1
|
Mei LN, Wang ZJ, Duan Y, Shen JS, Ye HB, Zhu YY, Luo XD. 4-Hydroxyboesenbergin B of Alpinia japonica protected gastrointestinal tract by inhibiting vancomycin-resistant enterococcus and balancing intestinal microbiota. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119737. [PMID: 40179999 DOI: 10.1016/j.jep.2025.119737] [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: 12/29/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alpinia japonica, a traditional herb utilized in Miao medicine in southwestern China, has been employed to alleviate symptoms such as stomachache, diarrhea, and abdominal pain, some of these symptoms may be associated with bacterial infections of the gastrointestinal tract. AIM OF THE STUDY To explore antimicrobial compounds related to traditional uses of A. japonica and its potential pathway in vitro and in vivo. MATERIALS AND METHODS Bioactive components of A. japonica were isolated by bioguide separation method. The antibacterial bioactivity of 4-hydroxyboesenbergin B (4-HB) was evaluated by time-kill curve and drug resistance induction. The pathway of 4-HB against VRE was investigated through network pharmacological analysis and validated by in vitro experiments and RT-qPCR assays. Moreover, a mouse gastrointestinal tract model was established to validate the antibacterial bioactivity of 4-HB in vivo. RESULTS 4-HB from A. japonica inhibited VRE (MIC = 16 μg/mL), rapidly killed the bacteria within 4 h at the 4 MIC concentration and exhibited low susceptibility to drug resistance. 4-HB specifically targeted VRE biofilms by down-regulating the expression of AtlA, SgrA, GelE, and Ace. As a result, 4-HB diminished the adhesion and aggregation ability of VRE, reduced the extracellular matrix content, disrupted biofilm structure and morphology, thereby reducing VRE resistance and virulence. Additionally, 4-HB significantly reduced VRE colonization, enhanced intestinal microbiota diversity, and promoted the restoration of intestinal microbiota balance in vivo. Notably, 4-HB enhanced the abundance of beneficial bacteria genera, such as Lactobacillus and Limosilactobacillus. CONCLUSIONS 4-HB has a significant ability to destroy VRE biofilms and balance intestinal microbiota, which might be responsible for the traditional use of A. japonica partly.
Collapse
Affiliation(s)
- Li-Na Mei
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Yu Duan
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Jia-Shan Shen
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Hong-Bo Ye
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences Kunming, 650201, China.
| |
Collapse
|
2
|
Higuita J, Arango M, Forga A, Cortes D, Graham D. An Updated Review of Enterococcus cecorum Infections in Poultry. Avian Dis 2025; 68:404-411. [PMID: 40249579 DOI: 10.1637/aviandiseases-d-24-00098] [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: 11/25/2024] [Accepted: 12/16/2024] [Indexed: 04/19/2025]
Abstract
Since the early 2000s, skeletal effects, specifically enterococcal spondylitis, related to pathogenic Enterococcus cecorum (EC), have been observed in older broiler chicken flocks. This skeletal involvement has typically been associated with persistent EC infections in the free thoracic vertebrae leading to paralysis. However, the emergence of virulent EC in young broiler chicken flocks causing clinical septicemia requires further investigation. The purpose of this review is to provide an update on EC-related research and pending industry needs.
Collapse
Affiliation(s)
- J Higuita
- Department of Poultry Science, University of Arkansas, Division of Agriculture, Fayetteville AR, 72701
| | - M Arango
- Department of Poultry Science, University of Arkansas, Division of Agriculture, Fayetteville AR, 72701
| | - A Forga
- Department of Poultry Science, University of Arkansas, Division of Agriculture, Fayetteville AR, 72701
| | - D Cortes
- Department of Poultry Science, University of Arkansas, Division of Agriculture, Fayetteville AR, 72701
| | - D Graham
- Department of Poultry Science, University of Arkansas, Division of Agriculture, Fayetteville AR, 72701,
| |
Collapse
|
3
|
Willett JLE, Dunny GM. Insights into ecology, pathogenesis, and biofilm formation of Enterococcus faecalis from functional genomics. Microbiol Mol Biol Rev 2025; 89:e0008123. [PMID: 39714182 PMCID: PMC11948497 DOI: 10.1128/mmbr.00081-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 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
|
4
|
Smith OER, Bharat TAM. Architectural dissection of adhesive bacterial cell surface appendages from a "molecular machines" viewpoint. J Bacteriol 2024; 206:e0029024. [PMID: 39499080 PMCID: PMC7616799 DOI: 10.1128/jb.00290-24] [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] [Indexed: 11/07/2024] Open
Abstract
The ability of bacteria to interact with and respond to their environment is crucial to their lifestyle and survival. Bacterial cells routinely need to engage with extracellular target molecules, in locations spatially separated from their cell surface. Engagement with distant targets allows bacteria to adhere to abiotic surfaces and host cells, sense harmful or friendly molecules in their vicinity, as well as establish symbiotic interactions with neighboring cells in multicellular communities such as biofilms. Binding to extracellular molecules also facilitates transmission of information back to the originating cell, allowing the cell to respond appropriately to external stimuli, which is critical throughout the bacterial life cycle. This requirement of bacteria to bind to spatially separated targets is fulfilled by a myriad of specialized cell surface molecules, which often have an extended, filamentous arrangement. In this review, we compare and contrast such molecules from diverse bacteria, which fulfil a range of binding functions critical for the cell. Our comparison shows that even though these extended molecules have vastly different sequence, biochemical and functional characteristics, they share common architectural principles that underpin bacterial adhesion in a variety of contexts. In this light, we can consider different bacterial adhesins under one umbrella, specifically from the point of view of a modular molecular machine, with each part fulfilling a distinct architectural role. Such a treatise provides an opportunity to discover fundamental molecular principles governing surface sensing, bacterial adhesion, and biofilm formation.
Collapse
Affiliation(s)
- Olivia E. R. Smith
- Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Tanmay A. M. Bharat
- Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
5
|
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
|
6
|
Mullally CA, Fahriani M, Mowlaboccus S, Coombs GW. Non- faecium non- faecalis enterococci: a review of clinical manifestations, virulence factors, and antimicrobial resistance. Clin Microbiol Rev 2024; 37:e0012123. [PMID: 38466110 PMCID: PMC11237509 DOI: 10.1128/cmr.00121-23] [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: 03/12/2024] Open
Abstract
SUMMARYEnterococci are a diverse group of Gram-positive bacteria that are typically found as commensals in humans, animals, and the environment. Occasionally, they may cause clinically relevant diseases such as endocarditis, septicemia, urinary tract infections, and wound infections. The majority of clinical infections in humans are caused by two species: Enterococcus faecium and Enterococcus faecalis. However, there is an increasing number of clinical infections caused by non-faecium non-faecalis (NFF) enterococci. Although NFF enterococcal species are often overlooked, studies have shown that they may harbor antimicrobial resistance (AMR) genes and virulence factors that are found in E. faecium and E. faecalis. In this review, we present an overview of the NFF enterococci with a particular focus on human clinical manifestations, epidemiology, virulence genes, and AMR genes.
Collapse
Affiliation(s)
- Christopher A Mullally
- Antimicrobial Resistance and Infectious Diseases (AMRID) Research Laboratory, Murdoch University, Murdoch, Western Australia, Australia
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Marhami Fahriani
- Antimicrobial Resistance and Infectious Diseases (AMRID) Research Laboratory, Murdoch University, Murdoch, Western Australia, Australia
| | - Shakeel Mowlaboccus
- Antimicrobial Resistance and Infectious Diseases (AMRID) Research Laboratory, Murdoch University, Murdoch, Western Australia, Australia
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
- PathWest Laboratory Medicine-WA, Department of Microbiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Geoffrey W Coombs
- Antimicrobial Resistance and Infectious Diseases (AMRID) Research Laboratory, Murdoch University, Murdoch, Western Australia, Australia
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
- PathWest Laboratory Medicine-WA, Department of Microbiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| |
Collapse
|
7
|
Sivaramalingam SS, Jothivel D, Govindarajan DK, Kadirvelu L, Sivaramakrishnan M, Chithiraiselvan DD, Kandaswamy K. Structural and functional insights of sortases and their interactions with antivirulence compounds. Curr Res Struct Biol 2024; 8:100152. [PMID: 38989133 PMCID: PMC11231552 DOI: 10.1016/j.crstbi.2024.100152] [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: 12/31/2023] [Revised: 05/18/2024] [Accepted: 06/11/2024] [Indexed: 07/12/2024] Open
Abstract
Sortase proteins play a crucial role as integral membrane proteins in anchoring bacterial surface proteins by recognizing them through a Cell-Wall Sorting (CWS) motif and cleaving them at specific sites before initiating pilus assembly. Both sortases and their substrate proteins are major virulence factors in numerous Gram-positive pathogens, making them attractive targets for antimicrobial intervention. Recognizing the significance of virulence proteins, a comprehensive exploration of their structural and functional characteristics is essential to enhance our understanding of pilus assembly in diverse Gram-positive bacteria. Therefore, this review article discusses the structural features of different classes of sortases and pilin proteins, primarily serving as substrates for sortase-assembled pili. Moreover, it thoroughly examines the molecular-level interactions between sortases and their inhibitors, providing insights from both structural and functional perspectives. In essence, this review article will provide a contemporary and complete understanding of both sortase pathways and various strategies to target them effectively to counteract the virulence.
Collapse
Affiliation(s)
- Sowmiya Sri Sivaramalingam
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | - Deepsikha Jothivel
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | - Deenadayalan Karaiyagowder Govindarajan
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore
| | - Lohita Kadirvelu
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | - Muthusaravanan Sivaramakrishnan
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
- Department of Biotechnology, Mepco Schlenk Engineering College, Tamil Nadu, India
| | - Dhivia Dharshika Chithiraiselvan
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | - Kumaravel Kandaswamy
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| |
Collapse
|
8
|
Feng Y, Wang S, Liu X, Han Y, Xu H, Duan X, Xie W, Tian Z, Yuan Z, Wan Z, Xu L, Qin S, He K, Huang J. Geometric constraint-triggered collagen expression mediates bacterial-host adhesion. Nat Commun 2023; 14:8165. [PMID: 38071397 PMCID: PMC10710423 DOI: 10.1038/s41467-023-43827-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Cells living in geometrically confined microenvironments are ubiquitous in various physiological processes, e.g., wound closure. However, it remains unclear whether and how spatially geometric constraints on host cells regulate bacteria-host interactions. Here, we reveal that interactions between bacteria and spatially constrained cell monolayers exhibit strong spatial heterogeneity, and that bacteria tend to adhere to these cells near the outer edges of confined monolayers. The bacterial adhesion force near the edges of the micropatterned monolayers is up to 75 nN, which is ~3 times higher than that at the centers, depending on the underlying substrate rigidities. Single-cell RNA sequencing experiments indicate that spatially heterogeneous expression of collagen IV with significant edge effects is responsible for the location-dependent bacterial adhesion. Finally, we show that collagen IV inhibitors can potentially be utilized as adjuvants to reduce bacterial adhesion and thus markedly enhance the efficacy of antibiotics, as demonstrated in animal experiments.
