L 2.44033 ± 0.1978 µm). The E. avium IRMC1622a genome has 14 (in CARD) antimicrobial resistance genes that were identified with several mechanisms of antimicrobial resistance, such as the efflux pump and conferring antibiotic resistance. The present study revealed that the E. avium IRMC1622a genome contains a high number of genes associated with virulence factors, and 14 matched pathogenic protein families and predicted as human pathogen (probability score 0.855). We report two (ISEnfa4 and ISEfa5) mobile genetic elements for the first time in the E. avium genome. CONCLUSIONS The study concludes that E. avium IRMC1622a is susceptible to all tested antibacterials except erythromycin. The IRMC1622a has 14 genes encoding antimicrobial resistance mechanisms, including the efflux pump and conferring antibiotic resistance. This could indicate a potential rise in E. avium resistance in healthcare facilities. These observations may raise concerns regarding E. avium resistance in healthcare. We need more research to understand the pathophysiology of E. avium, which leads to hospital-acquired infections.
Collapse
Affiliation(s)
- Reem AlJindan
- Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Nehal Mahmoud
- Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Doaa M AlEraky
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Noor B Almandil
- Department of Clinical Pharmacy Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia.
| | - Sayed AbdulAzeez
- Department of Genetic Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - J Francis Borgio
- Department of Genetic Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia.
| |
Collapse
|
40
|
Liu X, Xiong Y, Peng R, Zhang Y, Cai S, Deng Q, Yu Z, Wen Z, Chen Z, Hou T. Antibacterial activity and mechanisms of D-3263 against Staphylococcus aureus. BMC Microbiol 2024; 24:224. [PMID: 38926818 PMCID: PMC11201875 DOI: 10.1186/s12866-024-03377-3] [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: 02/19/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Multi-drug-resistant Staphylococcus aureus infections necessitate novel antibiotic development. D-3263, a transient receptor potential melastatin member 8 (TRPM8) agonist, has potential antineoplastic properties. Here, we reported the antibacterial and antibiofilm activities of D-3263. Minimum inhibitory concentrations (MICs) against S. aureus, Enterococcus faecalis and E. faecium were ≤ 50 µM. D-3263 exhibited bactericidal effects against clinical methicillin-resistant S. aureus (MRSA) and E. faecalis strains at 4× MIC. Subinhibitory D-3263 concentrations effectively inhibited S. aureus and E. faecalis biofilms, with higher concentrations also clearing mature biofilms. Proteomic analysis revealed differential expression of 29 proteins under 1/2 × MIC D-3263, influencing amino acid biosynthesis and carbohydrate metabolism. Additionally, D-3263 enhanced membrane permeability of S. aureus and E. faecalis. Bacterial membrane phospholipids phosphatidylethanolamine (PE), phosphatidylglycerol (PG), and cardiolipin (CL) dose-dependently increased D-3263 MICs. Overall, our data suggested that D-3263 exhibited potent antibacterial and antibiofilm activities against S. aureus by targeting the cell membrane.
Collapse
Affiliation(s)
- Xiaoju Liu
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Yanpeng Xiong
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Renhai Peng
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Yufang Zhang
- Department of Biology, Washington University in St. Louis, 1 Brookings Drive, St Louis, MO, 63130, USA
| | - Shuyu Cai
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Qiwen Deng
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Zhijian Yu
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Zewen Wen
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Zhong Chen
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
| | - Tieying Hou
- Department of Infectious Diseases, Shenzhen Key Laboratory for Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
- Department of Infectious Diseases and Shenzhen key Laboratory of Endogenous infection, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
| |
Collapse
|
41
|
