1
|
Abankwa A, Squires N, Sansone S, Asfaw T, Segal S. Evolving Enterococcus faecalis Biofilms and Urinary Tract Infection Relapse: Does Vaginal Estrogen Matter? UROGYNECOLOGY (PHILADELPHIA, PA.) 2025; 31:422-427. [PMID: 39715044 DOI: 10.1097/spv.0000000000001645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
IMPORTANCE Enterococcus faecalis urinary tract infection (UTI) is common in postmenopausal females and these bacteria create biofilms that may reduce treatment efficacy. The role of local vaginal estrogen therapy in susceptibility to E. faecalis infection is unclear. OBJECTIVE The aim of the study was to evaluate differences in the treatment of relapsing E. faecalis UTIs in postmenopausal women using vaginal estrogen compared to premenopausal women. STUDY DESIGN This was a secondary analysis of a retrospective cohort study of 71 female ambulatory patients seen within the gynecology or urogynecology practices between 2011 and 2020. Patients included had symptomatic E. faecalis UTI and a diagnosis of recurrent UTI. Patients with asymptomatic bacteriuria and concurrent pregnancy were excluded. Data was retrieved by chart review, stored, and analyzed utilizing descriptive statistics. A 2-sided Fisher exact test was performed to compare outcomes between postmenopausal and premenopausal patients and the prescription of additional rounds of antibiotics for relapse. RESULTS Within this cohort, 57.8% were postmenopausal and 42.2% were premenopausal. There was no statistically significant difference in the need for additional antibiotics between postmenopausal and premenopausal patients (10.8% vs 14.3%, P = 0.72), postmenopausal patients not using vaginal estrogen and premenopausal patients (0% vs 14.3%, P = 0.28), postmenopausal patients using vaginal estrogen and premenopausal patients (20% vs 14.3%, P = 0.70), and among postmenopausal vaginal estrogen users and nonusers (20% vs 0%, P = 0.11). CONCLUSIONS A small percentage of premenopausal and postmenopausal patients with recurrent UTI required additional antibiotics for E. faecalis relapse. However, there are no statistically significant differences between our estrogen-deficient or estrogenized postmenopausal patients, and premenopausal patients.
Collapse
Affiliation(s)
- Aileen Abankwa
- Department of Urology, New York Presbyterian-Weill Cornell Medicine, New York, NY
| | | | - Stephanie Sansone
- Department of Obstetrics and Gynecology, Hackensack Meridian Jersey Shore Medical Center, Neptune, NJ
| | | | | |
Collapse
|
2
|
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
|
3
|
Flores C, Rohn JL. Bacterial adhesion strategies and countermeasures in urinary tract infection. Nat Microbiol 2025; 10:627-645. [PMID: 39929975 DOI: 10.1038/s41564-025-01926-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/07/2025] [Indexed: 03/06/2025]
Abstract
Urinary tract infections (UTIs) are compounded by antimicrobial resistance, which increases the risk of UTI recurrence and antibiotic treatment failure. This also intensifies the burden of disease upon healthcare systems worldwide, and of morbidity and mortality. Uropathogen adhesion is a critical step in the pathogenic process, as has been mainly shown for Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, Streptococcus agalactiae, Proteus, Enterococcus and Staphylococcus species. Although many bacterial adhesion molecules from these uropathogens have been described, our understanding of their contributions to UTIs is limited. Here we explore knowledge gaps in the UTI field, as we discuss the broader repertoire of uropathogen adhesins, including their role beyond initial attachment and the counter-responses of the host immune system. Finally, we describe the development of therapeutic approaches that target uropathogenic adhesion strategies and provide potential alternatives to antibiotics.
Collapse
Affiliation(s)
- Carlos Flores
- Biozentrum, University of Basel, Basel, Switzerland.
| | - Jennifer L Rohn
- Centre for Urological Biology, Division of Medicine, University College London, London, UK.
| |
Collapse
|
4
|
Imani S, Lv S, Qian H, Cui Y, Li X, Babaeizad A, Wang Q. Current innovations in mRNA vaccines for targeting multidrug-resistant ESKAPE pathogens. Biotechnol Adv 2025; 79:108492. [PMID: 39637949 DOI: 10.1016/j.biotechadv.2024.108492] [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: 08/19/2024] [Revised: 10/30/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
The prevalence of multidrug-resistant (MDR) ESKAPE pathogens, including Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, and Pseudomonas aeruginosa, represents a critical global public health challenge. In response, mRNA vaccines offer an adaptable and scalable platform for immunotherapy against ESKAPE pathogens by encoding specific antigens that stimulate B-cell-driven antibody production and CD8+ T-cell-mediated cytotoxicity, effectively neutralizing these pathogens and combating resistance. This review examines recent advancements and ongoing challenges in the development of mRNA vaccines targeting MDR ESKAPE pathogens. We explore antigen selection, the nuances of mRNA vaccine technology, and the complex interactions between bacterial infections and antibiotic resistance. By assessing the potential efficacy of mRNA vaccines and addressing key barriers to their paraclinical implementation, this review highlights the promising function of mRNA-based immunization in combating MDR ESKAPE pathogens.
Collapse
Affiliation(s)
- Saber Imani
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Shuojie Lv
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Hongbo Qian
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Yulan Cui
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - XiaoYan Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Ali Babaeizad
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Qingjing Wang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China.
| |
Collapse
|
5
|
Stærk K, Heidtmann CV, Hjelmager JS, Ewald JD, Nielsen CU, Nielsen P, Andersen TE. The infectious capacity of Enterococcus faecalis, Staphylococcus aureus, and Staphylococcus saprophyticus in a porcine model of urinary tract infection. APMIS 2024; 132:807-813. [PMID: 39295304 DOI: 10.1111/apm.13469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/07/2024] [Indexed: 09/21/2024]
Abstract
The purpose of this study was to establish a porcine model of urinary tract infection (UTI) with gram-positive uropathogens. Ten female domestic pigs were experimentally inoculated with human UTI isolates of Enterococcus faecalis (n = 3), Staphylococcus saprophyticus (n = 3), or Staphylococcus aureus (n = 4) and followed with regular urine samples. Bladders and kidneys were aseptically removed at termination (5-7 days post infection) and assessed by gross pathology and bacterial enumeration. Enterococcus faecalis (n = 3 of 3) and S. aureus (n = 2 of 4) successfully colonized the pig bladders. Inoculation with S. saprophyticus never resulted in detectable bacteriuria. All infected pigs had cleared the infection spontaneously before termination. Surprisingly, three (of four) pigs inoculated with S. aureus led to spontaneous infection with opportunistic pathogens. Also, one pig colonized with E. faecalis resulted in spontaneous infection with E. coli. In conlusion, the pig supports experimental UTI with E. faecalis for up to 24 h but not prolonged infection. S. aureus and S. saprophyticus fails to cause UTI in pigs and other animals should be considered for studying these pathogens.
Collapse
Affiliation(s)
- Kristian Stærk
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | | | - Jesper Dupont Ewald
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Carsten Uhd Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark
| | - Poul Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark
| | - Thomas Emil Andersen
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Nappi F. Current Knowledge of Enterococcal Endocarditis: A Disease Lurking in Plain Sight of Health Providers. Pathogens 2024; 13:235. [PMID: 38535578 PMCID: PMC10974565 DOI: 10.3390/pathogens13030235] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 02/11/2025] Open
Abstract
Enterococcus faecalis is a bacterial pathogen that can cause opportunistic infections. Studies indicate that initial biofilm formation plays a crucial regulatory role in these infections, as well as in colonising and maintaining the gastrointestinal tract as a commensal member of the microbiome of most land animals. It has long been thought that vegetation of endocarditis resulting from bacterial attachment to the endocardial endothelium requires some pre-existing tissue damage, and in animal models of experimental endocarditis, mechanical valve damage is typically induced by cardiac catheterisation preceding infection. This section reviews historical and contemporary animal model studies that demonstrate the ability of E. faecalis to colonise the undamaged endovascular endothelial surface directly and produce robust microcolony biofilms encapsulated within a bacterially derived extracellular matrix. This report reviews both previous and current animal model studies demonstrating the resilient capacity of E. faecalis to colonise the undamaged endovascular endothelial surface directly and produce robust microcolony biofilms encapsulated in a bacterially derived extracellular matrix. The article also considers the morphological similarities when these biofilms develop on different host sites, such as when E. faecalis colonises the gastrointestinal epithelium as a commensal member of the common vertebrate microbiome, lurking in plain sight and transmitting systemic infection. These phenotypes may enable the organism to survive as an unrecognised infection in asymptomatic subjects, providing an infectious resource for subsequent clinical process of endocarditis.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
9
|
Daca A, Jarzembowski T. From the Friend to the Foe- Enterococcus faecalis Diverse Impact on the Human Immune System. Int J Mol Sci 2024; 25:2422. [PMID: 38397099 PMCID: PMC10888668 DOI: 10.3390/ijms25042422] [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: 01/30/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Enterococcus faecalis is a bacterium which accompanies us from the first days of our life. As a commensal it produces vitamins, metabolizes nutrients, and maintains intestinal pH. All of that happens in exchange for a niche to inhabit. It is not surprising then, that the bacterium was and is used as an element of many probiotics and its positive impact on the human immune system and the body in general is hard to ignore. This bacterium has also a dark side though. The plasticity and relative ease with which one acquires virulence traits, and the ability to hide from or even deceive and use the immune system to spread throughout the body make E. faecalis a more and more dangerous opponent. The statistics clearly show its increasing role, especially in the case of nosocomial infections. Here we present the summarization of current knowledge about E. faecalis, especially in the context of its relations with the human immune system.