Collapse
Affiliation(s)
- Yuting Feng
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Shuyi Wang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Xiaoye Liu
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, 102206, Beijing, China
| | - Yiming Han
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Hongwei Xu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Xiaocen Duan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Wenyue Xie
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Zhuoling Tian
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Zuoying Yuan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Zhuo Wan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
| | - Liang Xu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Siying Qin
- School of Life Sciences, Peking University, 100871, Beijing, China
| | - Kangmin He
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, 100871, Beijing, China.
| |
Collapse
|
9
|
Elashiry MM, Bergeron BE, Tay FR. Enterococcus faecalis in secondary apical periodontitis: Mechanisms of bacterial survival and disease persistence. Microb Pathog 2023; 183:106337. [PMID: 37683835 DOI: 10.1016/j.micpath.2023.106337] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
Enterococcus faecalis is a commensal bacterium commonly found in the human gastrointestinal tract. However, in individuals with compromised immune systems, the pathogen can lead to severe illness. This opportunistic pathogen is associated with secondary apical diseases and is adept at resisting antibiotics and other forms of treatment because of its numerous virulence factors. Enterococcus faecalis is capable of disrupting the normal functions of immune cells, thereby hindering the body's ability to eradicate the infection. However, intensive research is needed in further understanding the adverse immunomodulatory effects of E. faecalis. Potential strategies specific for eradicating E. faecalis have proven beneficial in the treatment of persistent secondary apical periodontitis.
Collapse
Affiliation(s)
- Mohamed M Elashiry
- Department of Endodontics, Dental College of Georgia, Augusta University, Georgia, USA; Department of Endodontics, Faculty of Dentistry, Ain Shams University, Cairo, Egypt.
| | - Brian E Bergeron
- Department of Endodontics, Dental College of Georgia, Augusta University, Georgia, USA
| | - Franklin R Tay
- Department of Endodontics, Dental College of Georgia, Augusta University, Georgia, USA
| |
Collapse
|
10
|
Codelia-Anjum A, Lerner LB, Elterman D, Zorn KC, Bhojani N, Chughtai B. Enterococcal Urinary Tract Infections: A Review of the Pathogenicity, Epidemiology, and Treatment. Antibiotics (Basel) 2023; 12:antibiotics12040778. [PMID: 37107140 PMCID: PMC10135011 DOI: 10.3390/antibiotics12040778] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Urinary tract infections (UTIs) are among the most common causes of infections worldwide and can be caused by numerous uropathogens. Enterococci are Gram-positive, facultative anaerobic commensal organisms of the gastrointestinal tract that are known uropathogens. Enterococcus spp. has become a leading cause of healthcare associated infections, ranging from endocarditis to UTIs. In recent years, there has been an increase in multidrug resistance due to antibiotic misuse, especially in enterococci. Additionally, infections due to enterococci pose a unique challenge due to their ability to survive in extreme environments, intrinsic antimicrobial resistance, and genomic malleability. Overall, this review aims to highlight the pathogenicity, epidemiology, and treatment recommendations (according to the most recent guidelines) of enterococci.
Collapse
Affiliation(s)
- Alia Codelia-Anjum
- Department of Urology, Weill Cornell Medical College, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Lori B Lerner
- Department of Urology, VA Boston Healthcare System, Boston, MA 02132, USA
| | - Dean Elterman
- Division of Urology, Department of Surgery, University Health Network, University of Toronto, Toronto, ON M5T 2SB, Canada
| | - Kevin C Zorn
- Division of Urology, Centre Hospitalier de l'Université de Monstréal, Montreal, QC H2X 0A9, Canada
| | - Naeem Bhojani
- Division of Urology, Centre Hospitalier de l'Université de Monstréal, Montreal, QC H2X 0A9, Canada
| | - Bilal Chughtai
- Department of Urology, Weill Cornell Medical College, New York Presbyterian Hospital, New York, NY 10065, USA
| |
Collapse
|
11
|
Rao Tatta E, Paul S, Kumavath R. Transcriptome Analysis revealed the Synergism of Novel Rhodethrin inhibition on Biofilm architecture, Antibiotic Resistance and Quorum sensing inEnterococcus faecalis. Gene 2023; 871:147436. [PMID: 37075926 DOI: 10.1016/j.gene.2023.147436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/31/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Enterococcus sp. emerged as an opportunistic nosocomial pathogen with the highest antibiotic resistance and mortality rate. Biofilm is problematic primarily since it is regulated by the global bacterial cell to cell communication mediated by the quorum sensing system. sing system. Thus, potential natural antagonists in a novel drug formulation against biofilm-forming Enterococcus faecalis is critical. We used RNA-Seq to evaluate the effects of the novel molecule rhodethrin with chloramphenicol induced on Enterococcus faecalis and DEGs were identified. In transcriptome sequence analysis, a total of 448 with control Vs rhodethrin, 1591 were in control Vs chloramphenicol, 379 genes were DEGs from control Vs synergies, in rhodethrin with chloramphenicol, 379 genes were differentially expressed, whereas 264 genes were significantly downregulated, indicating that 69.69% ofE. faecaliswas altered. The transcriptional sequence data further expression analysis qRT-PCR, and the results shed that the expression profiles of five significant biofilm formation responsible genes such as, Ace, AtpB, lepA, bopD, and typA, 3 genes involved in quorum sensing are sylA, fsrC and camE, and 4 genes involved in resistance were among including liaX, typA, EfrA, and lepA, were significantly suppressed expressions of the biofilm, quorum sensing, and resistance that are supported by transcriptome analysis.
Collapse
Affiliation(s)
- Eswar Rao Tatta
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya (PO), Kasaragod, Kerala 671320, India
| | - Sayan Paul
- Department of Biochemistry & Molecular Biology, the University of Texas Medical Branch at Galveston, Galveston, Texas 77555, USA
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya (PO), Kasaragod, Kerala 671320, India; Department of Biotechnology, School of Life Sciences, Pondicherry University, Puducherry 605014, India.
| |
Collapse
|
12
|
van Praagh J, Havenga K. What Is the Microbiome? A Description of a Social Network. Clin Colon Rectal Surg 2023; 36:91-97. [PMID: 36844706 PMCID: PMC9946720 DOI: 10.1055/s-0043-1760863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
The gut microbiome has coevolved with its hosts over the years, forming a complex and symbiotic relationship. It is formed by what we do, what we eat, where we live, and with whom we live. The microbiome is known to influence our health by training our immune system and providing nutrients for the human body. However, when the microbiome becomes out of balance and dysbiosis occurs, the microorganisms within can cause or contribute to diseases. This major influencer on our health is studied intensively, but it is unfortunately often overlooked by the surgeon and in surgical practice. Because of that, there is not much literature about the microbiome and its influence on surgical patients or procedures. However, there is evidence that it plays a major role, showing that it needs to be a topic of interest for the surgeon. This review is written to show the surgeon the importance of the microbiome and why it should be taken into consideration when preparing or treating patients.
Collapse
Affiliation(s)
- J.B. van Praagh
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Klaas Havenga
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
In Vivo Role of Two-Component Regulatory Systems in Models of Urinary Tract Infections. Pathogens 2023; 12:pathogens12010119. [PMID: 36678467 PMCID: PMC9861413 DOI: 10.3390/pathogens12010119] [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: 11/24/2022] [Revised: 12/23/2022] [Accepted: 01/08/2023] [Indexed: 01/12/2023] Open
Abstract
Two-component signaling systems (TCSs) are finely regulated mechanisms by which bacteria adapt to environmental conditions by modifying the expression of target genes. In bacterial pathogenesis, TCSs play important roles in modulating adhesion to mucosal surfaces, resistance to antibiotics, and metabolic adaptation. In the context of urinary tract infections (UTI), one of the most common types infections causing significant health problems worldwide, uropathogens use TCSs for adaptation, survival, and establishment of pathogenicity. For example, uropathogens can exploit TCSs to survive inside bladder epithelial cells, sense osmolar variations in urine, promote their ascension along the urinary tract or even produce lytic enzymes resulting in exfoliation of the urothelium. Despite the usefulness of studying the function of TCSs in in vitro experimental models, it is of primary necessity to study bacterial gene regulation also in the context of host niches, each displaying its own biological, chemical, and physical features. In light of this, the aim of this review is to provide a concise description of several bacterial TCSs, whose activity has been described in mouse models of UTI.
Collapse
|
14
|
Parga A, Manoil D, Brundin M, Otero A, Belibasakis GN. Gram-negative quorum sensing signalling enhances biofilm formation and virulence traits in gram-positive pathogen Enterococcus faecalis. J Oral Microbiol 2023; 15:2208901. [PMID: 37187675 PMCID: PMC10177678 DOI: 10.1080/20002297.2023.2208901] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
Acyl-homoserine lactones (AHLs) are typical quorum-sensing molecules of gram-negative bacteria. Recent evidence suggests that AHLs may also affect gram-positives, although knowledge of these interactions remains scarce. Here, we assessed the effect of AHLs on biofilm formation and transcriptional regulations in the gram-positive Enterococcus faecalis. Five E. faecalis strains were investigated herein. Crystal violet was employed to quantify the biomass formed, and confocal microscopy in combination with SYTO9/PI allowed the visualisation of biofilms' structure. The differential expression of 10 genes involved in quorum-sensing, biofilm formation and stress responses was evaluated using reverse-transcription-qPCR. The AHL exposure significantly increased biofilm production in strain ATCC 29212 and two isolates from infected dental roots, UmID4 and UmID5. In strains ATCC 29212 and UmID7, AHLs up-regulated the quorum-sensing genes (fsrC, cylA), the adhesins ace, efaA and asa1, together with the glycosyltransferase epaQ. In strain UmID7, AHL exposure additionally up-regulated two membrane-stress response genes (σV, groEL) associated with increased stress-tolerance and virulence. Altogether, our results demonstrate that AHLs promote biofilm formation and up-regulate a transcriptional network involved in virulence and stress tolerance in several E. faecalis strains. These data provide yet-unreported insights into E. faecalis biofilm responses to AHLs, a family of molecules long-considered the monopole of gram-negative signalling.