Rubilar-Huenchuman M, Ortega-Villanueva C, González IA, Palavecino CE. The Effect of Photodynamic Therapy on Enterococcus spp. and Its Application in Dentistry: A Scoping Review. Pharmaceutics 2024; 16:825. [PMID: 38931945 PMCID: PMC11207625 DOI: 10.3390/pharmaceutics16060825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Enterococci spp. are Gram-positive bacteria that cause mild to severe infections, many associated with the oral cavity, such as periapical infections and healthcare-associated infections (HAIs). Many of these infections become serious diseases that are difficult to resolve, specifically when multidrug-resistant (MDR) strains cause them. In recent years, the number of MDR strains of Enterococcus spp. has increased significantly. This increased prevalence of MDR strains produces significant pressure to generate more antimicrobial therapies, but there is a decline in the production of new antibiotics, driving the development of complementary therapies, such as photodynamic therapy (PDT). PDT combines a photosensitizer agent (PS), light, and oxygen to cause photooxidative stress in bacterial cells. PDT can eradicate Enterococcus spp. contaminations, improve the classic cleaning processes, and eradicate the bacteria in dental pieces. PDT's effectiveness can be improved with nanoparticles that function as carriers. Our work aims to describe the advances in PDT against Enterococcus spp. as a complement to antibiotic therapy, focusing on infections by Enterococcus faecium and Enterococcus faecalis, dental hygiene, and using nanoparticles to improve the antimicrobial effect. A systematic bibliographic search without a meta-analysis was conducted on various databases, using inclusion and exclusion criteria to identify the most relevant research. Of the 193 non-redundant articles found, 65 were selected for a systematic review, from which a summary table was created and a manual description was made. Photodynamic therapy for treating E. faecium and E. faecalis is a widely studied area, with promising results concerning bactericidal effectiveness and reductions in biofilm formation, particularly in regard to dental hygiene. Because most of the studies were conducted in vitro or ex vivo, the results indicated that there were not sufficient data to initiate clinical trials for safety and efficacy studies on humans.
Collapse
Affiliation(s)
- Mariaignacia Rubilar-Huenchuman
- Laboratorio de Microbiología Celular, Facultad de Medicina y Ciencias de la Salud, Universidad Central de Chile, Lord Cochrane 418, Santiago 8330546, Chile; (M.R.-H.); (C.O.-V.)
| | - Camilo Ortega-Villanueva
- Laboratorio de Microbiología Celular, Facultad de Medicina y Ciencias de la Salud, Universidad Central de Chile, Lord Cochrane 418, Santiago 8330546, Chile; (M.R.-H.); (C.O.-V.)
| | - Iván A. González
- Departamento de Química, Facultad de Ciencias Naturales, Matemática y del Medio Ambiente, Universidad Tecnológica Metropolitana, Las Palmeras 3360, Ñuñoa, Santiago 7800003, Chile;
| | - Christian Erick Palavecino
- Laboratorio de Microbiología Celular, Facultad de Medicina y Ciencias de la Salud, Universidad Central de Chile, Lord Cochrane 418, Santiago 8330546, Chile; (M.R.-H.); (C.O.-V.)
| |
Collapse
|
42
|
Klupt S, Fam KT, Zhang X, Chodisetti PK, Mehmood A, Boyd T, Grotjahn D, Park D, Hang HC. Secreted antigen A peptidoglycan hydrolase is essential for Enterococcus faecium cell separation and priming of immune checkpoint inhibitor therapy. eLife 2024; 13:RP95297. [PMID: 38857064 PMCID: PMC11164530 DOI: 10.7554/elife.95297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024] Open
Abstract
Enterococcus faecium is a microbiota species in humans that can modulate host immunity (Griffin and Hang, 2022), but has also acquired antibiotic resistance and is a major cause of hospital-associated infections (Van Tyne and Gilmore, 2014). Notably, diverse strains of E. faecium produce SagA, a highly conserved peptidoglycan hydrolase that is sufficient to promote intestinal immunity (Rangan et al., 2016; Pedicord et al., 2016; Kim et al., 2019) and immune checkpoint inhibitor antitumor activity (Griffin et al., 2021). However, the functions of SagA in E. faecium were unknown. Here, we report that deletion of sagA impaired E. faecium growth and resulted in bulged and clustered enterococci due to defective peptidoglycan cleavage and cell separation. Moreover, ΔsagA showed increased antibiotic sensitivity, yielded lower levels of active muropeptides, displayed reduced activation of the peptidoglycan pattern-recognition receptor NOD2, and failed to promote cancer immunotherapy. Importantly, the plasmid-based expression of SagA, but not its catalytically inactive mutant, restored ΔsagA growth, production of active muropeptides, and NOD2 activation. SagA is, therefore, essential for E. faecium growth, stress resistance, and activation of host immunity.