Collapse
Affiliation(s)
- Agnieszka Daca
- Department of Physiopathology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Tomasz Jarzembowski
- Department of Microbiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| |
Collapse
|
10
|
Șchiopu P, Toc DA, Colosi IA, Costache C, Ruospo G, Berar G, Gălbău ȘG, Ghilea AC, Botan A, Pană AG, Neculicioiu VS, Todea DA. An Overview of the Factors Involved in Biofilm Production by the Enterococcus Genus. Int J Mol Sci 2023; 24:11577. [PMID: 37511337 PMCID: PMC10380289 DOI: 10.3390/ijms241411577] [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] [Received: 06/14/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Enterococcus species are known for their ability to form biofilms, which contributes to their survival in extreme environments and involvement in persistent bacterial infections, especially in the case of multi-drug-resistant strains. This review aims to provide a comprehensive understanding of the mechanisms underlying biofilm formation in clinically important species such as Enterococcus faecalis and the less studied but increasingly multi-drug-resistant Enterococcus faecium, and explores potential strategies for their eradication. Biofilm formation in Enterococcus involves a complex interplay of genes and virulence factors, including gelatinase, cytolysin, Secreted antigen A, pili, microbial surface components that recognize adhesive matrix molecules (MSCRAMMs), and DNA release. Quorum sensing, a process of intercellular communication, mediated by peptide pheromones such as Cob, Ccf, and Cpd, plays a crucial role in coordinating biofilm development by targeting gene expression and regulation. Additionally, the regulation of extracellular DNA (eDNA) release has emerged as a fundamental component in biofilm formation. In E. faecalis, the autolysin N-acetylglucosaminidase and proteases such as gelatinase and serin protease are key players in this process, influencing biofilm development and virulence. Targeting eDNA may offer a promising avenue for intervention in biofilm-producing E. faecalis infections. Overall, gaining insights into the intricate mechanisms of biofilm formation in Enterococcus may provide directions for anti-biofilm therapeutic research, with the purpose of reducing the burden of Enterococcus-associated infections.
Collapse
Affiliation(s)
- Pavel Șchiopu
- Department of Microbiology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Pneumology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| | - Dan Alexandru Toc
- Department of Microbiology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Ioana Alina Colosi
- Department of Microbiology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Carmen Costache
- Department of Microbiology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Giuseppe Ruospo
- Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - George Berar
- Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Ștefan-Gabriel Gălbău
- Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Alexandra Cristina Ghilea
- Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Alexandru Botan
- Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Adrian-Gabriel Pană
- Department of Microbiology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Vlad Sever Neculicioiu
- Department of Microbiology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Doina Adina Todea
- Department of Pneumology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| |
Collapse
|
11
|
Mancuso G, Midiri A, Gerace E, Marra M, Zummo S, Biondo C. Urinary Tract Infections: The Current Scenario and Future Prospects. Pathogens 2023; 12:pathogens12040623. [PMID: 37111509 PMCID: PMC10145414 DOI: 10.3390/pathogens12040623] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections worldwide, occurring in both community and healthcare settings. Although the clinical symptoms of UTIs are heterogeneous and range from uncomplicated (uUTIs) to complicated (cUTIs), most UTIs are usually treated empirically. Bacteria are the main causative agents of these infections, although more rarely, other microorganisms, such as fungi and some viruses, have been reported to be responsible for UTIs. Uropathogenic Escherichia coli (UPEC) is the most common causative agent for both uUTIs and cUTIs, followed by other pathogenic microorganisms, such as Klebsiella pneumoniae, Proteus mirabilis, Enterococcus faecalis, and Staphylococcus spp. In addition, the incidence of UTIs caused by multidrug resistance (MDR) is increasing, resulting in a significant increase in the spread of antibiotic resistance and the economic burden of these infections. Here, we discuss the various factors associated with UTIs, including the mechanisms of pathogenicity related to the bacteria that cause UTIs and the emergence of increasing resistance in UTI pathogens.
Collapse
Affiliation(s)
- Giuseppe Mancuso
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Angelina Midiri
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | | | - Maria Marra
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Sebastiana Zummo
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Carmelo Biondo
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
12
|
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: 11] [Impact Index Per Article: 5.5] [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
|
13
|
Govindarajan DK, Kandaswamy K. Virulence factors of uropathogens and their role in host pathogen interactions. Cell Surf 2022; 8:100075. [PMID: 35198842 PMCID: PMC8841375 DOI: 10.1016/j.tcsw.2022.100075] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/01/2022] [Accepted: 02/06/2022] [Indexed: 12/26/2022] Open
Abstract
Gram-positive and Gram-negative bacterial pathogens are commonly found in Urinary Tract Infection (UTI), particularly infected in females like pregnant women, elder people, sexually active, or individuals prone to other risk factors for UTI. In this article, we review the expression of virulence surface proteins and their interaction with host cells for the most frequently isolated uropathogens: Escherichia coli, Enterococcus faecalis, Proteus mirabilis, Klebsiella pneumoniae, and Staphylococcus saprophyticus. In addition to the host cell interaction, surface protein regulation was also discussed in this article. The surface protein regulation serves as a key tool in differentiating the pathogen isotypes. Furthermore, it might provide insights on novel diagnostic methods to detect uropathogen that are otherwise easily overlooked due to limited culture-based assays. In essence, this review shall provide an in-depth understanding on secretion of virulence factors of various uropathogens and their role in host-pathogen interaction, this knowledge might be useful in the development of therapeutics against uropathogens.
Collapse
Affiliation(s)
| | - Kumaravel Kandaswamy
- Corresponding author at: Department of Biotechnology, Kumaraguru College of Technology (KCT), Chinnavedampatti, Coimbatore 641049, Tamil Nadu, India.
| |
Collapse
|
14
|
Zhang X, Feng H, He J, Muhammad A, Zhang F, Lu X. Features and Colonization Strategies of Enterococcus faecalis in the Gut of Bombyx mori. Front Microbiol 2022; 13:921330. [PMID: 35814682 PMCID: PMC9263704 DOI: 10.3389/fmicb.2022.921330] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/08/2022] [Indexed: 11/30/2022] Open
Abstract
The complex gut microbiome is a malleable microbial community that can undergo remodeling in response to many factors, including the gut environment and microbial properties. Enterococcus has emerged as one of the predominant gut commensal bacterial and plays a fundamental role in the host physiology and health of the major economic agricultural insect, Bombyx mori. Although extensive research on gut structure and microbiome diversity has been carried out, how these microbial consortia are established in multifarious niches within the gut has not been well characterized to date. Here, an Enterococcus species that was stably associated with its host, the model organism B. mori, was identified in the larval gut. GFP–tagged E. faecalis LX10 was constructed as a model bacterium to track the colonization mechanism in the intestine of B. mori. The results revealed that the minimum and optimum colonization results were obtained by feeding at doses of 105 CFU/silkworm and 107 CFU/silkworm, respectively, as confirmed by bioassays and fluorescence-activated cell sorting analyses (FACS). Furthermore, a comprehensive genome-wide exploration of signal sequences provided insight into the relevant colonization properties of E. faecalis LX10. E. faecalis LX10 grew well under alkaline conditions and stably reduced the intestinal pH through lactic acid production. Additionally, the genomic features responsible for lactic acid fermentation were characterized. We further expressed and purified E. faecalis bacteriocin and found that it was particularly effective against other gut bacteria, including Enterococcus casselifavus, Enterococcus mundtii, Serratia marcescens, Bacillus amyloliquefaciens, and Escherichia coli. In addition, the successful colonization of E. faecalis LX10 led to drastically increased expression of all adhesion genes (znuA, lepB, hssA, adhE, EbpA, and Lap), defense genes (cspp, tagF, and esp), regulation gene (BfmRS), secretion gene (prkC) and immune evasion genes (patA and patB), while the expression of iron acquisition genes (ddpD and metN) was largely unchanged or decreased. This work establishes an unprecedented conceptual model for understanding B. mori–gut microbiota interactions in an ecological context. Moreover, these results shed light on the molecular mechanisms of gut microbiota proliferation and colonization in the intestinal tract of this insect.
Collapse
Affiliation(s)
- Xiancui Zhang
- College of Animal Sciences, Institute of Sericulture and Apiculture, Zhejiang University, Hangzhou, China
| | - Huihui Feng
- College of Animal Sciences, Institute of Sericulture and Apiculture, Zhejiang University, Hangzhou, China
| | - Jintao He
- College of Animal Sciences, Institute of Sericulture and Apiculture, Zhejiang University, Hangzhou, China
| | - Abrar Muhammad
- College of Animal Sciences, Institute of Sericulture and Apiculture, Zhejiang University, Hangzhou, China
| | - Fan Zhang
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan, China
- *Correspondence: Fan Zhang,
| | - Xingmeng Lu
- College of Animal Sciences, Institute of Sericulture and Apiculture, Zhejiang University, Hangzhou, China
- Xingmeng Lu,
| |
Collapse
|
15
|
Xu W, Fang Y, Hu Q, Zhu K. Emerging Risks in Food: Probiotic Enterococci Pose a Threat to Public Health through the Food Chain. Foods 2021; 10:foods10112846. [PMID: 34829127 PMCID: PMC8623795 DOI: 10.3390/foods10112846] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Probiotics have been associated with clinical infections, toxicity, and antimicrobial resistance transfer, raising public concerns. Probiotic enterococci are emerging food risks as opportunistic pathogens, yet little attention has been paid to them. Herein, we collected 88 enterococcal isolates from probiotic products used for humans, companion animals, livestock, and aquaculture. Results showed that all 88 probiotic enterococcal isolates harbored diverse virulence genes, multiple antimicrobial resistance genes, and mobile genetic elements. Notably, 77 isolates were highly resistant to gentamicin. Representative enterococcal isolates exerted toxic activities in both in vitro and in vivo models. Collectively, our findings suggest that probiotic enterococci may be harmful to hosts and pose a potential threat to public health.
Collapse
Affiliation(s)
| | | | | | - Kui Zhu
- Correspondence: ; Tel.: +86-10-62733695
| |
Collapse
|
16
|
Barnes AMT, Frank KL, Dale JL, Manias DA, Powers JL, Dunny GM. Enterococcus faecalis colonizes and forms persistent biofilm microcolonies on undamaged endothelial surfaces in a rabbit endovascular infection model. FEMS MICROBES 2021; 2:xtab014. [PMID: 34734186 PMCID: PMC8557322 DOI: 10.1093/femsmc/xtab014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 09/23/2021] [Indexed: 01/03/2023] Open
Abstract
Infectious endocarditis (IE) is an uncommon disease with significant morbidity and mortality. The pathogenesis of IE has historically been described as a cascade of host-specific events beginning with endothelial damage and thrombus formation and followed by bacterial colonization of the nascent thrombus. Enterococcus faecalis is a Gram-positive commensal bacterial member of the gastrointestinal tract microbiota in most terrestrial animals and a leading cause of opportunistic biofilm-associated infections, including endocarditis. Here, we provide evidence that E. faecalis can colonize the endocardial surface without pre-existing damage and in the absence of thrombus formation in a rabbit endovascular infection model. Using previously described light and scanning electron microscopy techniques, we show that inoculation of a well-characterized E. faecalis lab strain in the marginal ear vein of New Zealand White rabbits resulted in rapid colonization of the endocardium throughout the heart within 4 days of administration. Unexpectedly, ultrastructural imaging revealed that the microcolonies were firmly attached directly to the endocardium in areas without morphological evidence of gross tissue damage. Further, the attached bacterial aggregates were not associated with significant cellular components of coagulation or host extracellular matrix damage repair (i.e. platelets). These results suggest that the canonical model of mechanical surface damage as a prerequisite for bacterial attachment to host sub-endothelial components is not required. Furthermore, these findings are consistent with a model of initial establishment of stable, endocarditis-associated E. faecalis biofilm microcolonies that may provide a reservoir for the eventual valvular infection characteristic of clinical endocarditis. The similarities between the E. faecalis colonization and biofilm morphologies seen in this rabbit endovascular infection model and our previously published murine gastrointestinal colonization model indicate that biofilm production and common host cell attachment factors are conserved in disparate mammalian hosts under both commensal and pathogenic contexts.