Collapse
Affiliation(s)
- Ana Parga
- Department of Microbiology and Parasitology, CIBUS-Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Daniel Manoil
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
- Division of cariology and endodontics, University Clinics of Dental Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- CONTACT Daniel Manoil Division of cariology and endodontics, University Clinics of Dental Medicine, Michel-Servet 1, Geneva1205, Switzerland
| | - Malin Brundin
- Division of Endodontics, Department of Odontology, Umeå University, Umeå, Sweden
| | - Ana Otero
- Department of Microbiology and Parasitology, CIBUS-Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Georgios N. Belibasakis
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
15
|
Liu Y, Li B, Wei Y. New understanding of gut microbiota and colorectal anastomosis leak: A collaborative review of the current concepts. Front Cell Infect Microbiol 2022; 12:1022603. [PMID: 36389160 PMCID: PMC9663802 DOI: 10.3389/fcimb.2022.1022603] [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: 08/18/2022] [Accepted: 10/06/2022] [Indexed: 01/24/2023] Open
Abstract
Anastomotic leak (AL) is a life-threatening postoperative complication following colorectal surgery, which has not decreased over time. Until now, no specific risk factors or surgical technique could be targeted to improve anastomotic healing. In the past decade, gut microbiota dysbiosis has been recognized to contribute to AL, but the exact effects are still vague. In this context, interpretation of the mechanisms underlying how the gut microbiota contributes to AL is significant for improving patients' outcomes. This review concentrates on novel findings to explain how the gut microbiota of patients with AL are altered, how the AL-specific pathogen colonizes and is enriched on the anastomosis site, and how these pathogens conduct their tissue breakdown effects. We build up a framework between the gut microbiota and AL on three levels. Firstly, factors that shape the gut microbiota profiles in patients who developed AL after colorectal surgery include preoperative intervention and surgical factors. Secondly, AL-specific pathogenic or collagenase bacteria adhere to the intestinal mucosa and defend against host clearance, including the interaction between bacterial adhesion and host extracellular matrix (ECM), the biofilm formation, and the weakened host commercial bacterial resistance. Thirdly, we interpret the potential mechanisms of pathogen-induced poor anastomotic healing.
Collapse
Affiliation(s)
- Yang Liu
- Pancreatic and Gastrointestinal Surgery Division, HwaMei Hospital, University of Chinese Academy of Science, Ningbo, China,Ningbo Clinical Research Center for Digestive System Tumors, Ningbo, China
| | - Bowen Li
- Pancreatic and Gastrointestinal Surgery Division, HwaMei Hospital, University of Chinese Academy of Science, Ningbo, China,Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunwei Wei
- Pancreatic and Gastrointestinal Surgery Division, HwaMei Hospital, University of Chinese Academy of Science, Ningbo, China,Ningbo Clinical Research Center for Digestive System Tumors, Ningbo, China,*Correspondence: Yunwei Wei,
| |
Collapse
|
16
|
Venkateswaran P, Lakshmanan PM, Muthukrishnan S, Bhagavathi H, Vasudevan S, Neelakantan P, Solomon AP. Hidden agenda of Enterococcus faecalis lifestyle transition: planktonic to sessile state. Future Microbiol 2022; 17:1051-1069. [PMID: 35899477 DOI: 10.2217/fmb-2021-0212] [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: 11/21/2022] Open
Abstract
Enterococcus faecalis, a human gastrointestinal tract commensal, is known to cause nosocomial infections. Interestingly, the pathogen's host colonization and persistent infections are possibly linked to its lifestyle changes from planktonic to sessile state. Also, the multidrug resistance and survival fitness acquired in the sessile stage of E. faecalis has challenged treatment regimes. This situation exists because of the critical role played by several root genes and their molecular branches, which are part of quorum sensing, aggregation substance, surface adhesions, stress-related response and sex pheromones in the sessile state. It is therefore imperative to decode the hidden agenda of E. faecalis and understand the significant factors influencing biofilm formation. This would, in turn, augment the development of novel strategies to tackle E. faecalis infections.
Collapse
Affiliation(s)
- Parvathy Venkateswaran
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Priya M Lakshmanan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Sudhiksha Muthukrishnan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Hema Bhagavathi
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Sahana Vasudevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | | | - Adline P Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| |
Collapse
|
17
|
Enterococcus Virulence and Resistant Traits Associated with Its Permanence in the Hospital Environment. Antibiotics (Basel) 2022; 11:antibiotics11070857. [PMID: 35884110 PMCID: PMC9311936 DOI: 10.3390/antibiotics11070857] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Enterococcus are opportunistic pathogens that have been gaining importance in the clinical setting, especially in terms of hospital-acquired infections. This problem has mainly been associated with the fact that these bacteria are able to present intrinsic and extrinsic resistance to different classes of antibiotics, with a great deal of importance being attributed to vancomycin-resistant enterococci. However, other aspects, such as the expression of different virulence factors including biofilm-forming ability, and its capacity of trading genetic information, makes this bacterial genus more capable of surviving harsh environmental conditions. All these characteristics, associated with some reports of decreased susceptibility to some biocides, all described in this literary review, allow enterococci to present a longer survival ability in the hospital environment, consequently giving them more opportunities to disseminate in these settings and be responsible for difficult-to-treat infections.
Collapse
|
18
|
Antibiotic resistance and virulence factors in lactobacilli: something to carefully consider. Food Microbiol 2022; 103:103934. [DOI: 10.1016/j.fm.2021.103934] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 10/25/2021] [Accepted: 11/04/2021] [Indexed: 01/06/2023]
|
19
|
Du Q, Yuan S, Zhao S, Fu D, Chen Y, Zhou Y, Cao Y, Gao Y, Xu X, Zhou X, He J. Coexistence of Candida albicans and Enterococcus faecalis increases biofilm virulence and periapical lesions in rats. BIOFOULING 2021; 37:964-974. [PMID: 34839774 DOI: 10.1080/08927014.2021.1993836] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The present study utilized an in vitro dual-species biofilm model and an in vivo rat post-treatment endodontic disease (PTED) model to investigate whether co-infection of Candida albicans and Enterococcus faecalis would aggravate periapical lesions. The results showed that co-culturing yielded a thicker and denser biofilm more tolerant to detrimental stresses compared with the mono-species biofilm, such as a starvation-alkalinity environment, mechanical shear force and bactericidal chemicals. Consistently, co-inoculation of E. faecalis and C. albicans significantly increased the extent of in vivo periapical lesions compared with mono-species infection. Specifically, coexistence of both microorganisms increased osteoclastic bone resorption and suppressed osteoblastic bone formation. The synergistic effects also up-regulated inflammatory cytokines including TNF-α and IL-6. In summary, coexistence of C. albicans and E. faecalis increased periapical lesions by enhanced biofilm virulence.
Collapse
Affiliation(s)
- Qian Du
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Shasha Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shuangyuan Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Di Fu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Yifei Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Yuan Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yangpei Cao
- Woody L. Hunt School of Dental Medicine, Texas Tech University Health Sciences Center, EI Paso, TX, USA
| | - Yuan Gao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jinzhi He
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Rezende-Pereira G, Albuquerque JP, Souza MC, Nogueira BA, Silva MG, Hirata R, Mattos-Guaraldi AL, Duarte RS, Neves FPG. Biofilm Formation on Breast Implant Surfaces by Major Gram-Positive Bacterial Pathogens. Aesthet Surg J 2021; 41:1144-1151. [PMID: 33378420 DOI: 10.1093/asj/sjaa416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Bacterial biofilm on surfaces of mammary implants is a predisposing factor for several outcomes. Because Gram-positive bacteria are potential agents of biomaterial-associated infections (BAIs), their abilities to form biofilm on breast implants should be elucidated. OBJECTIVES The aim of this study was to evaluate biofilm formation on different mammary prosthesis surfaces by major Gram-positive bacterial pathogens involved in BAIs. METHODS We initially evaluated biofilm formation on polystyrene plates with and without fibrinogen or collagen for 1 reference strain and 1 clinical isolate of Enterococcus faecalis, Staphylococcus aureus, Staphylococcus epidermidis, and Streptococcus pyogenes. We also tested the ability of clinical isolates to form biofilm on 4 different implant surfaces: polyurethane foam and smooth, microtextured, and standard textured silicone. Biofilm structure and cell viability were observed by scanning electron microscopy and confocal laser scanning microscopy. RESULTS All strains showed strong biofilm formation on polystyrene. After fibrinogen or collagen treatment, biofilm formation varied. With fibrinogen, reference strains of S. aureus and S. pyogenes increased biofilm formation (P < 0.05). Reference strains of all species and the clinical isolate of S. pyogenes increased biofilm formation after collagen treatment (P < 0.05). In general, S. aureus showed higher capacity to produce biofilm. Scanning electron microscopy showed that biofilm attached to all surfaces tested, with the presence of extracellular polymeric substances and voids. Viable cells were more frequent for E. faecalis and S. pyogenes. CONCLUSIONS All species produced biofilm on all prosthesis surfaces and under different conditions. Micrographies indicated thicker bacterial biofilm formation on microtextured and/or standard textured silicone by all species, except E. faecalis.
Collapse
Affiliation(s)
| | | | - Monica C Souza
- Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Barbara A Nogueira
- Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marlei G Silva
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Raphael Hirata
- Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana L Mattos-Guaraldi
- Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Rafael S Duarte
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Felipe P G Neves
- Instituto Biomédico, Universidade Federal Fluminense, Niterói, RJ, Brazil
| |
Collapse
|
21
|
Chilambi GS, Nordstrom HR, Evans DR, Kowalski RP, Dhaliwal DK, Jhanji V, Shanks RMQ, Van Tyne D. Genomic and phenotypic diversity of Enterococcus faecalis isolated from endophthalmitis. PLoS One 2021; 16:e0250084. [PMID: 33852628 PMCID: PMC8046195 DOI: 10.1371/journal.pone.0250084] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/30/2021] [Indexed: 11/30/2022] Open
Abstract
Enterococcus faecalis are hospital-associated opportunistic pathogens and also causative agents of post-operative endophthalmitis. Patients with enterococcal endophthalmitis often have poor visual outcomes, despite appropriate antibiotic therapy. Here we investigated the genomic and phenotypic characteristics of E. faecalis isolates collected from 13 patients treated at the University of Pittsburgh Medical Center Eye Center over 19 years. Comparative genomic analysis indicated that patients were infected with E. faecalis belonging to diverse multi-locus sequence types (STs) and resembled E. faecalis sampled from clinical, commensal, and environmental sources. We identified known E. faecalis virulence factors and antibiotic resistance genes in each genome, including genes conferring resistance to aminoglycosides, erythromycin, and tetracyclines. We assessed all isolates for their cytolysin production, biofilm formation, and antibiotic susceptibility, and observed phenotypic differences between isolates. Fluoroquinolone and cephalosporin susceptibilities were particularly variable between isolates, as were biofilm formation and cytolysin production. In addition, we found evidence of E. faecalis adaptation during recurrent endophthalmitis by identifying genetic variants that arose in sequential isolates sampled over eight months from the same patient. We identified a mutation in the DNA mismatch repair gene mutS that was associated with an increased rate of spontaneous mutation in the final isolate from the patient. Overall this study documents the genomic and phenotypic variability among E. faecalis causing endophthalmitis, as well as possible adaptive mechanisms underlying bacterial persistence during recurrent ocular infection.