Collapse
Affiliation(s)
- Steven Klupt
- Department of Immunology and Microbiology, Scripps ResearchLa JollaUnited States
| | - Kyong Tkhe Fam
- Department of Immunology and Microbiology, Scripps ResearchLa JollaUnited States
| | - Xing Zhang
- Department of Immunology and Microbiology, Scripps ResearchLa JollaUnited States
| | | | - Abeera Mehmood
- Department of Immunology and Microbiology, Scripps ResearchLa JollaUnited States
| | - Tumara Boyd
- Department of Integrative Structural & Computational Biology, Scripps ResearchLa JollaUnited States
| | - Danielle Grotjahn
- Department of Integrative Structural & Computational Biology, Scripps ResearchLa JollaUnited States
| | - Donghyun Park
- Department of Integrative Structural & Computational Biology, Scripps ResearchLa JollaUnited States
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps ResearchLa JollaUnited States
- Department of Chemistry, Scripps ResearchLa JollaUnited States
| |
Collapse
|
43
|
Cebeci T. Species prevalence, virulence genes, and antibiotic resistance of enterococci from food-producing animals at a slaughterhouse in Turkey. Sci Rep 2024; 14:13191. [PMID: 38851786 PMCID: PMC11162463 DOI: 10.1038/s41598-024-63984-y] [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: 03/07/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024] Open
Abstract
Healthy cattle, sheep, and goats can be reservoirs for gastrointestinal pathogenic fecal enterococci, some of which could be multidrug-resistant to antimicrobials. The objective of this study was to determine the prevalence and diversity of Enterococcus species in healthy sheep, goat, and cattle carcasses, as well as to analyze the antimicrobial resistance phenotype/genotype and the virulence gene content. During 2019-2020, carcass surface samples were collected from 150 ruminants in a slaughterhouse. A total of 90 enterococci, comprising five species, were obtained. The overall prevalence of enterococci was found to be 60%, out of which 37.7% were identified as Enterococcus (E.) hirae, 33.3% as E. casseliflavus, 15.5% as E. faecium, 12.2% as E. faecalis, and 1.1% as E. gallinarum. Virulence-associated genes of efaA (12.2%) were commonly observed in the Enterococcus isolates, followed by gelE (3.3%), asaI (3.3%), and ace (2.2%). High resistance to quinupristin-dalfopristin (28.8%), tetracycline (21.1%), ampicillin (20%), and rifampin (15.5%) was found in two, four, four, and five of the Enterococcus species group, respectively. The resistance of Enterococcus isolates to 11 antibiotic groups was determined and multidrug resistant (MDR) strains were found in 18.8% of Enterococcus isolates. Characteristic resistance genes were identified by PCR with an incidence of 6.6%, 2.2%, 1.1%, 1.1%, 1.1%, and 1.1% for the tetM, ermB, ermA, aac(6')Ie-aph(2")-la, VanC1, and VanC2 genes in Enterococcus isolates, respectively. Efflux pump genes causing multidrug resistance were detected in Enterococcus isolates (34.4%). The results showed that there were enterococci in the slaughterhouse with a number of genes linked to virulence that could be harmful to human health.
Collapse
Affiliation(s)
- Tugba Cebeci
- Department of Medical Services and Techniques, Espiye Vocational School, Giresun University, Giresun, Turkey.
| |
Collapse
|
44
|
Hallenbeck M, Chua M, Collins J. The role of the universal sugar transport system components PtsI (EI) and PtsH (HPr) in Enterococcus faecium. FEMS MICROBES 2024; 5:xtae018. [PMID: 38988831 PMCID: PMC11234649 DOI: 10.1093/femsmc/xtae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 07/12/2024] Open
Abstract
Vancomycin-resistant enterococci (VRE) pose a serious threat to public health because of their limited treatment options. Therefore, there is an increasing need to identify novel targets to develop new drugs. Here, we examined the roles of the universal PTS components, PtsI and PtsH, in Enterococcus faecium to determine their roles in carbon metabolism, biofilm formation, stress response, and the ability to compete in the gastrointestinal tract. Clean deletion of ptsHI resulted in a significant reduction in the ability to import and metabolize simple sugars, attenuated growth rate, reduced biofilm formation, and decreased competitive fitness both in vitro and in vivo. However, no significant difference in stress survival was observed when compared with the wild type. These results suggest that targeting universal or specific PTS may provide a novel treatment strategy by reducing the fitness of E. faecium.