Collapse
Affiliation(s)
- Aaron M T Barnes
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
| | - Kristi L Frank
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
| | - Jennifer L Dale
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
| | - Dawn A Manias
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
| | - Jennifer L Powers
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
| | - Gary M Dunny
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
| |
Collapse
|
17
|
Barnes AMT, Frank KL, Dunny GM. Enterococcal Endocarditis: Hiding in Plain Sight. Front Cell Infect Microbiol 2021; 11:722482. [PMID: 34527603 PMCID: PMC8435889 DOI: 10.3389/fcimb.2021.722482] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Enterococcus faecalis is a major opportunistic bacterial pathogen of increasing clinical relevance. A substantial body of experimental evidence suggests that early biofilm formation plays a critical role in these infections, as well as in colonization and persistence in the GI tract as a commensal member of the microbiome in most terrestrial animals. Animal models of experimental endocarditis generally involve inducing mechanical valve damage by cardiac catheterization prior to infection, and it has long been presumed that endocarditis vegetation formation resulting from bacterial attachment to the endocardial endothelium requires some pre-existing tissue damage. Here we review both historical and contemporary animal model studies demonstrating the robust ability of E. faecalis to directly attach and form stable microcolony biofilms encased within a bacterially-derived extracellular matrix on the undamaged endovascular endothelial surface. We also discuss the morphological similarities when these biofilms form on other host tissues, including when E. faecalis colonizes the GI epithelium as a commensal member of the normal vertebrate microbiome - hiding in plain sight where it can serve as a source for systemic infection via translocation. We propose that these phenotypes may allow the organism to persist as an undetected infection in asymptomatic individuals and thus provide an infectious reservoir for later clinical endocarditis.
Collapse
Affiliation(s)
- Aaron M. T. Barnes
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Kristi L. Frank
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN, United States
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Gary M. Dunny
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN, United States
| |
Collapse
|
18
|
Efficacy of Omadacycline against Escherichia coli in a Mouse Urinary Tract Infection Model. Antimicrob Agents Chemother 2021; 65:e0026921. [PMID: 33972239 DOI: 10.1128/aac.00269-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In a mouse urinary tract infection model, omadacycline (OMC) was comparable to gentamicin and better than ciprofloxacin (CIP) against a tetracycline-susceptible (TET-S), CIP-resistant (CIP-R) Escherichia coli strain. Gentamicin showed better efficacy than OMC against a TET-R, CIP-R E. coli strain, and OMC again showed better efficacy than CIP against this strain. OMC may warrant further study as a potential option for urinary tract infection treatment against CIP-R E. coli strains.
Collapse
|
19
|
Susmitha A, Bajaj H, Madhavan Nampoothiri K. The divergent roles of sortase in the biology of Gram-positive bacteria. ACTA ACUST UNITED AC 2021; 7:100055. [PMID: 34195501 PMCID: PMC8225981 DOI: 10.1016/j.tcsw.2021.100055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 12/16/2022]
Abstract
The bacterial cell wall contains numerous surface-exposed proteins, which are covalently anchored and assembled by a sortase family of transpeptidase enzymes. The sortase are cysteine transpeptidases that catalyzes the covalent attachment of surface protein to the cell wall peptidoglycan. Among the reported six classes of sortases, each distinct class of sortase plays a unique biological role in anchoring a variety of surface proteins to the peptidoglycan of both pathogenic and non-pathogenic Gram-positive bacteria. Sortases not only exhibit virulence and pathogenesis properties to host cells, but also possess a significant role in gut retention and immunomodulation in probiotic microbes. The two main distinct functions are to attach proteins directly to the cell wall or assemble pili on the microbial surface. This review provides a compendium of the distribution of different classes of sortases present in both pathogenic and non-pathogenic Gram-positive bacteria and also the noteworthy role played by them in bacterial cell wall assembly which enables each microbe to effectively interact with its environment.
Collapse
Affiliation(s)
- Aliyath Susmitha
- Microbial Processes and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Harsha Bajaj
- Microbial Processes and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India
| | - Kesavan Madhavan Nampoothiri
- Microbial Processes and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
20
|
Abstract
The bacterial type IV pilus (T4P) is a prominent virulence factor in many significant human pathogens, some of which have become increasingly antibiotic resistant. Antivirulence chemotherapeutics are considered a promising alternative to antibiotics because they target the disease process instead of bacterial viability. However, a roadblock to the discovery of anti-T4P compounds is the lack of a high-throughput screen (HTS) that can be implemented relatively easily and economically. Here, we describe the first HTS for the identification of inhibitors specifically against the T4P assembly ATPase PilB in vitro. Chloracidobacterium thermophilum PilB (CtPilB) had been demonstrated to have robust ATPase activity and the ability to bind its expected ligands in vitro. We utilized CtPilB and MANT-ATP, a fluorescent ATP analog, to develop a binding assay and adapted it for an HTS. As a proof of principle, we performed a pilot screen with a small compound library of kinase inhibitors and identified quercetin as a PilB inhibitor in vitro. Using Myxococcus xanthus as a model bacterium, we found quercetin to reduce its T4P-dependent motility and T4P assembly in vivo. These results validated our HTS as effective in identifying PilB inhibitors. This assay may prove valuable in seeking leads for the development of antivirulence chemotherapeutics against PilB, an essential and universal component of all bacterial T4P systems. IMPORTANCE Many bacterial pathogens use their type IV pili (T4P) to facilitate and maintain infection of a human host. Small chemical compounds that inhibit the production or assembly of T4P hold promise in the treatment and prevention of infections, especially in the era of increasing threats from antibiotic-resistant bacteria. However, few chemicals are known to have inhibitory or anti-T4P activity. Their identification has not been easy due to the lack of a method for the screening of compound collections or libraries on a large scale. Here, we report the development of an assay that can be scaled up to screen compound libraries for inhibitors of a critical T4P assembly protein. We further demonstrate that it is feasible to use whole cells to examine potential inhibitors for their activity against T4P assembly in a bacterium.
Collapse
|
21
|
Gaston JR, Johnson AO, Bair KL, White AN, Armbruster CE. Polymicrobial interactions in the urinary tract: is the enemy of my enemy my friend? Infect Immun 2021; 89:IAI.00652-20. [PMID: 33431702 DOI: 10.1128/iai.00652-20] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The vast majority of research pertaining to urinary tract infection has focused on a single pathogen in isolation, and predominantly Escherichia coli. However, polymicrobial urine colonization and infection are prevalent in several patient populations, including individuals with urinary catheters. The progression from asymptomatic colonization to symptomatic infection and severe disease is likely shaped by interactions between traditional pathogens as well as constituents of the normal urinary microbiota. Recent studies have begun to experimentally dissect the contribution of polymicrobial interactions to disease outcomes in the urinary tract, including their role in development of antimicrobial-resistant biofilm communities, modulating the innate immune response, tissue damage, and sepsis. This review aims to summarize the epidemiology of polymicrobial urine colonization, provide an overview of common urinary tract pathogens, and present key microbe-microbe and host-microbe interactions that influence infection progression, persistence, and severity.
Collapse
Affiliation(s)
- Jordan R Gaston
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo
| | - Alexandra O Johnson
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo
| | - Kirsten L Bair
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo
| | - Ashley N White
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo
| | - Chelsie E Armbruster
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo
| |
Collapse
|
22
|
Lupo F, Rousseau M, Canton T, Ingersoll MA. The Immune System Fails to Mount a Protective Response to Gram-Positive or Gram-Negative Bacterial Prostatitis. THE JOURNAL OF IMMUNOLOGY 2020; 205:2763-2777. [PMID: 33055280 DOI: 10.4049/jimmunol.2000587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/12/2020] [Indexed: 11/19/2022]
Abstract
Bacterial prostatitis affects 1% of men, with increased incidence in the elderly. Acute bacterial prostatitis frequently progresses to chronicity, marked by recurrent episodes interspersed with asymptomatic periods of variable duration. Antibiotic treatment is standard of care; however, dissemination of antimicrobially resistant uropathogens threatens therapy efficacy. Thus, development of nonantibiotic-based approaches to treat chronic disease is a priority. Currently, why chronic prostatitis arises is unclear, as the immune response to prostate infection is incompletely understood. As 80% of prostatitis cases are caused by Gram-negative uropathogenic Escherichia coli (UPEC) or Gram-positive Enterococcus faecalis, we used a mouse transurethral instillation model to address the hypothesis that an innate immune response fails to develop following prostate infection with these uropathogens, leading to chronic disease. Surprisingly, infection induced robust proinflammatory cytokine expression and myeloid cell infiltration. Following a second infection, cytokine responses and innate cell infiltration were largely comparable to primary infection. Characteristic of memory responses, more lymphoid cells infiltrated the prostate in a second infection compared with a first, suggesting that adaptive immunity develops to eliminate the pathogens. Unexpectedly, bacterial burden in prostates challenged with either UPEC or E. faecalis was equal or greater than primary infection despite that a protective adaptive response to UPEC infection was evident in the bladder of the same animals. Our findings support that chronic or recurrent prostatitis develops despite strong innate immune responses and may be the result of a failure to develop immune memory to infection, pointing to actionable targets for immunotherapy.