Collapse
Affiliation(s)
- Gayatri Shankar Chilambi
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Hayley R. Nordstrom
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Daniel R. Evans
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Regis P. Kowalski
- The Charles T. Campbell Ophthalmic Microbiology Laboratory, UPMC Eye Center, Ophthalmology and Visual Sciences Research Center, The Eye and Ear Institute, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Deepinder K. Dhaliwal
- The Charles T. Campbell Ophthalmic Microbiology Laboratory, UPMC Eye Center, Ophthalmology and Visual Sciences Research Center, The Eye and Ear Institute, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Vishal Jhanji
- The Charles T. Campbell Ophthalmic Microbiology Laboratory, UPMC Eye Center, Ophthalmology and Visual Sciences Research Center, The Eye and Ear Institute, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Robert M. Q. Shanks
- The Charles T. Campbell Ophthalmic Microbiology Laboratory, UPMC Eye Center, Ophthalmology and Visual Sciences Research Center, The Eye and Ear Institute, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Daria Van Tyne
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
22
|
Arora S, Gordon J, Hook M. Collagen Binding Proteins of Gram-Positive Pathogens. Front Microbiol 2021; 12:628798. [PMID: 33613497 PMCID: PMC7893114 DOI: 10.3389/fmicb.2021.628798] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Collagens are the primary structural components of mammalian extracellular matrices. In addition, collagens regulate tissue development, regeneration and host defense through interaction with specific cellular receptors. Their unique triple helix structure, which requires a glycine residue every third amino acid, is the defining structural feature of collagens. There are 28 genetically distinct collagens in humans. In addition, several other unrelated human proteins contain a collagen domain. Gram-positive bacteria of the genera Staphylococcus, Streptococcus, Enterococcus, and Bacillus express cell surface proteins that bind to collagen. These proteins of Gram-positive pathogens are modular proteins that can be classified into different structural families. This review will focus on the different structural families of collagen binding proteins of Gram-positive pathogen. We will describe how these proteins interact with the triple helix in collagens and other host proteins containing a collagenous domain and discuss how these interactions can contribute to the pathogenic processes.
Collapse
Affiliation(s)
- Srishtee Arora
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, United States
| | - Jay Gordon
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, United States
| | - Magnus Hook
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, United States
| |
Collapse
|
23
|
Suryaletha K, Chandrika SK, Thomas S. Comprehensive genomics depict accessory genes encoding pathogenicity and biofilm determinants in Enterococcus faecalis. Future Microbiol 2021; 16:175-184. [PMID: 33528277 DOI: 10.2217/fmb-2020-0111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: Enterococcus faecalis is a leading nosocomial pathogen in biofilm-associated polymicrobial infections. The study aims to understand pathogenicity and biofilm determinants of the pathogen by genome analysis. Methodology: Genome sequencing of a strong biofilm forming clinical isolate Enterococcus faecalis SK460 devoid of Fsr quorum-signaling system, was performed and comparative genomics was carried out among a set of pathogenic biofilm formers and nonpathogenic weak biofilm formers. Results: Analysis revealed a pool of virulence and adhesion related factors associated with pathogenicity. Absence of CRISPR-Cas system facilitated acquisition of pheromone responsive plasmid, pathogenicity island and phages. Comprehensive analysis identified a subset of accessory genes encoding polysaccharide lyase, sugar phosphotransferase system, phage proteins and transcriptional regulators exclusively in pathogenic biofilm formers. Conclusion: The study identified a set of genes specific to pathogenic biofilm formers and these can act as targets which in turn help to develop future treatment endeavors against enterococcal infections.
Collapse
Affiliation(s)
- Karthika Suryaletha
- Cholera & Biofilm Research Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695 014, Kerala, India
| | - Sivakumar K Chandrika
- Distributed Information Sub-Centre, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695 014, Kerala, India
| | - Sabu Thomas
- Cholera & Biofilm Research Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695 014, Kerala, India
| |
Collapse
|
24
|
Gholizadeh P, Aghazadeh M, Ghotaslou R, Ahangarzadeh Rezaee M, Pirzadeh T, Köse Ş, Ganbarov K, Yousefi M, Kafil HS. CRISPR- cas system in the acquisition of virulence genes in dental-root canal and hospital-acquired isolates of Enterococcus faecalis. Virulence 2020; 11:1257-1267. [PMID: 32930628 PMCID: PMC7549939 DOI: 10.1080/21505594.2020.1809329] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/23/2020] [Accepted: 08/08/2020] [Indexed: 01/20/2023] Open
Abstract
Enterococcus faecalis is one of the important causative agents of nosocomial and life-threatening infections in human. Several studies have demonstrated that the presence of CRISPR-cas is associated with antibiotic susceptibility and lack of virulence traits. In this study, we aimed to assess the phenotypic and genotypic virulence determinants in relation to CRISPR elements from the dental-root canals and hospital-acquired isolates of E. faecalis. Eighty-eight hospital-acquired and 73 dental-root canal isolates of E. faecalis were assessed in this study. Phenotypic screening of the isolates included biofilm formation, and gelatinase and hemolysis activities. Genotypical screening using PCR was further used to evaluate the presence of CRISPR elements and different virulence-associated genes such as efaA, esp, cylA, hyl, gelE, ace, ebpR, and asa1. Biofilm formation, gelatinase, and hemolysis activities were detected in 93.8%, 29.2%, and 19.2% of the isolates, respectively. The most prevalent virulence-associated gene was ace, which was followed by efaA, whereas cylA was the least identified. The presence of CRISPR1-cas, orphan CRISPR2, and CRISPR3-cas was determined in 13%, 55.3%, and 17.4% of the isolates, respectively. CRISPR elements were significantly more prevalent in the dental-root canal isolates. An inverse significant correlation was found between CRISPR-cas loci, esp, and gelE, while direct correlations were observed in the case of cylA, hyl, gelE (among CRISPR-loci 1 and 3), asa1, ace, biofilm formation, and hemolysis activity. Findings, therefore, indicate that CRISPR-cas might prevent the acquisition of some respective pathogenicity factors in some isolates, though not all; so selective forces could not influence pathogenic traits. Abbreviations: BHI: brain-heart infusion agar; CRISPRs: Clustered regularly interspaced short palindromic repeats; Esp: Cell wall-associated protein; ENT: ear-nose-throat; ICU: intensive care units; OD: optical densities; PCR: polymerase chain reaction; SDS: sodium dodecyl sulfate; UTI: urinary tract infection.
Collapse
Affiliation(s)
- Pourya Gholizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, I.R. Iran
| | - Mohammad Aghazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, I.R. Iran
| | - Reza Ghotaslou
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, I.R. Iran
| | | | - Tahereh Pirzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, I.R. Iran
| | - Şükran Köse
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Tepecik Training and Research Hospital, İzmir, Turkey
| | | | - Mehdi Yousefi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, I.R. Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, I.R. Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, I.R. Iran
| |
Collapse
|
25
|
Ghalavand Z, Alebouyeh M, Ghanati K, Azimi L, Rashidan M. Genetic relatedness of the Enterococcus faecalis isolates in stool and urine samples of patients with community-acquired urinary tract infection. Gut Pathog 2020; 12:42. [PMID: 32944085 PMCID: PMC7488108 DOI: 10.1186/s13099-020-00380-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/01/2020] [Indexed: 11/10/2022] Open
Abstract
Background Community-acquired urinary tract infection (CA-UTI) could be caused by endogenous or exogenous routes. To show this relationship, we investigated molecular fingerprints and genotypes of paired Enterococcus faecalis isolated from the urine of symptomatic patients and their fecal samples. Results Out of the studied patients, 63 pairs of E. faecalis isolates were obtained simultaneously from their urine and feces samples. All the strains were sensitive to vancomycin, linezolid, nitrofurantoin, and daptomycin (MIC value: ≤ 4 µg/ml), while resistance to tetracycline (urine: 88.9%; stool: 76.2%) and minocycline (urine: 87.3%, stool: 71.4%) was detected in most of them. The most common detected virulence genes were included efbA, ace, and gelE. RAPD-PCR and PFGE analyses showed the same patterns of molecular fingerprints between paired of the isolates in 26.9% and 15.8% of the patients, respectively. Conclusions Similarity of E. faecalis strains between the urine and feces samples confirmed the occurrence of endogenous infection via contamination with colonized bacteria in the intestinal tract. Carriage of a complete virulence genotype in the responsible strains was statistically in correlation with endogenous UTI, which shows their possible involvement in pathogenicity of uropathogenic E. faecalis strains.
Collapse
Affiliation(s)
- Zohreh Ghalavand
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Alebouyeh
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kiandokht Ghanati
- National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences and Health Services, Tehran, Iran
| | - Leila Azimi
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Rashidan
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Xiao B, Zou Z, Bhandari J, Zhang Y, Yan G. Exposure to diode laser (810nm) affects the bacterial adherence and biofilm formation in a E. faecalis biofilm model. Photodiagnosis Photodyn Ther 2020; 31:101772. [DOI: 10.1016/j.pdpdt.2020.101772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/14/2020] [Accepted: 04/02/2020] [Indexed: 10/24/2022]
|
27
|
Involvement of Chromosomally Encoded Homologs of the RRNPP Protein Family in Enterococcus faecalis Biofilm Formation and Urinary Tract Infection Pathogenesis. J Bacteriol 2020; 202:JB.00063-20. [PMID: 32540933 DOI: 10.1128/jb.00063-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/02/2020] [Indexed: 11/20/2022] Open
Abstract
Enterococcus faecalis is an opportunistic pathogen capable of causing infections, including endocarditis and urinary tract infections (UTI). One of the well-characterized quorum-sensing pathways in E. faecalis involves coordination of the conjugal transfer of pheromone-responsive plasmids by PrgX, a member of the RRNPP protein family. Members of this protein family in various Firmicutes have also been shown to contribute to numerous cellular processes, including sporulation, competence, conjugation, nutrient sensing, biofilm formation, and virulence. As PrgX is a plasmid-encoded RRNPP family member, we surveyed the genome of the multidrug-resistant strain V583 for additional RRNPP homologs using computational searches and refined those identified hits for predicted structural similarities to known RRNPP family members. This led us to investigate the contribution of the chromosomally encoded RRNPP homologs to biofilm processes and pathogenesis in a catheter-associated urinary tract infection (CAUTI) model. In this study, we identified five such homologs and report that 3 of the 5 homologs, EF0073, EF1599, and EF1316, affect biofilm formation as well as outcomes in the CAUTI model.IMPORTANCE Enterococcus faecalis causes health care-associated infections and displays resistance to a variety of broad-spectrum antibiotics by acquisition of resistance traits as well as the ability to form biofilms. Even though a growing number of factors related to biofilm formation have been identified, mechanisms that contribute to biofilm formation are still largely unknown. Members of the RRNPP protein family regulate a diverse set of biological reactions in low-G+C Gram-positive bacteria (Firmicutes). Here, we identify three predicted structural homologs of the RRNPP family, EF0073, EF1599, and EF1316, which affect biofilm formation and CAUTI pathogenesis.