Collapse
Affiliation(s)
- Michelle Hallenbeck
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, United States
| | - Michelle Chua
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, United States
| | - James Collins
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, United States
- Center for Microbiomics, Inflammation and Pathogenicity, University of Louisville, Louisville, KY 40202, United States
| |
Collapse
|
45
|
Tadesse BT, Svetlicic E, Zhao S, Berhane N, Jers C, Solem C, Mijakovic I. Bad to the bone? - Genomic analysis of Enterococcus isolates from diverse environments reveals that most are safe and display potential as food fermentation microorganisms. Microbiol Res 2024; 283:127702. [PMID: 38552381 DOI: 10.1016/j.micres.2024.127702] [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: 11/29/2023] [Revised: 02/09/2024] [Accepted: 03/20/2024] [Indexed: 04/17/2024]
Abstract
Enterococci comprise a group of lactic acid bacteria (LAB) with considerable potential to serve as food fermentation microorganisms. Unfortunately, enterococci have received a lot of negative attention, due to the occurrence of pathogenic and multidrug resistant strains. In this study, we used genomics to select safe candidates among the forty-four studied enterococcal isolates. The genomes of the forty-four strains were fully sequenced and assessed for presence of virulence and antibiotic resistance genes. Nineteen isolates belonging to the species Enterococcus lactis, Enterococcus faecium, Enterococcus durans, and Enterococcus thailandicus, were deemed safe from the genome analysis. The presence of secondary metabolite gene clusters for bacteriocins was assessed, and twelve candidates were found to secrete antimicrobial compounds effective against Listeria monocytogenes isolated from cheese and Staphylococcus aureus. Physiological characterization revealed nineteen industrial potentials; all strains grew well at 42 °C and acidified 1.5 hours faster than their mesophilic counterpart Lactococcus lactis, with which they share metabolism and flavor forming ability. We conclude that a large fraction of the examined enterococci were safe and could serve as excellent food fermentation microorganisms with inherent bioprotective abilities.
Collapse
Affiliation(s)
- Belay Tilahun Tadesse
- National Food Institute, Research Group for Microbial Biotechnology and Biorefining, Technical University of Denmark, Kgs Lyngby DK-2800, Denmark; Novo Nordisk Foundation Center for Biosustainability, Kongens Lyngby, Denmark
| | - Ema Svetlicic
- Novo Nordisk Foundation Center for Biosustainability, Kongens Lyngby, Denmark
| | - Shuangqing Zhao
- National Food Institute, Research Group for Microbial Biotechnology and Biorefining, Technical University of Denmark, Kgs Lyngby DK-2800, Denmark
| | - Nega Berhane
- Institute of Biotechnology, University of Gondar, Ethiopia
| | - Carsten Jers
- Novo Nordisk Foundation Center for Biosustainability, Kongens Lyngby, Denmark
| | - Christian Solem
- National Food Institute, Research Group for Microbial Biotechnology and Biorefining, Technical University of Denmark, Kgs Lyngby DK-2800, Denmark.
| | - Ivan Mijakovic
- Novo Nordisk Foundation Center for Biosustainability, Kongens Lyngby, Denmark; Systems and Synthetic Biology Division, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
| |
Collapse
|
46
|
Al-Ghamdi AY. Caffeic acid phenethyl ester attenuates Enterococcus faecalis infection in vivo: antioxidants and NF-κB have a protective role against stomach damage. J Med Life 2024; 17:574-581. [PMID: 39296435 PMCID: PMC11407487 DOI: 10.25122/jml-2023-0544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 04/16/2024] [Indexed: 09/21/2024] Open
Abstract
The mammalian gastrointestinal tract hosts a significant microbial symbiont community, an intriguing feature of this complex organ system. This study aimed to investigate the anti-inflammatory, antioxidant, and protective effects of caffeic acid phenethyl ester (CAPE) against Enterococcus faecalis infection in the stomach at a dose of 106 CFU in Swiss mice. A total of 30 mice were randomly assigned to three groups of ten mice each. Group I was the negative control, Group II was infected orally with E. faecalis for 18 days, and Group III was infected with E. faecalis and treated with CAPE orally at a daily dose of 4 mg/kg for 18 days. We assessed the antioxidant activities of stomach homogenate and the immunohistochemical expressions of the transcription factor nuclear factor kappa B (NF-κB) and proliferating cell nuclear antigen (PCNA). Histopathological examination was performed on the stomachs of all mice. Group II had decreased levels of antioxidant activity and positive expressions of NF-κB and PCNA. Histological observations revealed an increase in mucosal and glandular thickness compared with Group I. Group III, treated with CAPE, showed a significant increase in antioxidant activities and a significant decrease in NF-κB and PCNA immunoreactivities compared with Group II. In addition, Group III showed restoration of the normal thickness of the non-glandular and glandular parts of the stomach. Our results revealed that E. faecalis infection has damaging effects on the stomach and proved that CAPE has promising protective, anti-inflammatory, and antioxidant effects against E. faecalis. Further studies may investigate the potential therapeutic effects of CAPE against E. faecalis infection.