Collapse
Affiliation(s)
- Federico Lupo
- Department of Immunology, Institut Pasteur, 75015 Paris, France; and INSERM U1223, 75015 Paris, France
| | - Matthieu Rousseau
- Department of Immunology, Institut Pasteur, 75015 Paris, France; and INSERM U1223, 75015 Paris, France
| | - Tracy Canton
- Department of Immunology, Institut Pasteur, 75015 Paris, France; and INSERM U1223, 75015 Paris, France
| | - Molly A Ingersoll
- Department of Immunology, Institut Pasteur, 75015 Paris, France; and INSERM U1223, 75015 Paris, France
| |
Collapse
|
23
|
Basiouni S, Fayed MAA, Tarabees R, El-Sayed M, Elkhatam A, Töllner KR, Hessel M, Geisberger T, Huber C, Eisenreich W, Shehata AA. Characterization of Sunflower Oil Extracts from the Lichen Usnea barbata. Metabolites 2020; 10:metabo10090353. [PMID: 32878015 PMCID: PMC7570345 DOI: 10.3390/metabo10090353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/21/2022] Open
Abstract
The increasing global emergence of multidrug resistant (MDR) pathogens is categorized as one of the most important health problems. Therefore, the discovery of novel antimicrobials is of the utmost importance. Lichens provide a rich source of natural products including unique polyketides and polyphenols. Many of them display pharmaceutical benefits. The aim of this study was directed towards the characterization of sunflower oil extracts from the fruticose lichen, Usnea barbata. The concentration of the major polyketide, usnic acid, was 1.6 mg/mL extract as determined by NMR analysis of the crude mixture corresponding to 80 mg per g of the dried lichen. The total phenolics and flavonoids were determined by photometric assays as 4.4 mg/mL (gallic acid equivalent) and 0.27 mg/mL (rutin equivalent) corresponding to 220 mg/g and 13.7 mg/g lichen, respectively. Gram-positive (e.g., Enterococcus faecalis) and Gram-negative bacteria, as well as clinical isolates of infected chickens were sensitive against these extracts as determined by agar diffusion tests. Most of these activities increased in the presence of zinc salts. The data suggest the potential usage of U. barbata extracts as natural additives and mild antibiotics in animal husbandry, especially against enterococcosis in poultry.
Collapse
Affiliation(s)
- Shereen Basiouni
- Clinical Pathology Department, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Egypt;
| | - Marwa A. A. Fayed
- Pharmacognosy Department, Faculty of Pharmacy, University of Sadat City, Sadat 32897, Egypt;
| | - Reda Tarabees
- Institute for Bacteriology and Mycology, Faculty of Veterinary Medicine, University of Sadat City, Sadat 32897, Egypt; (R.T.); (M.E.-S.)
| | - Mohamed El-Sayed
- Institute for Bacteriology and Mycology, Faculty of Veterinary Medicine, University of Sadat City, Sadat 32897, Egypt; (R.T.); (M.E.-S.)
| | - Ahmed Elkhatam
- Department for Parasitology, Faculty of Veterinary Medicine, University of Sadat City, Sadat 32897, Egypt;
| | - Klaus-Rainer Töllner
- Research and Development Section, PerNaturam GmbH, An der Trift 8, 56290 Gödenroth, Germany; (K.-R.T.); (M.H.)
| | - Manfred Hessel
- Research and Development Section, PerNaturam GmbH, An der Trift 8, 56290 Gödenroth, Germany; (K.-R.T.); (M.H.)
| | - Thomas Geisberger
- Chair of Biochemistry, Department of Chemistry, Technical University Munich, Lichtenbergstraße 4, 85748 Garching, Germany; (T.G.); (C.H.)
| | - Claudia Huber
- Chair of Biochemistry, Department of Chemistry, Technical University Munich, Lichtenbergstraße 4, 85748 Garching, Germany; (T.G.); (C.H.)
| | - Wolfgang Eisenreich
- Chair of Biochemistry, Department of Chemistry, Technical University Munich, Lichtenbergstraße 4, 85748 Garching, Germany; (T.G.); (C.H.)
- Correspondence: (W.E.); (A.A.S.); Tel.: +49-089-289-13336 (W.E.); +49-06762-96362-137 (A.A.S.)
| | - Awad A. Shehata
- Research and Development Section, PerNaturam GmbH, An der Trift 8, 56290 Gödenroth, Germany; (K.-R.T.); (M.H.)
- Avian and Rabbit Diseases Department, Faculty of Veterinary Medicine, University of Sadat City, Sadat 32897, Egypt
- Correspondence: (W.E.); (A.A.S.); Tel.: +49-089-289-13336 (W.E.); +49-06762-96362-137 (A.A.S.)
| |
Collapse
|
24
|
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
|
25
|
Enterococcus faecalis Manganese Exporter MntE Alleviates Manganese Toxicity and Is Required for Mouse Gastrointestinal Colonization. Infect Immun 2020; 88:IAI.00058-20. [PMID: 32229614 DOI: 10.1128/iai.00058-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/21/2020] [Indexed: 01/13/2023] Open
Abstract
Bacterial pathogens encounter a variety of nutritional environments in the human host, including nutrient metal restriction and overload. Uptake of manganese (Mn) is essential for Enterococcus faecalis growth and virulence; however, it is not known how this organism prevents Mn toxicity. In this study, we examine the role of the highly conserved MntE transporter in E. faecalis Mn homeostasis and virulence. We show that inactivation of mntE results in growth restriction in the presence of excess Mn, but not other metals, demonstrating its specific role in Mn detoxification. Upon growth in the presence of excess Mn, an mntE mutant accumulates intracellular Mn, iron (Fe), and magnesium (Mg), supporting a role for MntE in Mn and Fe export and a role for Mg in offsetting Mn toxicity. Growth of the mntE mutant in excess Fe also results in increased levels of intracellular Fe, but not Mn or Mg, providing further support for MntE in Fe efflux. Inactivation of mntE in the presence of excess iron also results in the upregulation of glycerol catabolic genes and enhanced biofilm growth, and addition of glycerol is sufficient to augment biofilm growth for both the mntE mutant and its wild-type parental strain, demonstrating that glycerol availability significantly enhances biofilm formation. Finally, we show that mntE contributes to colonization of the antibiotic-treated mouse gastrointestinal (GI) tract, suggesting that E. faecalis encounters excess Mn in this niche. Collectively, these findings demonstrate that the manganese exporter MntE plays a crucial role in E. faecalis metal homeostasis and virulence.
Collapse
|
26
|
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
|
27
|
Risks associated with enterococci as probiotics. Food Res Int 2019; 129:108788. [PMID: 32036912 DOI: 10.1016/j.foodres.2019.108788] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/07/2019] [Accepted: 10/29/2019] [Indexed: 01/01/2023]
Abstract
Probiotics are naturally occurring microorganisms that confer health benefits by altering host commensal microbiota, modulating immunity, enhancing intestinal barrier function, or altering pain perception. Enterococci are human and animal intestinal commensals that are used as probiotics and in food production. These microorganisms, however, express many virulence traits including cytolysin, proteases, aggregation substance, capsular polysaccharide, enterococcal surface protein, biofilm formation, extracellular superoxide, intestinal translocation, and resistance to innate immunity that can lead to serious hospital-acquired infections. In addition, enterococci are facile in acquiring antibiotic resistance genes to many clinically important antibiotics encoded on a wide variety of conjugative plasmids, transposons, and bacteriophages. The pathogenicity and disease burden caused by enterococci render them poor choices as probiotics. No large, randomized, placebo-controlled clinical trials have demonstrated the safety and efficacy of any enterococcal probiotic. As a result, no enterococcal probiotic has been approved by the United States Food and Drug Administration for the treatment, cure, or amelioration of human disease. In 2007, the European Food Safety Authority concluded that enterococci do not meet the standard for "Qualified Presumption of Safety". Enterococcal strains used or proposed for use as probiotics should be carefully screened for efficacy and safety.
Collapse
|
28
|
Ryu BH, Hong J, Jung J, Kim MJ, Sung H, Kim MN, Chong YP, Kim SH, Lee SO, Kim YS, Woo JH, Choi SH. Clinical characteristics and treatment outcomes of Enterococcus durans bacteremia: a 20-year experience in a tertiary care hospital. Eur J Clin Microbiol Infect Dis 2019; 38:1743-1751. [PMID: 31243595 DOI: 10.1007/s10096-019-03605-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/28/2019] [Indexed: 11/25/2022]
Abstract
While the clinical characteristics and treatment outcomes of Enterococcus faecalis and E. faecium bacteremia are well known, those of E. durans bacteremia are still largely unclear. We retrospectively identified 80 adult E. durans bacteremia cases treated at our 2700-bed tertiary care hospital between January 1997 and December 2016. We compared the clinical characteristics and treatment outcomes of the adult patients with E. durans bacteremia (case group) with those of E. faecalis and E. faecium bacteremia cases (two control groups). The case and control groups were matched for sex, age, and date of onset of bacteremia. E. durans was responsible for 1.2% of all enterococcal bacteremia cases at our hospital. Of 80 cases, 39 (48.8%) had biliary tract infection and 18 (22.5%) had urinary tract infection. Community-onset bacteremia was more frequent in the case group than in the control groups (56.2% vs. 35.0% vs. 21.2%, p < 0.01). Infective endocarditis tended to be more common in the E. durans group (7.5% vs. 1.2% vs. 1.2%, p = 0.05). The majority of E. durans isolates were susceptible to penicillin (66/76, 86.8%), ampicillin (67/76, 88.2%), and vancomycin (75/76, 98.7%). The case group had significantly lower all-cause mortality (20.0% vs. 31.2% vs. 42.5%, p < 0.01) and bacteremia-related mortality (2.5% vs. 16.2% vs. 18.8%, p < 0.01) than the control groups. E. durans bacteremia mainly originates from the biliary or urinary tract and is associated with a lower risk of mortality.