Collapse
|
28
|
Brown AO, Singh KV, Cruz MR, Kaval KG, Francisco LE, Murray BE, Garsin DA. Cardiac Microlesions Form During Severe Bacteremic Enterococcus faecalis Infection. J Infect Dis 2020; 223:508-516. [PMID: 32597945 DOI: 10.1093/infdis/jiaa371] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/22/2020] [Indexed: 12/22/2022] Open
Abstract
Enterococcus faecalis is a significant cause of hospital-acquired bacteremia. Herein, the discovery is reported that cardiac microlesions form during severe bacteremic E. faecalis infection in mice. The cardiac microlesions were identical in appearance to those formed by Streptococcus pneumoniae during invasive pneumococcal disease. However, E. faecalis does not encode the virulence determinants implicated in pneumococcal microlesion formation. Rather, disulfide bond forming protein A (DsbA) was found to be required for E. faecalis virulence in a Caenorhabditis elegans model and was necessary for efficient cardiac microlesion formation. Furthermore, E. faecalis promoted cardiomyocyte apoptotic and necroptotic cell death at sites of microlesion formation. Additionally, loss of DsbA caused an increase in proinflammatory cytokines, unlike the wild-type strain, which suppressed the immune response. In conclusion, we establish that E. faecalis is capable of forming cardiac microlesions and identify features of both the bacterium and the host response that are mechanistically involved.
Collapse
Affiliation(s)
- Armand O Brown
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kavindra V Singh
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Melissa R Cruz
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Karan Gautam Kaval
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Liezl E Francisco
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Barbara E Murray
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center at Houston, Houston, Texas, USA.,Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Danielle A Garsin
- Department of Microbiology and Molecular Genetics, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
29
|
Tan CAZ, Antypas H, Kline KA. Overcoming the challenge of establishing biofilms in vivo: a roadmap for Enterococci. Curr Opin Microbiol 2020; 53:9-18. [PMID: 32062025 DOI: 10.1016/j.mib.2020.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 12/28/2022]
Abstract
Enterococcus faecalis forms single and mixed-species biofilms on both tissue and medical devices in the host, often under exposure to fluid flow, giving rise to infections that are recalcitrant to treatment. The factors that drive enterococcal biofilm formation in the host, however, remain unclear. Recent reports in other pathogens show how surface sensing by bacteria can trigger the transition from planktonic to sessile lifestyle. Fluid flow can enhance initial adhesion, but also influence quorum sensing. Biofilm-specific factors, as well as biofilm size and extracellular polymeric substances, can compromise opsonization and phagocytosis. Bacterial interspecies synergy can create favorable conditions in the host for biofilm formation. Through these concepts, we define the knowledge gaps in understanding host-associated E. faecalis biofilm formation and propose a roadmap for future investigations.
Collapse
Affiliation(s)
- Casandra Ai Zhu Tan
- Singapore Centre for Environmental Life Sciences Engineering, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Haris Antypas
- Singapore Centre for Environmental Life Sciences Engineering, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Kimberly A Kline
- Singapore Centre for Environmental Life Sciences Engineering, School of Biological Sciences, Nanyang Technological University, Singapore.
| |
Collapse
|
30
|
Orihuel A, Terán L, Renaut J, Planchon S, Valacco MP, Masias E, Minahk C, Vignolo G, Moreno S, De Almeida AM, Saavedra L, Fadda S. Physiological and proteomic response of Escherichia coli O157:H7 to a bioprotective lactic acid bacterium in a meat environment. Food Res Int 2019; 125:108622. [PMID: 31554055 DOI: 10.1016/j.foodres.2019.108622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/12/2019] [Accepted: 08/15/2019] [Indexed: 01/21/2023]
Abstract
The enterohemorrhagic Escherichia (E.) coli (EHEC) is a pathogen of great concern for public health and the meat industry all over the world. The high economic losses in meat industry and the high costs of the illness highlight the necessity of additional efforts to control this pathogen. Previous studies have demonstrated the inhibitory activity of Enterococcus mundtii CRL35 towards EHEC, showing a specific proteomic response during the co-culture. In the present work, additional studies of the EHEC-Ent. mundtii interaction were carried out: i) differential protein expression of E. coli O157:H7 NCTC12900 growing in co-culture with Ent. mundtii in a meat environment, ii) the reciprocal influence between these two microorganisms in the adhesion to extracellular matrix (ECM) proteins and iii) the possible induction of the phage W933, coding for Shiga toxin (Stx1), by Ent. mundtii CRL35. Proteomic analysis showed a significant repression of a number of E. coli NCTC12900 proteins in co-culture respect to its single culture, these mostly related to the metabolism and transport of amino acids and nucleotides. On the other hand, statistically significant overexpression of EHEC proteins involved in stress, energy production, amino acid metabolism and transcription was observed at 30 h respect to 6 h when EHEC grew in co-culture. Data are available via ProteomeXchange with identifier PXD014588. Besides, EHEC showed a decreased adhesion capacity to ECM proteins in the presence of the bioprotective strain. Finally, Ent. mundtii CRL35 did not induce the lytic cycle of W933 bacteriophage, thus indicating its potential safe use for eliminating this pathogen. Overall, this study expands the knowledge of EHEC- Ent. mundtii CRL35 interaction in a meat environment, which will certainly contribute to find out effective biological strategies to eliminate this pathogen.
Collapse
Affiliation(s)
- Alejandra Orihuel
- Technology, Centro de Referencia para Lactobacilos, Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucumán, Tucumán, Argentina
| | - Lucrecia Terán
- Genetics, Centro de Referencia para Lactobacilos, Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucumán, Tucumán, Argentina
| | - Jenny Renaut
- LIST - Luxembourg Institute of Science and Technology "Environmental Research and Innovation" (ERIN) Department, Belvaux, Luxembourg
| | - Sébastien Planchon
- LIST - Luxembourg Institute of Science and Technology "Environmental Research and Innovation" (ERIN) Department, Belvaux, Luxembourg
| | - María Pía Valacco
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Emilse Masias
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT. Chacabuco 461, T4000ILI - San Miguel de Tucumán, Argentina
| | - Carlos Minahk
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT. Chacabuco 461, T4000ILI - San Miguel de Tucumán, Argentina
| | - Graciela Vignolo
- Technology, Centro de Referencia para Lactobacilos, Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucumán, Tucumán, Argentina
| | - Silvia Moreno
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - André M De Almeida
- LEAF - Linking Landscape, Environment, Agriculture and Food, Instituto Superior de Agronomia, University of Lisbon, Lisbon, Portugal
| | - Lucila Saavedra
- Genetics, Centro de Referencia para Lactobacilos, Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucumán, Tucumán, Argentina
| | - Silvina Fadda
- Technology, Centro de Referencia para Lactobacilos, Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucumán, Tucumán, Argentina.
| |
Collapse
|
31
|
Ali IAA, Cheung BPK, Yau JYY, Matinlinna JP, Lévesque CM, Belibasakis GN, Neelakantan P. The influence of substrate surface conditioning and biofilm age on the composition of
Enterococcus faecalis
biofilms. Int Endod J 2019; 53:53-61. [DOI: 10.1111/iej.13202] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 08/09/2019] [Indexed: 02/06/2023]
Affiliation(s)
- I. A. A. Ali
- Faculty of Dentistry The University of Hong Kong Hong Kong SAR
| | - B. P. K. Cheung
- Faculty of Dentistry The University of Hong Kong Hong Kong SAR
| | - J. Y. Y. Yau
- Faculty of Dentistry The University of Hong Kong Hong Kong SAR
| | | | - C. M. Lévesque
- Faculty of Dentistry University of Toronto Toronto ON Canada
| | - G. N. Belibasakis
- Division of Oral Diseases Department of Dental Medicine Karolinska Institute Huddinge Sweden
| | - P. Neelakantan
- Faculty of Dentistry The University of Hong Kong Hong Kong SAR
| |
Collapse
|
32
|
Adaptation to Adversity: the Intermingling of Stress Tolerance and Pathogenesis in Enterococci. Microbiol Mol Biol Rev 2019; 83:83/3/e00008-19. [PMID: 31315902 DOI: 10.1128/mmbr.00008-19] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Enterococcus is a diverse and rugged genus colonizing the gastrointestinal tract of humans and numerous hosts across the animal kingdom. Enterococci are also a leading cause of multidrug-resistant hospital-acquired infections. In each of these settings, enterococci must contend with changing biophysical landscapes and innate immune responses in order to successfully colonize and transit between hosts. Therefore, it appears that the intrinsic durability that evolved to make enterococci optimally competitive in the host gastrointestinal tract also ideally positioned them to persist in hospitals, despite disinfection protocols, and acquire new antibiotic resistances from other microbes. Here, we discuss the molecular mechanisms and regulation employed by enterococci to tolerate diverse stressors and highlight the role of stress tolerance in the biology of this medically relevant genus.
Collapse
|
33
|
Fiore E, Van Tyne D, Gilmore MS. Pathogenicity of Enterococci. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0053-2018. [PMID: 31298205 PMCID: PMC6629438 DOI: 10.1128/microbiolspec.gpp3-0053-2018] [Citation(s) in RCA: 273] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Indexed: 12/19/2022] Open
Abstract
Enterococci are unusually well adapted for survival and persistence in a variety of adverse environments, including on inanimate surfaces in the hospital environment and at sites of infection. This intrinsic ruggedness undoubtedly played a role in providing opportunities for enterococci to interact with other overtly drug-resistant microbes and acquire additional resistances on mobile elements. The rapid rise of antimicrobial resistance among hospital-adapted enterococci has rendered hospital-acquired infections a leading therapeutic challenge. With about a quarter of a genome of additional DNA conveyed by mobile elements, there are undoubtedly many more properties that have been acquired that help enterococci persist and spread in the hospital setting and cause diseases that have yet to be defined. Much remains to be learned about these ancient and rugged microbes, particularly in the area of pathogenic mechanisms involved with human diseases.
Collapse
Affiliation(s)
- Elizabeth Fiore
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| | - Daria Van Tyne
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| | - Michael S Gilmore
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
34
|
Sortase-Dependent Proteins Promote Gastrointestinal Colonization by Enterococci. Infect Immun 2019; 87:IAI.00853-18. [PMID: 30804098 DOI: 10.1128/iai.00853-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/18/2019] [Indexed: 12/19/2022] Open
Abstract
The human gastrointestinal tract (GIT) is inhabited by a dense microbial community of symbionts. Enterococci are among the earliest members of this community and remain core members of the GIT microbiota throughout life. Enterococci have also recently emerged as opportunistic pathogens and major causes of nosocomial infections. Although recognized as a prerequisite for infection, colonization of the GIT by enterococci remains poorly understood. One way that bacteria adapt to dynamic ecosystems like the GIT is through the use of their surface proteins to sense and interact with components of their immediate environment. In Gram-positive bacteria, a subset of surface proteins relies on an enzyme called sortase for covalent attachment to the cell wall. Here, we show that the housekeeping sortase A (SrtA) enzyme promotes intestinal colonization by enterococci. Furthermore, we show that the enzymatic activity of SrtA is key to the ability of Enterococcus faecalis to bind mucin (a major component of the GIT mucus). We also report the GIT colonization phenotypes of E. faecalis mutants lacking selected sortase-dependent proteins (SDPs). Further examination of the mucin binding ability of these mutants suggests that adhesion to mucin contributes to intestinal colonization by E. faecalis.