Collapse
|
47
|
Van Looveren N, IJdema F, van der Heijden N, Van Der Borght M, Vandeweyer D. Microbial dynamics and vertical transmission of Escherichia coli across consecutive life stages of the black soldier fly (Hermetia illucens). Anim Microbiome 2024; 6:29. [PMID: 38797818 PMCID: PMC11129375 DOI: 10.1186/s42523-024-00317-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND The black soldier fly (BSF, Hermetia illucens L.) is one of the most promising insects for bioconversion of organic waste, which often carry a high microbial load with potential foodborne pathogens. Although horizontal transmission (from rearing substrate to larvae) has been extensively studied, less is known about vertical transmission of microorganisms, and particularly of foodborne pathogens, across different BSF life stages. RESULTS This study investigated the microbial dynamics and vertical transmission of Escherichia coli across different life stages (larvae, prepupae, pupae and adults) of one BSF life cycle and its associated substrate (chicken feed) and frass, based on a combination of general microbial counts (based on culture-dependent techniques) and the bacterial community composition (based on 16S rRNA gene sequencing). Multiple interactions between the microbiota of the substrate, frass and BSF larvae were affirmed. The larvae showed relative consistency among both the microbial counts and bacterial community composition. Diversification of the bacterial communities started during the pupal stage, while most notable changes of the microbial counts and bacterial community compositions occurred during metamorphosis to adults. Furthermore, vertical transmission of E. coli was investigated after substrate inoculation with approximately 7.0 log cfu/g of kanamycin-resistant E. coli, and monitoring E. coli counts from larval to adult stage. Although the frass still contained substantial levels of E. coli (> 4.5 log cfu/g) and E. coli was taken up by the larvae, limited vertical transmission of E. coli was observed with a decreasing trend until the prepupal stage. E. coli counts were below the detection limit (1.0 log cfu/g) for all BSF samples from the end of the pupal stage and the adult stage. Additionally, substrate inoculation of E. coli did not have a substantial impact on the bacterial community composition of the substrate, frass or different BSF life stages. CONCLUSIONS The fluctuating microbial counts and bacterial community composition underscored the dynamic character of the microbiota of BSF life stages. Additionally, vertical transmission throughout one BSF life cycle was not observed for E. coli. Hence, these findings paved the way for future case studies on vertical transmission of foodborne pathogens across consecutive BSF life stages or other insect species.
Collapse
Affiliation(s)
- Noor Van Looveren
- KU Leuven, Geel Campus, Department of Microbial and Molecular Systems (M2S), Research Group for Insect Production and Processing, Kleinhoefstraat 4, Geel, 2440, Belgium
| | - Freek IJdema
- KU Leuven, Geel Campus, Department of Microbial and Molecular Systems (M2S), Research Group for Insect Production and Processing, Kleinhoefstraat 4, Geel, 2440, Belgium
| | - Niels van der Heijden
- KU Leuven, Geel Campus, Department of Microbial and Molecular Systems (M2S), Research Group for Insect Production and Processing, Kleinhoefstraat 4, Geel, 2440, Belgium
| | - Mik Van Der Borght
- KU Leuven, Geel Campus, Department of Microbial and Molecular Systems (M2S), Research Group for Insect Production and Processing, Kleinhoefstraat 4, Geel, 2440, Belgium
| | - Dries Vandeweyer
- KU Leuven, Geel Campus, Department of Microbial and Molecular Systems (M2S), Research Group for Insect Production and Processing, Kleinhoefstraat 4, Geel, 2440, Belgium.