Collapse
Affiliation(s)
- Byung-Han Ryu
- Department of Infectious Diseases, Gyeongsang National University Changwon Hospital, Changwon, Republic of Korea
| | - Jeongmin Hong
- Department of Infectious Diseases, Daegu Catholic University Medical Center, Daegu, Republic of Korea
| | - Jiwon Jung
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min Jae Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Heungsup Sung
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi-Na Kim
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong Pil Chong
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Oh Lee
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yang Soo Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jun Hee Woo
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Ho Choi
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
29
|
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
|
30
|
Flores-Mireles A, Hreha TN, Hunstad DA. Pathophysiology, Treatment, and Prevention of Catheter-Associated Urinary Tract Infection. Top Spinal Cord Inj Rehabil 2019; 25:228-240. [PMID: 31548790 PMCID: PMC6743745 DOI: 10.1310/sci2503-228] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Urinary tract infections (UTIs) are among the most common microbial infections in humans and represent a substantial burden on the health care system. UTIs can be uncomplicated, as when affecting healthy individuals, or complicated, when affecting individuals with compromised urodynamics and/or host defenses, such as those with a urinary catheter. There are clear differences between uncomplicated UTI and catheter-associated UTI (CAUTI) in clinical manifestations, causative organisms, and pathophysiology. Therefore, uncomplicated UTI and CAUTI cannot be approached similarly, or the risk of complications and treatment failure may increase. It is imperative to understand the key aspects of each condition to develop successful treatment options and improve patient outcomes. Here, we will review the epidemiology, pathogen prevalence, differential mechanisms used by uropathogens, and treatment and prevention of uncomplicated UTI and CAUTI.
Collapse
Affiliation(s)
| | - Teri N. Hreha
- Washington University School of Medicine, Saint Louis, Missouri
| | | |
Collapse
|
31
|
Colomer-Winter C, Lemos JA, Flores-Mireles AL. Biofilm Assays on Fibrinogen-coated Silicone Catheters and 96-well Polystyrene Plates. Bio Protoc 2019; 9:e3196. [PMID: 31106237 DOI: 10.21769/bioprotoc.3196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Biofilm formation is a well-known bacterial strategy that protects cells from hostile environments. During infection, bacteria found in a biofilm community are less sensitive to antibiotics and to the immune response, often allowing them to colonize and persist in the host niche. Not surprisingly, biofilm formation on medical devices, such as urinary catheters, is a major problem in hospital settings. To be able to eliminate such biofilms, it is important to understand the key bacterial factors that contribute to their formation. A common practice in the lab setting is to study biofilms grown in laboratory media. However, these media do not fully reflect the host environment conditions, potentially masking relevant biological determinants. This is the case during urinary catheterization, where a key element for Enterococcus faecalis and Staphylococcus aureus colonization and biofilm formation is the release of fibrinogen (Fg) into the bladder and its deposition on the urinary catheter. To recapitulate bladder conditions during catheter-associated urinary tract infection (CAUTI), we have developed a fibrinogen-coated catheter and 96-well plate biofilm assay in urine. Notably, enterococcal biofilm factors identified in these in vitro assays proved to be important for biofilm formation in vivo in a mouse model of CAUTI. Thus, the method described herein can be used to uncover biofilm-promoting factors that are uniquely relevant in the host environment, and that can be exploited to develop new antibacterial therapies.
Collapse
Affiliation(s)
- Cristina Colomer-Winter
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - José A Lemos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Ana L Flores-Mireles
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
32
|
Chen M, Pan H, Lou Y, Wu Z, Zhang J, Huang Y, Yu W, Qiu Y. Epidemiological characteristics and genetic structure of linezolid-resistant Enterococcus faecalis. Infect Drug Resist 2018; 11:2397-2409. [PMID: 30538507 PMCID: PMC6251436 DOI: 10.2147/idr.s181339] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Objectives The aim of this study was to investigate the mechanism of linezolid resistance and evaluate the risk factors for linezolid-resistant Enterococcus faecalis (LZR-Efa) infections. Methods A total of 730 E. faecalis isolates were collected, and whole-genome sequencing and bioinformatics analysis were performed. Meanwhile, risk factors related to linezolid resistance were analyzed by binary logistic regression. Results Twenty-six LZR-Efa were isolated from various clinical samples, and 24 isolates were multidrug resistant. Four isolates were daptomycin nonsusceptible, while all LZR-Efa were susceptible to vancomycin. Thirteen different sequence types (STs) were identified, and the most prevalent type was ST16 (23.1%). The genes dfrE, lsaA, and emeA were identified in all isolates. A total of 23 E. faecalis were positive for optrA gene, and six amino acids mutations were identified among 18 LZR-Efa in OptrA. The 23S rRNA mutation was found in 16 LZR-Efa isolates. However, the presence of cfr was not identified. Furthermore, there were 41 virulence genes detected, and 10 genes (ace, bopD, cpsA, cpsB, ebpB, ebpC, efaA, fss1, fss2, and srtC) were found in all isolates. A total of nine isolates were positive for multiple virulent factors (ace, asa1, cylA, efaA, esp, and gelE). There was no difference in the number of virulence factors among different specimens (P=0.825). It is of note that all patients had not been prescribed linezolid or traveled abroad previously. Moreover, previous use of carbapenems was a risk factor for LZR-Efa infections. Conclusion The main trends of LZR-Efa, with lower level of resistance, were sporadic mainly in the department of surgery. optrA and 23S rRNA were the main resistance mechanisms. In addition, carbapenems use was an independent predictor of LZR-Efa infections.
Collapse
Affiliation(s)
- Meijuan Chen
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China,
| | - Hongying Pan
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China,
| | - Yaling Lou
- Department of Infectious Diseases, Dongyang People's Hospital, Jinhua, China
| | - Zhe Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China, ,
| | - Jiajie Zhang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China,
| | - Yicheng Huang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China,
| | - Wei Yu
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China, .,State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China, ,
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China, ,
| |
Collapse
|
33
|
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
|
34
|
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]
|
35
|
Ahmadrajabi R, Dalfardi MS, Farsinejad A, Saffari F. Distribution of Ebp pili among clinical and fecal isolates ofEnterococcus faecalisand evaluation for human platelet activation. APMIS 2018; 126:314-319. [DOI: 10.1111/apm.12813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 12/12/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Roya Ahmadrajabi
- Faculty of Medicine, Microbiology Section; Bam University of Medical Sciences; Bam Iran
- Department of Microbiology; School of Medicine; Kerman University of Medical Sciences; Kerman Iran
| | - Mohammad Sadegh Dalfardi
- Department of Microbiology; School of Medicine; Kerman University of Medical Sciences; Kerman Iran
| | - Alireza Farsinejad
- Department of Hematology and Laboratory Sciences; Faculty of Allied Medicine; Kerman University of Medical Science; Kerman Iran
- Pathology and Stem Cell Research Center; Kerman University of Medical Sciences; Kerman Iran
| | - Fereshteh Saffari
- Department of Microbiology; School of Medicine; Kerman University of Medical Sciences; Kerman Iran
| |
Collapse
|
36
|
Goh HMS, Yong MHA, Chong KKL, Kline KA. Model systems for the study of Enterococcal colonization and infection. Virulence 2017; 8:1525-1562. [PMID: 28102784 PMCID: PMC5810481 DOI: 10.1080/21505594.2017.1279766] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/30/2016] [Accepted: 01/04/2017] [Indexed: 02/07/2023] Open
Abstract
Enterococcus faecalis and Enterococcus faecium are common inhabitants of the human gastrointestinal tract, as well as frequent opportunistic pathogens. Enterococci cause a range of infections including, most frequently, infections of the urinary tract, catheterized urinary tract, bloodstream, wounds and surgical sites, and heart valves in endocarditis. Enterococcal infections are often biofilm-associated, polymicrobial in nature, and resistant to antibiotics of last resort. Understanding Enterococcal mechanisms of colonization and pathogenesis are important for identifying new ways to manage and intervene with these infections. We review vertebrate and invertebrate model systems applied to study the most common E. faecalis and E. faecium infections, with emphasis on recent findings examining Enterococcal-host interactions using these models. We discuss strengths and shortcomings of each model, propose future animal models not yet applied to study mono- and polymicrobial infections involving E. faecalis and E. faecium, and comment on the significance of anti-virulence strategies derived from a fundamental understanding of host-pathogen interactions in model systems.
Collapse
Affiliation(s)
- H. M. Sharon Goh
- Singapore Centre for Environmental Life Sciences Engineering, School of Biological Sciences, Nanyang Technological University, Singapore
| | - M. H. Adeline Yong
- Singapore Centre for Environmental Life Sciences Engineering, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Kelvin Kian Long Chong
- Singapore Centre for Environmental Life Sciences Engineering, School of Biological Sciences, Nanyang Technological University, Singapore
- Singapore Centre for Environmental Life Sciences Engineering, Interdisciplinary Graduate School, Nanyang Technological University, Singapore
| | - Kimberly A. Kline
- Singapore Centre for Environmental Life Sciences Engineering, School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
37
|
Gram-Positive Uropathogens, Polymicrobial Urinary Tract Infection, and the Emerging Microbiota of the Urinary Tract. Microbiol Spectr 2017; 4. [PMID: 27227294 DOI: 10.1128/microbiolspec.uti-0012-2012] [Citation(s) in RCA: 225] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Gram-positive bacteria are a common cause of urinary-tract infection (UTI), particularly among individuals who are elderly, pregnant, or who have other risk factors for UTI. Here we review the epidemiology, virulence mechanisms, and host response to the most frequently isolated Gram-positive uropathogens: Staphylococcus saprophyticus, Enterococcus faecalis, and Streptococcus agalactiae. We also review several emerging, rare, misclassified, and otherwise underreported Gram-positive pathogens of the urinary tract including Aerococcus, Corynebacterium, Actinobaculum, and Gardnerella. The literature strongly suggests that urologic diseases involving Gram-positive bacteria may be easily overlooked due to limited culture-based assays typically utilized for urine in hospital microbiology laboratories. Some UTIs are polymicrobial in nature, often involving one or more Gram-positive bacteria. We herein review the risk factors and recent evidence for mechanisms of bacterial synergy in experimental models of polymicrobial UTI. Recent experimental data has demonstrated that, despite being cleared quickly from the bladder, some Gram-positive bacteria can impact pathogenic outcomes of co-infecting organisms. When taken together, the available evidence argues that Gram-positive bacteria are important uropathogens in their own right, but that some can be easily overlooked because they are missed by routine diagnostic methods. Finally, a growing body of evidence demonstrates that a surprising variety of fastidious Gram-positive bacteria may either reside in or be regularly exposed to the urinary tract and further suggests that their presence is widespread among women, as well as men. Experimental studies in this area are needed; however, there is a growing appreciation that the composition of bacteria found in the bladder could be a potentially important determinant in urologic disease, including susceptibility to UTI.