Collapse
|
35
|
Bolocan AS, Upadrasta A, Bettio PHDA, Clooney AG, Draper LA, Ross RP, Hill C. Evaluation of Phage Therapy in the Context of Enterococcus faecalis and Its Associated Diseases. Viruses 2019; 11:E366. [PMID: 31010053 PMCID: PMC6521178 DOI: 10.3390/v11040366] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/11/2022] Open
Abstract
Bacteriophages (phages) or bacterial viruses have been proposed as natural antimicrobial agents to fight against antibiotic-resistant bacteria associated with human infections. Enterococcus faecalis is a gut commensal, which is occasionally found in the mouth and vaginal tract, and does not usually cause clinical problems. However, it can spread to other areas of the body and cause life-threatening infections, such as septicemia, endocarditis, or meningitis, in immunocompromised hosts. Although E. faecalis phage cocktails are not commercially available within the EU or USA, there is an accumulated evidence from in vitro and in vivo studies that have shown phage efficacy, which supports the idea of applying phage therapy to overcome infections associated with E. faecalis. In this review, we discuss the potency of bacteriophages in controlling E. faecalis, in both in vitro and in vivo scenarios. E. faecalis associated bacteriophages were compared at the genome level and an attempt was made to categorize phages with respect to their suitability for therapeutic application, using orthocluster analysis. In addition, E. faecalis phages have been examined for the presence of antibiotic-resistant genes, to ensure their safe use in clinical conditions. Finally, the domain architecture of E. faecalis phage-encoded endolysins are discussed.
Collapse
Affiliation(s)
- Andrei S Bolocan
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| | - Aditya Upadrasta
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| | - Pedro H de Almeida Bettio
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| | - Adam G Clooney
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| | - Lorraine A Draper
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork P61 C996, Ireland.
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| |
Collapse
|
36
|
Igbinosa EO, Beshiru A. Antimicrobial Resistance, Virulence Determinants, and Biofilm Formation of Enterococcus Species From Ready-to-Eat Seafood. Front Microbiol 2019; 10:728. [PMID: 31057497 PMCID: PMC6482160 DOI: 10.3389/fmicb.2019.00728] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/25/2019] [Indexed: 12/22/2022] Open
Abstract
Enterococcus species form an important population of commensal bacteria and have been reported to possess numerous virulence factors considered significantly important in exacerbating diseases caused by them. The present study was designed to characterize antibiotic-resistant and virulent enterococci from ready-to-eat (RTE) seafood. A total of 720 RTE shrimp samples comprising sauced shrimp (n = 288), boiled shrimp (n = 216), and smoked shrimp (n = 216) obtained from open markets in Delta State, Nigeria, were assessed. Standard classical methods and polymerase chain reaction (PCR) were used in identifying the Enterococcus species. Potential virulence factors (β-hemolysis, gelatinase activity, S-layer, and biofilm formation) were assessed using standard procedures. The antibiotic susceptibility profile of the identified enterococci isolates was assayed using the Kirby–Bauer disc diffusion method. PCR was further used to screen selected antibiotic resistance and virulence genes. Prevalence of Enterococcus species from shrimp varieties is as follows: sauced, 26 (9.03%); boiled, 6 (2.78%); and smoked, 27 (12.50%), with an overall prevalence of 59 (8.19%) based on the occurrence of black hallow colonies after incubation. Enterococcus species detected include E. faecalis, 17 (28.8%); E. faecium, 29 (49.2%); E. gallinarum, 6 (10.2%); E. casseliflavus, 2 (3.4%); E. hirae, 3 (5.1%); and E. durans, 2 (3.4%). Biofilm occurrence among the shrimp varieties is as follows: 19/26 (73.1%) for sauced shrimps, 5/6 (83.3%) for boiled shrimps, and 16/27 (59.3%) for smoked shrimps. The phenotypic expression of the enterococci virulence revealed the following: S-layer, 59 (100%); gelatinase production, 19 (32.2%); and β-hemolysis, 21 (35.6%). An average of 3–11 virulence genes were detected in the Enterococcus species. The resistance profile of Enterococcus species is as follows: erythromycin, 29 (49.2%); vancomycin, 22 (37.3%); and tetracycline, 27 (45.8%). The frequency of occurrence of antibiotic resistance genes from the phenotypic resistant enterococci isolates to the macrolide, glycopeptide, and tetracycline antibiotics is as follows: ermA, 13/29 (44.8%); vanA, 14/22 (63.6%); tetA, 14/27 (51.9%); tetM, 15/27 (55.6%); ermB, 4/29 (13.8%); and vanB, 5/22 (22.7%). Findings from this study reveal the antibiotic resistance of enterococci strains of such species as E. durans, E. casseliflavus, E. gallinarum, and E. hirae. This study further revealed that RTE food products are reservoirs of potential virulent enterococci with antibiotic-resistant capabilities. This provides useful data for risk assessment and indicates that these foods may present a potential public health risk to consumers.
Collapse
Affiliation(s)
- Etinosa O Igbinosa
- Applied Microbial Processes and Environmental Health Research Group, Department of Microbiology, Faculty of Life Sciences, University of Benin, Benin City, Nigeria.,Sustainable Development Office, University of Benin, Benin City, Nigeria
| | - Abeni Beshiru
- Applied Microbial Processes and Environmental Health Research Group, Department of Microbiology, Faculty of Life Sciences, University of Benin, Benin City, Nigeria
| |
Collapse
|
37
|
Chong KKL, Tay WH, Janela B, Yong AMH, Liew TH, Madden L, Keogh D, Barkham TMS, Ginhoux F, Becker DL, Kline KA. Enterococcus faecalis Modulates Immune Activation and Slows Healing During Wound Infection. J Infect Dis 2019; 216:1644-1654. [PMID: 29045678 PMCID: PMC5854026 DOI: 10.1093/infdis/jix541] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 10/13/2017] [Indexed: 01/13/2023] Open
Abstract
Enterococcus faecalis is one of the most frequently isolated bacterial species in wounds yet little is known about its pathogenic mechanisms in this setting. Here, we used a mouse wound excisional model to characterize the infection dynamics of E faecalis and show that infected wounds result in 2 different states depending on the initial inoculum. Low-dose inocula were associated with short-term, low-titer colonization whereas high-dose inocula were associated with acute bacterial replication and long-term persistence. High-dose infection and persistence were also associated with immune cell infiltration, despite suppression of some inflammatory cytokines and delayed wound healing. During high-dose infection, the multiple peptide resistance factor, which is involved in resisting immune clearance, contributes to E faecalis fitness. These results comprehensively describe a mouse model for investigating E faecalis wound infection determinants, and suggest that both immune modulation and resistance contribute to persistent, nonhealing wounds.
Collapse
Affiliation(s)
- Kelvin Kian Long Chong
- Singapore Centre for Environmental Life Sciences Engineering, Singapore.,Nanyang Technological University Institute for Health Technologies, Singapore
| | - Wei Hong Tay
- Singapore Centre for Environmental Life Sciences Engineering, Singapore.,Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore
| | - Baptiste Janela
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Adeline Mei Hui Yong
- Singapore Centre for Environmental Life Sciences Engineering, Singapore.,School of Biological Sciences, Singapore
| | - Tze Horng Liew
- Singapore Centre for Environmental Life Sciences Engineering, Singapore
| | - Leigh Madden
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Damien Keogh
- Singapore Centre for Environmental Life Sciences Engineering, Singapore
| | | | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | | | - Kimberly A Kline
- Singapore Centre for Environmental Life Sciences Engineering, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| |
Collapse
|
38
|
Williams DL, Smith SR, Peterson BR, Allyn G, Cadenas L, Epperson RT, Looper RE. Growth substrate may influence biofilm susceptibility to antibiotics. PLoS One 2019; 14:e0206774. [PMID: 30870411 PMCID: PMC6417642 DOI: 10.1371/journal.pone.0206774] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/21/2019] [Indexed: 12/04/2022] Open
Abstract
The CDC biofilm reactor is a robust culture system with high reproducibility in which biofilms can be grown for a wide variety of analyses. Multiple material types are available as growth substrates, yet data from biofilms grown on biologically relevant materials is scarce, particularly for antibiotic efficacy against differentially supported biofilms. In this study, CDC reactor holders were modified to allow growth of biofilms on collagen, a biologically relevant substrate. Susceptibility to multiple antibiotics was compared between biofilms of varying species grown on collagen versus standard polycarbonate coupons. Data indicated that in 13/18 instances, biofilms on polycarbonate were more susceptible to antibiotics than those on collagen, suggesting that when grown on a complex substrate, biofilms may be more tolerant to antibiotics. These outcomes may influence the translatability of antibiotic susceptibility profiles that have been collected for biofilms on hard plastic materials. Data may also help to advance information on antibiotic susceptibility testing of biofilms grown on biologically relevant materials for future in vitro and in vivo applications.
Collapse
Affiliation(s)
- Dustin L. Williams
- George E. Wahlen Department of Veterans Affairs, Salt Lake City, UT, United States of America
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, United States of America
- Department of Pathology, University of Utah, Salt Lake City, UT, United States of America
- Department of Bioengineering, University of Utah, Salt Lake City, UT, United States of America
- Department of Physical Medicine and Rehabilitation, Uniformed Services University, Bethesda, MD, United States of America
- * E-mail:
| | - Scott R. Smith
- George E. Wahlen Department of Veterans Affairs, Salt Lake City, UT, United States of America
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, United States of America
| | - Brittany R. Peterson
- George E. Wahlen Department of Veterans Affairs, Salt Lake City, UT, United States of America
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, United States of America
| | - Gina Allyn
- George E. Wahlen Department of Veterans Affairs, Salt Lake City, UT, United States of America
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, United States of America
| | - Lousili Cadenas
- George E. Wahlen Department of Veterans Affairs, Salt Lake City, UT, United States of America
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, United States of America
| | - Richard Tyler Epperson
- George E. Wahlen Department of Veterans Affairs, Salt Lake City, UT, United States of America
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, United States of America
| | - Ryan E. Looper
- Department of Chemistry, University of Utah, Salt Lake City, UT, United States of America
| |
Collapse
|
39
|
Karimi A, Ghalavand Z, Fallah F, Eslami P, Parvin M, Alebouyeh M, Rashidan M. Prevalence of virulence determinants and antibiotic resistance patterns of Enterococcus faecalis strains in patients with community-acquired urinary tract infections in Iran. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2018; 28:599-608. [PMID: 30044128 DOI: 10.1080/09603123.2018.1497777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/03/2018] [Indexed: 06/08/2023]
Abstract
The aim of this study was to characterize virulence factors and antibiotic resistance patterns in E. faecalis strains obtained from community-acquired urinary tract infections. A total of 70 E. faecalis isolates from Labbafinejad Hospital in Tehran were collected. Antibiotic resistance and virulence determinants were examined by phenotypic and molecular methods. Among 70 E. faecalis isolates, efba (97.1%), ace (95.7%), and gelE (94.3%) were the most prevalent virulence genes. The most common antibiotic resistance pattern was tetracycline (88.6%) and minocycline (87.1%). Multi-drug resistant phenotype was detected among 10% of them. Our results showed capability of E. faecalis strains for infection of the urinary tract in community. Involvement of virulence determinants in the pathogenesis of community acquired E. faecalis strains was proposed due to their high prevalence rates. Food producing animals were proposed as their environmental reservoirs, due to dominance of tetracycline resistance phenotype among them.