| |
Collapse
|
48
|
Labecka L, Ķibilds J, Cīrulis A, Čeirāne ED, Zeltiņa I, Reinis A, Vilima B, Rudzīte D, Erts R, Mauliņa I, Bandere D, Krūmiņa A. Evaluation of Antimicrobial Resistancein Clinical Isolates of Enterococcus spp. Obtained from Hospital Patients in Latvia. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:850. [PMID: 38929467 PMCID: PMC11206013 DOI: 10.3390/medicina60060850] [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: 02/09/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
Background and Objective: Enterococci are typically found in a healthy human gastrointestinal tract but can cause severe infections in immunocompromised patients. Such infections are treated with antibiotics. This study addresses the rising concern of antimicrobial resistance (AMR) in Enterococci, focusing on the prevalence of vancomycin-resistant enterococcus (VRE) strains. Materials and Methods: The pilot study involved 140 Enterococci isolates collected between 2021 and 2022 from two multidisciplinary hospitals (with and without local therapeutic drug monitoring protocol of vancomycin) in Latvia. Microbiological assays and whole genome sequencing were used. AMR gene prevalence with resistance profiles were determined and the genetic relationship and outbreak evaluation were made by applying core genome multi-locus sequence typing (cgMLST). Results: The acquired genes and mutations were responsible for resistance against 10 antimicrobial classes, including 25.0% of isolates expressing resistance to vancomycin, predominantly of the vanB type. Genetic diversity among E. faecalis and E. faecium isolates was observed and seven potential outbreak clusters were identified, three of them containing sequence types ST6, ST78 and ST80. The prevalence of vancomycin resistance was highest in the hospital without a therapeutic drug-monitoring protocol and in E. faecium. Notably, a case of linezolid resistance due to a mutation was documented. Conclusions: The study illustrates the concerning prevalence of multidrug-resistant Enterococci in Latvian hospitals, showcasing the rather widespread occurrence of vancomycin-resistant strains. This highlights the urgency of implementing efficient infection control mechanisms and the need for continuous VRE surveillance in Latvia to define the scope and pattern of the problem, influencing clinical decision making and planning further preventative measures.
Collapse
Affiliation(s)
- Linda Labecka
- Institute of Food Safety, Animal Health and Environment “BIOR”, Lejupes Street 3, LV-1076 Riga, Latvia (A.C.); (A.K.)
| | - Juris Ķibilds
- Institute of Food Safety, Animal Health and Environment “BIOR”, Lejupes Street 3, LV-1076 Riga, Latvia (A.C.); (A.K.)
| | - Aivars Cīrulis
- Institute of Food Safety, Animal Health and Environment “BIOR”, Lejupes Street 3, LV-1076 Riga, Latvia (A.C.); (A.K.)
- Faculty of Biology, University of Latvia, Jelgavas Street 1, LV-1004 Riga, Latvia
| | - Evelīna Diāna Čeirāne
- Faculty of Medicine, Riga Stradins University, Dzirciema Street 16, LV-1007 Riga, Latvia
| | - Indra Zeltiņa
- Department of Infectology, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia;
- Riga East Clinical University Hospital, Hipokrata Street 2, LV-1038 Riga, Latvia; (D.R.); (I.M.)
| | - Aigars Reinis
- Department of Biology and Microbiology, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia;
- Pauls Stradiņš Clinical University Hospital, Pilsoņu Street 13, LV-1002 Riga, Latvia
| | - Barba Vilima
- Vidzeme Hospital, Jumaras Street 195, LV-4201 Valmiera, Latvia;
| | - Dace Rudzīte
- Riga East Clinical University Hospital, Hipokrata Street 2, LV-1038 Riga, Latvia; (D.R.); (I.M.)
| | - Renārs Erts
- Faculty of Medicine, University of Latvia, Raiņa bulvāris 19, LV-1586 Riga, Latvia;
| | - Inga Mauliņa
- Riga East Clinical University Hospital, Hipokrata Street 2, LV-1038 Riga, Latvia; (D.R.); (I.M.)
- Vidzeme Hospital, Jumaras Street 195, LV-4201 Valmiera, Latvia;
- Department of Pharmaceutical Chemistry, Riga Stradiņš University, LV-1007 Riga, Latvia;
| | - Dace Bandere
- Department of Pharmaceutical Chemistry, Riga Stradiņš University, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, LV-1658 Riga, Latvia
| | - Angelika Krūmiņa
- Institute of Food Safety, Animal Health and Environment “BIOR”, Lejupes Street 3, LV-1076 Riga, Latvia (A.C.); (A.K.)