Collapse
|
38
|
Singh KV, Tran TT, Nannini EC, Tam VH, Arias CA, Murray BE. Efficacy of Ceftaroline against Methicillin-Susceptible Staphylococcus aureus Exhibiting the Cefazolin High-Inoculum Effect in a Rat Model of Endocarditis. Antimicrob Agents Chemother 2017; 61:e00324-17. [PMID: 28483961 PMCID: PMC5487651 DOI: 10.1128/aac.00324-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/04/2017] [Indexed: 12/16/2022] Open
Abstract
Certain Staphylococcus aureus strains exhibit an inoculum effect (InE) with cefazolin (CFZ) that has been associated with therapeutic failures in high-inoculum infections. We assessed the in vitro activities of ceftaroline (CPT), CFZ, and nafcillin (NAF) against 17 type A β-lactamase (βla)-producing, methicillin-susceptible S. aureus (MSSA) strains, including the previously reported TX0117, which exhibits the CFZ InE, and its βla-cured derivative, TX0117c. Additionally, we determined the pharmacokinetics of CPT in rats after single intramuscular doses of 20 and 40 mg/kg of body weight and evaluated the activities of CPT (40 mg/kg every 8 h [q8h]), CFZ, and NAF against TX0117 and TX0117c in a rat model of infective endocarditis. No InE was observed for CPT or NAF, whereas a marked InE was detected for CFZ (MIC, 8 to ≥128 μg/ml). CPT and NAF treatment against TX0117 resulted in mean bacterial counts of 2.3 and 2.1 log10 CFU/g in vegetations, respectively, compared to a mean of 5.9 log10 CFU/g in the CFZ-treated group (CPT and NAF versus CFZ, P = 0.001; CPT versus NAF, P = 0.9830). Both CFZ and CPT were efficacious against the βla-cured derivative, TX0117c, compared to time zero (t0) (P = <0.0001 and 0.0015, respectively). Our data reiterate the in vivo consequences of the CFZ InE and show that CPT is not affected by this phenomenon. CPT might be considered for high-inoculum infections caused by MSSA exhibiting the CFZ InE.
Collapse
Affiliation(s)
- Kavindra V Singh
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Center for Antimicrobial Resistance and Microbial Genomics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Truc T Tran
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Center for Antimicrobial Resistance and Microbial Genomics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Esteban C Nannini
- Division of Infectious Diseases, School of Medicine, Universidad Nacional de Rosario,. Instituto de Inmunología Clínica y Experimental Rosario (IDICER), CONICET, Rosario, Argentina
| | - Vincent H Tam
- College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Cesar A Arias
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Microbiology and Molecular Genetics, University of Texas Medical School at Houston, Houston, Texas, USA
- Molecular Genetics and Antimicrobial Resistance Unit, Universidad El Bosque, Bogota, Colombia
- Center for Antimicrobial Resistance and Microbial Genomics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Barbara E Murray
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Microbiology and Molecular Genetics, University of Texas Medical School at Houston, Houston, Texas, USA
- Center for Antimicrobial Resistance and Microbial Genomics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| |
Collapse
|
39
|
Abstract
Urinary tract infections (UTI) are among the most common bacterial infections in humans, affecting millions of people every year. UTI cause significant morbidity in women throughout their lifespan, in infant boys, in older men, in individuals with underlying urinary tract abnormalities, and in those that require long-term urethral catheterization, such as patients with spinal cord injuries or incapacitated individuals living in nursing homes. Serious sequelae include frequent recurrences, pyelonephritis with sepsis, renal damage in young children, pre-term birth, and complications of frequent antimicrobial use including high-level antibiotic resistance and Clostridium difficile colitis. Uropathogenic E. coli (UPEC) cause the vast majority of UTI, but less common pathogens such as Enterococcus faecalis and other enterococci frequently take advantage of an abnormal or catheterized urinary tract to cause opportunistic infections. While antibiotic therapy has historically been very successful in controlling UTI, the high rate of recurrence remains a major problem, and many individuals suffer from chronically recurring UTI, requiring long-term prophylactic antibiotic regimens to prevent recurrent UTI. Furthermore, the global emergence of multi-drug resistant UPEC in the past ten years spotlights the need for alternative therapeutic and preventative strategies to combat UTI, including anti-infective drug therapies and vaccines. In this chapter, we review recent advances in the field of UTI pathogenesis, with an emphasis on the identification of promising drug and vaccine targets. We then discuss the development of new UTI drugs and vaccines, highlighting the challenges these approaches face and the need for a greater understanding of urinary tract mucosal immunity.
Collapse
|
40
|
Role of the Emp Pilus Subunits of Enterococcus faecium in Biofilm Formation, Adherence to Host Extracellular Matrix Components, and Experimental Infection. Infect Immun 2016; 84:1491-1500. [PMID: 26930703 DOI: 10.1128/iai.01396-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/20/2016] [Indexed: 11/20/2022] Open
Abstract
Enterococcus faecium is an important cause of hospital-associated infections, including urinary tract infections (UTIs), bacteremia, and infective endocarditis. Pili have been shown to play a role in the pathogenesis of Gram-positive bacteria, including E. faecium We previously demonstrated that a nonpiliated ΔempABC::cat derivative of E. faecium TX82 was attenuated in biofilm formation and in a UTI model. Here, we studied the contributions of the individual pilus subunits EmpA, EmpB, and EmpC to pilus architecture, biofilm formation, adherence to extracellular matrix (ECM) proteins, and infection. We identified EmpA as the tip of the pili and found that deletion of empA reduced biofilm formation to the same level as deletion of the empABC operon, a phenotype that was restored by reconstituting in situ the empA gene. Deletion of empB also caused a reduction in biofilm, while EmpC was found to be dispensable. Significant reductions in adherence to fibrinogen and collagen type I were observed with deletion of empA and empB, while deletion of empC had no adherence defect. Furthermore, we showed that each deletion mutant was significantly attenuated in comparison to the isogenic parental strain, TX82, in a mixed-inoculum UTI model (P < 0.001 to 0.048), that reconstitution of empA restored virulence in the UTI model, and that deletion of empA also resulted in attenuation in an infective endocarditis model (P = 0.0088). Our results indicate that EmpA and EmpB, but not EmpC, contribute to biofilm and adherence to ECM proteins; however, all the Emp pilins are important for E. faecium to cause infection in the urinary tract.
Collapse
|
41
|
Spaulding CN, Hultgren SJ. Adhesive Pili in UTI Pathogenesis and Drug Development. Pathogens 2016; 5:E30. [PMID: 26999218 PMCID: PMC4810151 DOI: 10.3390/pathogens5010030] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 02/15/2016] [Accepted: 03/07/2016] [Indexed: 01/08/2023] Open
Abstract
Urinary tract infections (UTIs) are one of the most common bacterial infections, affecting 150 million people each year worldwide. High recurrence rates and increasing antimicrobial resistance among uropathogens are making it imperative to develop alternative strategies for the treatment and prevention of this common infection. In this Review, we discuss how understanding the: (i) molecular and biophysical basis of host-pathogen interactions; (ii) consequences of the molecular cross-talk at the host pathogen interface in terms of disease progression; and (iii) pathophysiology of UTIs is leading to efforts to translate this knowledge into novel therapeutics to treat and prevent these infections.
Collapse
Affiliation(s)
- Caitlin N Spaulding
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Scott J Hultgren
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
42
|
La Rosa SL, Montealegre MC, Singh KV, Murray BE. Enterococcus faecalis Ebp pili are important for cell-cell aggregation and intraspecies gene transfer. MICROBIOLOGY-SGM 2016; 162:798-802. [PMID: 26967674 DOI: 10.1099/mic.0.000276] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Enterococcus faecalis is an opportunistic pathogen that ranks among the leading causes of biofilm-associated infections. We previously demonstrated that the endocarditis- and biofilm-associated pili (Ebp) of E. faecalis play a major role in biofilm formation, adherence to abiotic surfaces and experimental infections. In this study, derivatives of E. faecalis strain OG1 were engineered to further characterize functions of Ebp pili. Loss of pili resulted in a 36-fold decrease in the number of closely associated cells when OG1RFΔebpABC was mixed with OG1SSpΔebpABC, compared with mixing the Ebp+ parental strains. In addition, using the Ebp+ parental strains as donor and recipient, we found a statistically significant increase (280-360 %, P < 0.05) in the frequency of plasmid transfer versus using Ebp- mutants in the conjugation experiments. These results demonstrate a previously unrecognized role of Ebp pili, namely, as important contributors to microscale cell aggregation and horizontal spread of genetic material.
Collapse
Affiliation(s)
- Sabina Leanti La Rosa
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Health Science Center,Houston, Texas,USA
| | - Maria Camila Montealegre
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Health Science Center,Houston, Texas,USA.,Department of Microbiology and Molecular Genetics, University of Texas Health Science Center,Houston, Texas,USA
| | - Kavindra V Singh
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Health Science Center,Houston, Texas,USA.,Center for the Study of Emerging and Re-emerging Pathogens, University of Texas Health Science Center,Houston, Texas,USA
| | - Barbara E Murray
- Center for the Study of Emerging and Re-emerging Pathogens, University of Texas Health Science Center,Houston, Texas,USA.,Department of Microbiology and Molecular Genetics, University of Texas Health Science Center,Houston, Texas,USA.,Department of Internal Medicine, Division of Infectious Diseases, University of Texas Health Science Center,Houston, Texas,USA
| |
Collapse
|
43
|
Chahales P, Thanassi DG. Structure, Function, and Assembly of Adhesive Organelles by Uropathogenic Bacteria. Microbiol Spectr 2015; 3:10.1128/microbiolspec.UTI-0018-2013. [PMID: 26542038 PMCID: PMC4638162 DOI: 10.1128/microbiolspec.uti-0018-2013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Indexed: 01/02/2023] Open
Abstract
Bacteria assemble a wide range of adhesive proteins, termed adhesins, to mediate binding to receptors and colonization of surfaces. For pathogenic bacteria, adhesins are critical for early stages of infection, allowing the bacteria to initiate contact with host cells, colonize different tissues, and establish a foothold within the host. The adhesins expressed by a pathogen are also critical for bacterial-bacterial interactions and the formation of bacterial communities, including biofilms. The ability to adhere to host tissues is particularly important for bacteria that colonize sites such as the urinary tract, where the flow of urine functions to maintain sterility by washing away non-adherent pathogens. Adhesins vary from monomeric proteins that are directly anchored to the bacterial surface to polymeric, hair-like fibers that extend out from the cell surface. These latter fibers are termed pili or fimbriae, and were among the first identified virulence factors of uropathogenic Escherichia coli. Studies since then have identified a range of both pilus and non-pilus adhesins that contribute to bacterial colonization of the urinary tract, and have revealed molecular details of the structures, assembly pathways, and functions of these adhesive organelles. In this review, we describe the different types of adhesins expressed by both Gram-negative and Gram-positive uropathogens, what is known about their structures, how they are assembled on the bacterial surface, and the functions of specific adhesins in the pathogenesis of urinary tract infections.