Collapse
Affiliation(s)
- Abdullah Karimi
- a Pediatric Infection Research Center, Research Institute for Children Health , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Zohreh Ghalavand
- b Department of Microbiology, School of Medicine , Shahid Beheshti University of Medical Sciences , Tehran , IR Iran
| | - Fatemeh Fallah
- a Pediatric Infection Research Center, Research Institute for Children Health , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Parisa Eslami
- c Department of Microbiology , Milad Hospital , Tehran , IR Iran
| | - Mahmoud Parvin
- d Department of Pathology, Labbafinejad Hospital , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Masoud Alebouyeh
- e Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases , Shahid Beheshsti University of Medical Sciences , Tehran , Iran
| | - Marjan Rashidan
- f School of medicine , Shahroud University of Medical Sciences , Shahroud , Iran
| |
Collapse
|
40
|
Banla LI, Salzman NH, Kristich CJ. Colonization of the mammalian intestinal tract by enterococci. Curr Opin Microbiol 2018; 47:26-31. [PMID: 30439685 DOI: 10.1016/j.mib.2018.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/08/2018] [Accepted: 10/16/2018] [Indexed: 02/08/2023]
Abstract
Enterococci are colonizers of the mammalian gastrointestinal tract (GIT) and normally live in healthy association with their human host. However, enterococci are also major causes of healthcare-acquired infections, prompting the US Centers for Disease Control and Prevention to declare vancomycin-resistant enterococci (VRE) a serious threat to public health. Because of both intrinsic and acquired antibiotic resistance, enterococci proliferate in the GIT during antibiotic therapy, leading to dissemination and disease. The recognition that colonization of the GIT is a pre-requisite for enterococcal infections has prompted research to study mechanisms used by enterococci to colonize this niche. This review discusses major findings of recent research to understand GIT colonization by enterococci using diverse experimental models, each of which exhibits unique strengths. This work has revealed enterococcal transcriptional reprogramming in the GIT, contributions of specific enterococcal genes encoded by the core genome to GIT colonization, the impact of genome plasticity, and roles for intra-species and inter-species interactions in modulation of GIT colonization.
Collapse
Affiliation(s)
- Leou Ismael Banla
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Nita H Salzman
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.
| | - Christopher J Kristich
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States.
| |
Collapse
|
41
|
Singh KV, Pinkston KL, Gao P, Harvey BR, Murray BE. Anti-Ace monoclonal antibody reduces Enterococcus faecalis aortic valve infection in a rat infective endocarditis model. Pathog Dis 2018; 76:5185112. [PMID: 30445491 DOI: 10.1093/femspd/fty084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/15/2018] [Indexed: 11/13/2022] Open
Abstract
Ace (Adhesin to collagen from Enterococcus faecalis) is a cell-wall anchored protein that is expressed conditionally and is important for virulence in a rat infective endocarditis (IE) model. Previously, we showed that rats immunized with the collagen binding domain of Ace (domain A), or administered anti-Ace domain A polyclonal antibody, were less susceptible to E. faecalis endocarditis than sham-immunized controls. In this work, we demonstrated that a sub nanomolar monoclonal antibody (mAb), anti-Ace mAb70, significantly diminished E. faecalis binding to ECM collagen IV in in vitro adherence assays and that, in the endocarditis model, anti-Ace mAb70 pre-treatment significantly reduced E. faecalis infection of aortic valves. The effectiveness of anti-Ace mAb against IE in the rat model suggests it might serve as a beneficial agent for passive protection against E. faecalis infections.
Collapse
Affiliation(s)
- Kavindra V Singh
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St. Houston, TX 77030, USA.,UTHealth's Center for Antimicrobial Resistance and Microbial Genomics (CARMiG), 6431 Fannin St., Houston, TX 77030
| | - Kenneth L Pinkston
- Center for Molecular Imaging, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, 1825 Pressler St, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Peng Gao
- Center for Molecular Imaging, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, 1825 Pressler St, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Barrett R Harvey
- Center for Molecular Imaging, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, 1825 Pressler St, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, 6431 Fannin St. Houston, TX 77030, USA
| | - Barbara E Murray
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St. Houston, TX 77030, USA.,UTHealth's Center for Antimicrobial Resistance and Microbial Genomics (CARMiG), 6431 Fannin St., Houston, TX 77030.,Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, 6431 Fannin St. Houston, TX 77030, USA
| |
Collapse
|
42
|
Ch’ng JH, Chong KKL, Lam LN, Wong JJ, Kline KA. Biofilm-associated infection by enterococci. Nat Rev Microbiol 2018; 17:82-94. [DOI: 10.1038/s41579-018-0107-z] [Citation(s) in RCA: 253] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
43
|
Tartari T, Wichnieski C, Bachmann L, Jafelicci M, Silva RM, Letra A, van der Hoeven R, Duarte MAH, Bramante CM. Effect of the combination of several irrigants on dentine surface properties, adsorption of chlorhexidine and adhesion of microorganisms to dentine. Int Endod J 2018; 51:1420-1433. [DOI: 10.1111/iej.12960] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/30/2018] [Indexed: 11/28/2022]
Affiliation(s)
- T. Tartari
- Department of Operative Dentistry, Endodontics, and Dental Materials; Bauru School of Dentistry; University of São Paulo; Bauru SP Brazil
| | - C. Wichnieski
- Department of Endodontics; Pontifical Catholic University of Paraná; Curitiba PR Brazil
| | - L. Bachmann
- Department of Physics; Faculty of Philosophy, Science and Literature of Ribeirão Preto; University of São Paulo; Ribeirão Preto SP Brazil
| | - M. Jafelicci
- Department of Physical Chemistry; Institute of Chemistry; São Paulo State University; Araraquara SP Brazil
| | - R. M. Silva
- Department of Endodontics; School of Dentistry; University of Texas Health Science Center at Houston; Houston TX USA
| | - A. Letra
- Department of Endodontics; School of Dentistry; University of Texas Health Science Center at Houston; Houston TX USA
- Department of Diagnostic and Biomedical Sciences; School of Dentistry; University of Texas Health Science Center at Houston; Houston TX USA
| | - R. van der Hoeven
- Department of Diagnostic and Biomedical Sciences; School of Dentistry; University of Texas Health Science Center at Houston; Houston TX USA
| | - M. A. H. Duarte
- Department of Operative Dentistry, Endodontics, and Dental Materials; Bauru School of Dentistry; University of São Paulo; Bauru SP Brazil
| | - C. M. Bramante
- Department of Operative Dentistry, Endodontics, and Dental Materials; Bauru School of Dentistry; University of São Paulo; Bauru SP Brazil
| |
Collapse
|
44
|
Ó Cuív P, Giri R, Hoedt EC, McGuckin MA, Begun J, Morrison M. Enterococcus faecalis AHG0090 is a Genetically Tractable Bacterium and Produces a Secreted Peptidic Bioactive that Suppresses Nuclear Factor Kappa B Activation in Human Gut Epithelial Cells. Front Immunol 2018; 9:790. [PMID: 29720977 PMCID: PMC5915459 DOI: 10.3389/fimmu.2018.00790] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/29/2018] [Indexed: 12/20/2022] Open
Abstract
Enterococcus faecalis is an early coloniser of the human infant gut and contributes to the development of intestinal immunity. To better understand the functional capacity of E. faecalis, we constructed a broad host range RP4 mobilizable vector, pEHR513112, that confers chloramphenicol resistance and used a metaparental mating approach to isolate E. faecalis AHG0090 from a fecal sample collected from a healthy human infant. We demonstrated that E. faecalis AHG0090 is genetically tractable and could be manipulated using traditional molecular microbiology approaches. E. faecalis AHG0090 was comparable to the gold-standard anti-inflammatory bacterium Faecalibacterium prausnitzii A2-165 in its ability to suppress cytokine-mediated nuclear factor kappa B (NF-κB) activation in human gut-derived LS174T goblet cell like and Caco-2 enterocyte-like cell lines. E. faecalis AHG0090 and F. prausnitzii A2-165 produced secreted low molecular weight NF-κB suppressive peptidic bioactives. Both bioactives were sensitive to heat and proteinase K treatments although the E. faecalis AHG0090 bioactive was more resilient to both forms of treatment. As expected, E. faecalis AHG0090 suppressed IL-1β-induced NF-κB-p65 subunit nuclear translocation and expression of the NF-κB regulated genes IL-6, IL-8 and CXCL-10. Finally, we determined that E. faecalis AHG0090 is distantly related to other commensal strains and likely encodes niche factors that support effective colonization of the infant gut.
Collapse
Affiliation(s)
- Páraic Ó Cuív
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Rabina Giri
- Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Emily C Hoedt
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Michael A McGuckin
- Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Jakob Begun
- Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Mark Morrison
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
45
|
Enterococcus faecalis Demonstrates Pathogenicity through Increased Attachment in an Ex Vivo Polymicrobial Pulpal Infection. Infect Immun 2018; 86:IAI.00871-17. [PMID: 29483293 DOI: 10.1128/iai.00871-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 02/21/2018] [Indexed: 11/20/2022] Open
Abstract
This study investigated the host response to a polymicrobial pulpal infection consisting of Streptococcus anginosus and Enterococcus faecalis, bacteria commonly implicated in dental abscesses and endodontic failure, using a validated ex vivo rat tooth model. Tooth slices were inoculated with planktonic cultures of S. anginosus or E. faecalis alone or in coculture at S. anginosus/E. faecalis ratios of 50:50 and 90:10. Attachment was semiquantified by measuring the area covered by fluorescently labeled bacteria. Host response was established by viable histological cell counts, and inflammatory response was measured using reverse transcription-quantitative PCR (RT-qPCR) and immunohistochemistry. A significant reduction in cell viability was observed for single and polymicrobial infections, with no significant differences between infection types (∼2,000 cells/mm2 for infected pulps compared to ∼4,000 cells/mm2 for uninfected pulps). E. faecalis demonstrated significantly higher levels of attachment (6.5%) than S. anginosus alone (2.3%) and mixed-species infections (3.4% for 50:50 and 2.3% for 90:10), with a remarkable affinity for the pulpal vasculature. Infections with E. faecalis demonstrated the greatest increase in tumor necrosis factor alpha (TNF-α) (47.1-fold for E. faecalis, 14.6-fold for S. anginosus, 60.1-fold for 50:50, and 25.0-fold for 90:10) and interleukin 1β (IL-1β) expression (54.8-fold for E. faecalis, 8.8-fold for S. anginosus, 54.5-fold for 50:50, and 39.9-fold for 90:10) compared to uninfected samples. Immunohistochemistry confirmed this, with the majority of inflammation localized to the pulpal vasculature and odontoblast regions. Interestingly, E. faecalis supernatant and heat-killed E. faecalis treatments were unable to induce the same inflammatory response, suggesting E. faecalis pathogenicity in pulpitis is linked to its greater ability to attach to the pulpal vasculature.