- Department of Infectology, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia;
| |
Collapse
|
49
|
van Heule M, El-Sheikh Ali H, Monteiro HF, Scoggin K, Fedorka C, Weimer BC, Ball B, Daels P, Dini P. Characterization of the equine placental microbial population during nocardioform placentitis. Theriogenology 2024; 225:172-179. [PMID: 38810343 DOI: 10.1016/j.theriogenology.2024.05.025] [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: 02/02/2024] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 05/31/2024]
Abstract
Nocardioform placentitis is a poorly understood disease of equine late gestation. The presence of nocardioform, filamentous branching gram-positive bacteria, has been linked to the disease, with Crossiella equi, Amycolatopsis spp., and Streptomyces spp. being the most frequently identified bacteria. However, these bacteria are not found in all clinical cases in addition to being isolated from healthy, normal postpartum placentas. To better understand this form of placentitis, we analyzed the microbial composition in the equine placenta (chorioallantois) of both healthy postpartum (control; n = 11) and nocardioform-affected samples (n = 22) using 16S rDNA sequencing. We found a lower Shannon index in nocardioform samples, a higher Chao1 index in nocardioform samples, and a difference in beta diversity between control and nocardioform samples (p < 0.05), suggesting the presence of dysbiosis during the disease. In the majority of the NP samples (77 %), one of the following genera-Amycolatopsis, Crossiella, Lentzea, an unidentified member of the Pseudonocardiaceae family, Mycobacterium, or Enterococcus -represented over 70 % of the relative abundance. Overall, the data suggest that a broader spectrum of potential opportunistic pathogens could be involved in nocardioform placentitis, extending beyond the traditionally recognized bacteria, resulting in a similar histomorphological profile.
Collapse
Affiliation(s)
- Machteld van Heule
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA; Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, University of Ghent, Merelbeke, 9800, Belgium
| | - Hossam El-Sheikh Ali
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40503, USA
| | - Hugo Fernando Monteiro
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Kirsten Scoggin
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40503, USA
| | - Carleigh Fedorka
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40503, USA
| | - Bart C Weimer
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Barry Ball
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40503, USA
| | - Peter Daels
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, University of Ghent, Merelbeke, 9800, Belgium
| | - Pouya Dini
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
50
|
Elnar AG, Kim GB. In Vitro and In Silico Characterization of N-Formylated Two-Peptide Bacteriocin from Enterococcus faecalis CAUM157 with Anti-Listeria Activity. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10265-9. [PMID: 38743207 DOI: 10.1007/s12602-024-10265-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Enterococcus faecalis CAUM157 (KACC 81148BP), a Gram-positive bacteria isolated from raw cow's milk, was studied for its bacteriocin production. The antimicrobial activity of CAUM157 was attributed to a two-peptide class IIb bacteriocin with potent activity against food-borne pathogen Listeria monocytogenes and periodontal disease-causing pathogens (Prevotella intermedia KCTC 15693 T and Fusobacterium nucleatum KCTC 2488 T). M157 bacteriocins exhibit high temperature and pH stability and resist hydrolytic enzyme degradation and detergent denaturation, potentially due to their structural conformation. Based on amino acid sequence, M157A and M157B were predicted to be 5.176 kDa and 5.182 kDa in size, respectively. However, purified bacteriocins and chemically synthesized N-formylated M157 peptides both showed 5.204 kDa (M157A) and 5.209 kDa (M157B) molecular mass, confirming the formylation of the N-terminal methionine of both peptides produced by strain CAUM157. Furthermore, the strain demonstrated favorable growth and fermentation with minimal bacteriocin production when cultured in whey-based media, whereas a 1.0% tryptone or soytone supplementation resulted in higher bacteriocin production. Although Ent. faecalis CAUM157 innately harbors genes for virulence factors and antimicrobial resistance (e.g., tetracycline and erythromycin), its bacteriocin production is valuable in circumventing the need for live microorganisms, particularly in food applications for pathogen control.
Collapse
Affiliation(s)
- Arxel G Elnar
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Geun-Bae Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea.
| |
Collapse
|