Collapse
Affiliation(s)
- Peter Chahales
- Center for Infectious Diseases and Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY 11794
| | - David G Thanassi
- Center for Infectious Diseases and Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY 11794
| |
Collapse
|
44
|
The fibronectin-binding protein Fnm contributes to adherence to extracellular matrix components and virulence of Enterococcus faecium. Infect Immun 2015; 83:4653-61. [PMID: 26371130 DOI: 10.1128/iai.00885-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/10/2015] [Indexed: 12/29/2022] Open
Abstract
The interaction between bacteria and fibronectin is believed to play an important role in the pathogenicity of clinically important Gram-positive cocci. In the present study, we identified a gene encoding a predicted fibronectin-binding protein of Enterococcus faecium (fnm), a homologue of Streptococcus pneumoniae pavA, in the genomes of E. faecium strain TX82 and all other sequenced E. faecium isolates. Full-length recombinant Fnm from strain TX82 bound to immobilized fibronectin in a concentration-dependent manner and also appeared to bind collagen type V and laminin, but not other proteins, such as transferrin, heparin, bovine serum albumin, mucin, or collagen IV. We demonstrated that the N-terminal fragment of Fnm is required for full fibronectin binding, since truncation of this region caused a 2.4-fold decrease (P < 0.05) in the adhesion of E. faecium TX82 to fibronectin. Deletion of fnm resulted in a significant reduction (P < 0.001) in the ability of the mutant, TX6128, to bind fibronectin relative to that of the wild-type strain; in situ reconstitution of fnm in the deletion mutant strain restored adherence. In addition, the Δfnm mutant was highly attenuated relative to TX82 (P ≤ 0.0001) in a mixed-inoculum rat endocarditis model. Taken together, these results demonstrate that Fnm affects the adherence of E. faecium to fibronectin and is important in the pathogenesis of experimental endocarditis.
Collapse
|
45
|
Frank KL, Vergidis P, Brinkman CL, Greenwood Quaintance KE, Barnes AMT, Mandrekar JN, Schlievert PM, Dunny GM, Patel R. Evaluation of the Enterococcus faecalis Biofilm-Associated Virulence Factors AhrC and Eep in Rat Foreign Body Osteomyelitis and In Vitro Biofilm-Associated Antimicrobial Resistance. PLoS One 2015; 10:e0130187. [PMID: 26076451 PMCID: PMC4467866 DOI: 10.1371/journal.pone.0130187] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/18/2015] [Indexed: 02/01/2023] Open
Abstract
Enterococcus faecalis can cause healthcare-associated biofilm infections, including those of orthopedic devices. Treatment of enterococcal prosthetic joint infection is difficult, in part, due to biofilm-associated antimicrobial resistance. We previously showed that the E. faecalis OG1RF genes ahrC and eep are in vitro biofilm determinants and virulence factors in animal models of endocarditis and catheter-associated urinary tract infection. In this study, we evaluated the role of these genes in a rat acute foreign body osteomyelitis model and in in vitro biofilm-associated antimicrobial resistance. Osteomyelitis was established for one week following the implantation of stainless steel orthopedic wires inoculated with E. faecalis strains OG1RF, ΩahrC, and ∆eep into the proximal tibiae of rats. The median bacterial loads recovered from bones and wires did not differ significantly between the strains at multiple inoculum concentrations. We hypothesize that factors present at the infection site that affect biofilm formation, such as the presence or absence of shear force, may account for the differences in attenuation in the various animal models we have used to study the ΩahrC and ∆eep strains. No differences among the three strains were observed in the planktonic and biofilm antimicrobial susceptibilities to ampicillin, vancomycin, daptomycin, linezolid, and tetracycline. These findings suggest that neither ahrC nor eep directly contribute to E. faecalis biofilm-associated antimicrobial resistance. Notably, the experimental evidence that the biofilm attachment mutant ΩahrC displays biofilm-associated antimicrobial resistance suggests that surface colonization alone is sufficient for E. faecalis cells to acquire the biofilm antimicrobial resistance phenotype.
Collapse
Affiliation(s)
- Kristi L. Frank
- Department of Microbiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Paschalis Vergidis
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Cassandra L. Brinkman
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kerryl E. Greenwood Quaintance
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Aaron M. T. Barnes
- Department of Microbiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jayawant N. Mandrekar
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Patrick M. Schlievert
- Department of Microbiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Gary M. Dunny
- Department of Microbiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
46
|
Flores-Mireles AL, Pinkner JS, Caparon MG, Hultgren SJ. EbpA vaccine antibodies block binding of Enterococcus faecalis to fibrinogen to prevent catheter-associated bladder infection in mice. Sci Transl Med 2015; 6:254ra127. [PMID: 25232179 DOI: 10.1126/scitranslmed.3009384] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Enterococci bacteria are a frequent cause of catheter-associated urinary tract infections, the most common type of hospital-acquired infection. Treatment has become increasingly challenging because of the emergence of multiantibiotic-resistant enterococcal strains and their ability to form biofilms on catheters. We identified and targeted a critical step in biofilm formation and developed a vaccine that prevents catheter-associated urinary tract infections in mice. In the murine model, formation of catheter-associated biofilms by Enterococcus faecalis depends on EbpA, which is the minor subunit at the tip of a heteropolymeric surface fiber known as the endocarditis- and biofilm-associated pilus (Ebp). We show that EbpA is an adhesin that mediates bacterial attachment to host fibrinogen, which is released and deposited on catheters after introduction of the catheter into the mouse bladder. Fibrinogen-binding activity resides in the amino-terminal domain of EbpA (EbpA(NTD)), and vaccination with EbpA and EbpA(NTD), but not its carboxyl-terminal domain or other Ebp subunits, inhibited biofilm formation in vivo and protected against catheter-associated urinary tract infection. Analyses in vitro demonstrated that protection was associated with a serum antibody response that blocked EbpA binding to fibrinogen and the formation of a fibrinogen-dependent biofilm on catheters. This approach may provide a new strategy for the prevention of catheter-associated urinary tract infections.
Collapse
Affiliation(s)
- Ana L Flores-Mireles
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Jerome S Pinkner
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Michael G Caparon
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110-1093, USA.
| | - Scott J Hultgren
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110-1093, USA.
| |
Collapse
|
47
|
Carey AJ, Tan CK, Ipe DS, Sullivan MJ, Cripps AW, Schembri MA, Ulett GC. Urinary tract infection of mice to model human disease: Practicalities, implications and limitations. Crit Rev Microbiol 2015; 42:780-99. [PMID: 26006172 DOI: 10.3109/1040841x.2015.1028885] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections in humans. Murine models of human UTI are vital experimental tools that have helped to elucidate UTI pathogenesis and advance knowledge of potential treatment and infection prevention strategies. Fundamentally, several variables are inherent in different murine models, and understanding the limitations of these variables provides an opportunity to understand how models may be best applied to research aimed at mimicking human disease. In this review, we discuss variables inherent in murine UTI model studies and how these affect model usage, data analysis and data interpretation. We examine recent studies that have elucidated UTI host-pathogen interactions from the perspective of gene expression, and review new studies of biofilm and UTI preventative approaches. We also consider potential standards for variables inherent in murine UTI models and discuss how these might expand the utility of models for mimicking human disease and uncovering new aspects of pathogenesis.
Collapse
Affiliation(s)
- Alison J Carey
- a Menzies Health Institute Queensland & School of Medical Sciences, Griffith University , Gold Coast , Australia
| | - Chee K Tan
- a Menzies Health Institute Queensland & School of Medical Sciences, Griffith University , Gold Coast , Australia
| | - Deepak S Ipe
- a Menzies Health Institute Queensland & School of Medical Sciences, Griffith University , Gold Coast , Australia
| | - Matthew J Sullivan
- a Menzies Health Institute Queensland & School of Medical Sciences, Griffith University , Gold Coast , Australia
| | - Allan W Cripps
- b Menzies Health Institute Queensland, Griffith University , Gold Coast , Australia , and
| | - Mark A Schembri
- c School of Chemistry and Molecular Biosciences, University of Queensland , Brisbane , Australia
| | - Glen C Ulett
- a Menzies Health Institute Queensland & School of Medical Sciences, Griffith University , Gold Coast , Australia
| |
Collapse
|
48
|
The Enterococcus faecalis EbpA Pilus Protein: Attenuation of Expression, Biofilm Formation, and Adherence to Fibrinogen Start with the Rare Initiation Codon ATT. mBio 2015; 6:e00467-15. [PMID: 26015496 PMCID: PMC4447247 DOI: 10.1128/mbio.00467-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The endocarditis and biofilm-associated pili (Ebp) are important in Enterococcus faecalis pathogenesis, and the pilus tip, EbpA, has been shown to play a major role in pilus biogenesis, biofilm formation, and experimental infections. Based on in silico analyses, we previously predicted that ATT is the EbpA translational start codon, not the ATG codon, 120 bp downstream of ATT, which is annotated as the translational start. ATT is rarely used to initiate protein synthesis, leading to our hypothesis that this codon participates in translational regulation of Ebp production. To investigate this possibility, site-directed mutagenesis was used to introduce consecutive stop codons in place of two lysines at positions 5 and 6 from the ATT, to replace the ATT codon in situ with ATG, and then to revert this ATG to ATT; translational fusions of ebpA to lacZ were also constructed to investigate the effect of these start codons on translation. Our results showed that the annotated ATG does not start translation of EbpA, implicating ATT as the start codon; moreover, the presence of ATT, compared to the engineered ATG, resulted in significantly decreased EbpA surface display, attenuated biofilm, and reduced adherence to fibrinogen. Corroborating these findings, the translational fusion with the native ATT as the initiation codon showed significantly decreased expression of β-galactosidase compared to the construct with ATG in place of ATT. Thus, these results demonstrate that the rare initiation codon of EbpA negatively regulates EbpA surface display and negatively affects Ebp-associated functions, including biofilm and adherence to fibrinogen. IMPORTANCE Enterococcus faecalis is among the leading causes of serious infections in the hospital setting, and the endocarditis and biofilm-associated pili (Ebp) have been shown to play significant roles in E. faecalis pathogenesis. Understanding the regulation of virulence is important for the development of new approaches to counteract multidrug-resistant pathogens. We previously predicted that ATT, which has been reported to start protein synthesis only in rare instances, is the most likely translational start codon of EbpA in E. faecalis. Here, we demonstrate that ATT is the initiation codon of EbpA and, relative to a constructed ATG start codon, results in smaller amounts of EbpA on the surface of the cells, attenuating biofilm formation and fibrinogen adherence, phenotypes associated with the ability of E. faecalis to cause infections. This provides the first example of pilus regulation through the use of an ATT initiation codon.