Collapse
|
46
|
Fan W, Huang Z, Fan B. Effects of prolonged exposure to moderate static magnetic field and its synergistic effects with alkaline pH on Enterococcus faecalis. Microb Pathog 2017; 115:117-122. [PMID: 29241767 DOI: 10.1016/j.micpath.2017.12.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/09/2017] [Accepted: 12/09/2017] [Indexed: 01/02/2023]
Abstract
Static magnetic field (SMF) has been shown to biologically affect various microorganisms, but its effects on Enterococcus faecalis, which is associated with multiple dental infections, have not been reported yet. Besides, Enterococcus faecalis was found to be resistant to the alkaline environment provided by a major dental antimicrobial, calcium hydroxide. Therefore, the antibacterial activity of prolonged exposure to moderate SMF (170 mT) and its possible synergistic activity with alkaline pH (pH = 9) were evaluated in the study. The ability to form a biofilm under these conditions was examined by crystal violet assay. Real-time quantitative PCR was performed to evaluate the relative expression of stress (dnaK and groEL) and virulence (efaA, ace, gelE and fsrC) related genes. As the results indicated, cell proliferation was inhibited after 120 h of SMF exposure. What's more, the combined treatment of SMF and alkaline pH showed significantly improved antimicrobial action when compared to single SMF and alkaline pH treatment for more than 24 h and 72 h respectively. However, the ability to form a biofilm was also enhanced under SMF and alkaline pH treatments. SMF can induce stress response by up-regulating the expression of dnaK and elevate virulence gene expression (efaA and ace). These responses were more significant and more genes were up-regulated including groEL, gelE and fsrC when exposed to SMF and alkaline pH simultaneously. Hence, combination of SMF and alkaline pH could be a promising disinfection strategy in dental area and other areas associated with Enterococcus faecalis infections.
Collapse
Affiliation(s)
- Wei Fan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, People's Republic of China
| | - Zhuo Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, People's Republic of China
| | - Bing Fan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, People's Republic of China.
| |
Collapse
|
47
|
Belmouhand M, Krohn PS, Svendsen LB, Henriksen A, Hansen CP, Achiam MP. The occurrence of Enterococcus faecium and faecalis Is significantly associated With anastomotic leakage After pancreaticoduodenectomy. Scand J Surg 2017; 107:107-113. [PMID: 28980499 DOI: 10.1177/1457496917731188] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Enterococcus has emerged as a virulent species; Enterococcus faecium especially has arisen as a source of nosocomial infections. Furthermore, specific Enterococcus faecalis species are significantly associated with anastomotic leakage in rodent studies. The objective of this study was to investigate whether the occurrence of Enterococci ( E. faecium and E. faecalis) obtained from drain samples was associated with leakage in humans undergoing pancreaticoduodenectomy. MATERIALS AND METHODS All patients undergoing pancreaticoduodenectomy had a peritoneal drain sample sent for culturing between postoperative days 3 and 10. Postoperative pancreatic fistulas were defined and classified according to the International Study Group of Pancreatic Fistula. Bile leakage was radiologically verified. Postoperative complications were classified according to the Dindo-Clavien classification. RESULTS A total of 70 patients were eligible and enrolled in this study. Anastomosis leakage was observed in 19 patients; 1 leakage corresponding to the hepaticojejunostomy and 18 pancreatic fistulas were identified. In total, 10 patients (53%) with leakage had Enterococci-positive drain samples versus 12 patients (24%) without leakage [odds ratio (OR) = 5.1, 95% confidence interval (CI) = 1.4-19.4, p = 0.02]. Preoperative biliary drainage with either endoscopic stenting or a percutaneous transhepatic cholangiography catheter was associated with the occurrence of Enterococci in drain samples (OR = 5.67, 95% CI = 1.8-12.9, p = 0.003), but preoperative biliary drainage was not associated with leakage (OR = 0.45, 95% CI = 0.1-1.7, p = 0.23). CONCLUSION Enterococci in drain sample cultures in patients undergoing pancreaticoduodenectomy occurs significantly more among patients with anastomotic leakage compared to patients without leakage.
Collapse
Affiliation(s)
- M Belmouhand
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - P S Krohn
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - L B Svendsen
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - A Henriksen
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - C P Hansen
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - M P Achiam
- Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
48
|
The Enterococcus faecalis virulence factor ElrA interacts with the human Four-and-a-Half LIM Domains Protein 2. Sci Rep 2017; 7:4581. [PMID: 28676674 PMCID: PMC5496941 DOI: 10.1038/s41598-017-04875-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/22/2017] [Indexed: 12/22/2022] Open
Abstract
The commensal bacterium Enterococcus faecalis is a common cause of nosocomial infections worldwide. The increasing prevalence of multi-antibiotic resistant E. faecalis strains reinforces this public health concern. Despite numerous studies highlighting several pathology-related genetic traits, the molecular mechanisms of E. faecalis virulence remain poorly understood. In this work, we studied 23 bacterial proteins that could be considered as virulence factors or involved in the Enterococcus interaction with the host. We systematically tested their interactions with human proteins using the Human ORFeome library, a set of 12,212 human ORFs, in yeast. Among the thousands of tested interactions, one involving the E. faecalis virulence factor ElrA and the human protein FHL2 was evidenced by yeast two-hybrid and biochemically confirmed. Further molecular characterizations allowed defining an FHL2-interacting domain (FID) of ElrA. Deletion of the FID led to an attenuated in vivo phenotype of the mutated strain clearly indicating that this interaction is likely to contribute to the multifactorial virulence of this opportunistic pathogen. Altogether, our results show that FHL2 is the first host cellular protein directly targeted by an E. faecalis virulence factor and that this interaction is involved in Enterococcus pathogenicity.
Collapse
|
49
|
Tartari T, Bachmann L, Zancan RF, Vivan RR, Duarte MAH, Bramante CM. Analysis of the effects of several decalcifying agents alone and in combination with sodium hypochlorite on the chemical composition of dentine. Int Endod J 2017; 51 Suppl 1:e42-e54. [DOI: 10.1111/iej.12764] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 03/13/2017] [Indexed: 11/27/2022]
Affiliation(s)
- T. Tartari
- Department of Operative Dentistry, Endodontics and Dental Materials; Bauru School of Dentistry; University of São Paulo; Bauru SP Brazil
| | - L. Bachmann
- Department of Physics; Faculty of Philosophy, Science and Literature of Ribeirão Preto; University of São Paulo; Ribeirão Preto SP, Brazil
| | - R. F. Zancan
- Department of Operative Dentistry, Endodontics and Dental Materials; Bauru School of Dentistry; University of São Paulo; Bauru SP Brazil
| | - R. R. Vivan
- Department of Operative Dentistry, Endodontics and Dental Materials; Bauru School of Dentistry; University of São Paulo; Bauru SP Brazil
| | - M. A. H. Duarte
- Department of Operative Dentistry, Endodontics and Dental Materials; Bauru School of Dentistry; University of São Paulo; Bauru SP Brazil
| | - C. M. Bramante
- Department of Operative Dentistry, Endodontics and Dental Materials; Bauru School of Dentistry; University of São Paulo; Bauru SP Brazil
| |
Collapse
|
50
|
Bachmann R, Leonard D, Delzenne N, Kartheuser A, Cani PD. Novel insight into the role of microbiota in colorectal surgery. Gut 2017; 66:738-749. [PMID: 28153961 DOI: 10.1136/gutjnl-2016-312569] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/21/2016] [Accepted: 12/24/2016] [Indexed: 12/12/2022]
Abstract
Recent literature undeniably supports the idea that the microbiota has a strong influence on the healing process of an intestinal anastomosis. Understanding the mechanisms by which the bacterial community of the gut influences intestinal healing could open the door for new preventive and therapeutic approaches. Among the different mechanisms, data have shown that the production of specific reactive oxygen species (ROS) and the activation of specific formyl peptide receptors (FPRs) regulate intestinal wound healing. Evidence suggests that specific gut microbes such as Lactobacillus spp and Akkermansia muciniphila help to regulate healing processes through both ROS-dependent and FPR-dependent mechanisms. In this review, we will discuss the current knowledge and future perspectives concerning the impact of microbiota on wound healing. We will further review available evidence on whether mechanical bowel preparation and the use of specific antibiotics are beneficial or harmful procedures, an ongoing matter of debate. These practices have a profound effect on the gut microbiota composition at the level of both the mucosal and the luminal compartments. Therefore, a key question remains unanswered: should we continue to prepare the gut before surgical intervention? Current knowledge and data do not clearly support the use of one technique or another to avoid complications such as anastomotic leak. There is an urgent need for appropriate interventions with a deep microbiota analysis to investigate both the surgical technical benefits of a proper anastomosis compared with the potential effect of the gut microbes (beneficial vs harmful) on the processes of wound healing and anastomotic leakage reduction.
Collapse
Affiliation(s)
- Radu Bachmann
- Colorectal Surgery Unit, Cliniques universitaires Saint-Luc, Brussels, Belgium.,Cliniques des Pathologies Tumorales du Colon et de Rectum (CPTCR), Institut Roi Albert II, Cliniques universitaires Saint Luc, Brussels, Belgium
| | - Daniel Leonard
- Colorectal Surgery Unit, Cliniques universitaires Saint-Luc, Brussels, Belgium.,Cliniques des Pathologies Tumorales du Colon et de Rectum (CPTCR), Institut Roi Albert II, Cliniques universitaires Saint Luc, Brussels, Belgium
| | - Nathalie Delzenne
- Université catholique de Louvain, Louvain Drug Research Institute, Metabolism and Nutrition research group, Brussels, Belgium
| | - Alex Kartheuser
- Colorectal Surgery Unit, Cliniques universitaires Saint-Luc, Brussels, Belgium.,Cliniques des Pathologies Tumorales du Colon et de Rectum (CPTCR), Institut Roi Albert II, Cliniques universitaires Saint Luc, Brussels, Belgium
| | - Patrice D Cani
- Université catholique de Louvain, Louvain Drug Research Institute, Metabolism and Nutrition research group, Brussels, Belgium.,WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Brussels, Belgium
| |
Collapse
|