Collapse
|
49
|
Gozalan A, Coskun-Ari FF, Ozdem B, Unaldi O, Celikbilek N, Kirca F, Aydogan S, Muderris T, Guven T, Acikgoz ZC, Durmaz R. Molecular characterization of vancomycin-resistant Enterococcus faecium strains isolated from carriage and clinical samples in a tertiary hospital, Turkey. J Med Microbiol 2015; 64:759-766. [PMID: 25976005 DOI: 10.1099/jmm.0.000088] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This study aimed to determine the presence of vancomycin resistance (vanA and vanB) and virulence genes (esp, asa1, gelE, ace, hyl, cylA, cpd and ebpA) in vancomycin-resistant Enterococcus faecium (VREfm) strains and to analyse the clonal relationships among the strains. E. faecium strains were identified from rectal and clinical specimens by biochemical tests and the API-20 Strep kit. Susceptibility testing was performed using disc-diffusion and broth-dilution methods. PFGE was used for molecular typing of the VREfm strains. The vancomycin resistance and virulence genes were amplified by two-step multiplex PCR. All 55 VREfm isolates were resistant to penicillin G, ampicillin and high-level gentamicin but were susceptible to quinupristin/dalfopristin and linezolid. Multiplex PCR analysis indicated that all isolates harboured vanA and that 41 (75 %) were positive for virulence genes. The esp gene was the most common virulence factor and was detected in nine (41 %) invasive and 32 (96.7 %) non-invasive isolates. Multiple virulence genes were observed only in two non-invasive isolates; one harboured esp and ebpA and the other harboured esp, ebpA, asa1, gelE and cpd. PFGE typing yielded 16 different types, seven of which were clusters with two to 14 strains each. The clustering rates of the rectal swab, blood and urine isolates were 72.7 %, 61.5 % and 87.5 %, respectively. The genetic similarity observed among the VREfm isolates indicated cross-transmission in the hospital. Further studies on the virulence factors present in the strains might provide insight into the acquisition of these traits and their contribution to increased prevalence of VREfm.
Collapse
Affiliation(s)
- Aysegul Gozalan
- Atatürk Education and Research Hospital, Medical Microbiology Laboratory, Ankara, Turkey
| | - Fatma Filiz Coskun-Ari
- National Molecular Microbiology Reference Laboratory, Public Health Institution of Turkey, Ankara, Turkey
| | - Birsen Ozdem
- Atatürk Education and Research Hospital, Medical Microbiology Laboratory, Ankara, Turkey
| | - Ozlem Unaldi
- National Molecular Microbiology Reference Laboratory, Public Health Institution of Turkey, Ankara, Turkey
| | - Nevreste Celikbilek
- Atatürk Education and Research Hospital, Medical Microbiology Laboratory, Ankara, Turkey
| | - Fisun Kirca
- Atatürk Education and Research Hospital, Medical Microbiology Laboratory, Ankara, Turkey
| | - Sibel Aydogan
- Atatürk Education and Research Hospital, Medical Microbiology Laboratory, Ankara, Turkey
| | - Tuba Muderris
- Atatürk Education and Research Hospital, Medical Microbiology Laboratory, Ankara, Turkey
| | - Tumer Guven
- Yildirim Beyazit University, Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ankara, Turkey
| | - Ziya Cibali Acikgoz
- Atatürk Education and Research Hospital, Medical Microbiology Laboratory, Ankara, Turkey.,Yildirim Beyazit University, Faculty of Medicine, Department of Microbiology, Ankara, Turkey
| | - Riza Durmaz
- National Molecular Microbiology Reference Laboratory, Public Health Institution of Turkey, Ankara, Turkey.,Yildirim Beyazit University, Faculty of Medicine, Department of Microbiology, Ankara, Turkey
| |
Collapse
|
50
|
Zischka M, Künne CT, Blom J, Wobser D, Sakιnç T, Schmidt-Hohagen K, Dabrowski PW, Nitsche A, Hübner J, Hain T, Chakraborty T, Linke B, Goesmann A, Voget S, Daniel R, Schomburg D, Hauck R, Hafez HM, Tielen P, Jahn D, Solheim M, Sadowy E, Larsen J, Jensen LB, Ruiz-Garbajosa P, Quiñones Pérez D, Mikalsen T, Bender J, Steglich M, Nübel U, Witte W, Werner G. Comprehensive molecular, genomic and phenotypic analysis of a major clone of Enterococcus faecalis MLST ST40. BMC Genomics 2015; 16:175. [PMID: 25887115 PMCID: PMC4374294 DOI: 10.1186/s12864-015-1367-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/20/2015] [Indexed: 11/28/2022] Open
Abstract
Background Enterococcus faecalis is a multifaceted microorganism known to act as a beneficial intestinal commensal bacterium. It is also a dreaded nosocomial pathogen causing life-threatening infections in hospitalised patients. Isolates of a distinct MLST type ST40 represent the most frequent strain type of this species, distributed worldwide and originating from various sources (animal, human, environmental) and different conditions (colonisation/infection). Since enterococci are known to be highly recombinogenic we determined to analyse the microevolution and niche adaptation of this highly distributed clonal type. Results We compared a set of 42 ST40 isolates by assessing key molecular determinants, performing whole genome sequencing (WGS) and a number of phenotypic assays including resistance profiling, formation of biofilm and utilisation of carbon sources. We generated the first circular closed reference genome of an E. faecalis isolate D32 of animal origin and compared it with the genomes of other reference strains. D32 was used as a template for detailed WGS comparisons of high-quality draft genomes of 14 ST40 isolates. Genomic and phylogenetic analyses suggest a high level of similarity regarding the core genome, also demonstrated by similar carbon utilisation patterns. Distribution of known and putative virulence-associated genes did not differentiate between ST40 strains from a commensal and clinical background or an animal or human source. Further analyses of mobile genetic elements (MGE) revealed genomic diversity owed to: (1) a modularly structured pathogenicity island; (2) a site-specifically integrated and previously unknown genomic island of 138 kb in two strains putatively involved in exopolysaccharide synthesis; and (3) isolate-specific plasmid and phage patterns. Moreover, we used different cell-biological and animal experiments to compare the isolate D32 with a closely related ST40 endocarditis isolate whose draft genome sequence was also generated. D32 generally showed a greater capacity of adherence to human cell lines and an increased pathogenic potential in various animal models in combination with an even faster growth in vivo (not in vitro). Conclusion Molecular, genomic and phenotypic analysis of representative isolates of a major clone of E. faecalis MLST ST40 revealed new insights into the microbiology of a commensal bacterium which can turn into a conditional pathogen. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1367-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melanie Zischka
- Division of Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Burgstr. 37, D-38855, Wernigerode, Germany. .,Present address: Institute for Pathology, Hannover Medical School (MHH), Hannover, Germany.
| | - Carsten T Künne
- Functional Genomics of Bacterial Pathogens, Institute for Medical Microbiology, Justus Liebig University Giessen and German Center for Infection Research (DZIF), Partner site Giessen-Marburg-Langen, Campus Giessen, Giessen, Germany. .,Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| | - Jochen Blom
- Center for Biotechnology (CeBiTec)/University of Bielefeld, Bielefeld, Germany. .,Institute for Bioinformatics and Systems Biology, Justus Liebig University Giessen, Giessen, Germany.
| | - Dominique Wobser
- Division of Infectious Diseases, Department of Medicine, University Hospital Freiburg, Freiburg, Germany.
| | - Türkân Sakιnç
- Division of Infectious Diseases, Department of Medicine, University Hospital Freiburg, Freiburg, Germany.
| | - Kerstin Schmidt-Hohagen
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany.
| | - P Wojtek Dabrowski
- Robert Koch Institute, ZBS 1 Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens, Berlin, Germany.
| | - Andreas Nitsche
- Robert Koch Institute, ZBS 1 Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens, Berlin, Germany.
| | - Johannes Hübner
- Division of Infectious Diseases, Department of Medicine, University Hospital Freiburg, Freiburg, Germany. .,Division of Pediatric Infectious Diseases, Hauner Children's Hospital, Ludwig-Maximilians University Munich, Munich, Germany.
| | - Torsten Hain
- Functional Genomics of Bacterial Pathogens, Institute for Medical Microbiology, Justus Liebig University Giessen and German Center for Infection Research (DZIF), Partner site Giessen-Marburg-Langen, Campus Giessen, Giessen, Germany.
| | - Trinad Chakraborty
- Functional Genomics of Bacterial Pathogens, Institute for Medical Microbiology, Justus Liebig University Giessen and German Center for Infection Research (DZIF), Partner site Giessen-Marburg-Langen, Campus Giessen, Giessen, Germany.
| | - Burkhard Linke
- Center for Biotechnology (CeBiTec)/University of Bielefeld, Bielefeld, Germany. .,Institute for Bioinformatics and Systems Biology, Justus Liebig University Giessen, Giessen, Germany.
| | - Alexander Goesmann
- Center for Biotechnology (CeBiTec)/University of Bielefeld, Bielefeld, Germany. .,Institute for Bioinformatics and Systems Biology, Justus Liebig University Giessen, Giessen, Germany.
| | - Sonja Voget
- Goettingen Genomics Laboratory, Georg August University, Goettingen, Germany.
| | - Rolf Daniel
- Goettingen Genomics Laboratory, Georg August University, Goettingen, Germany.
| | - Dietmar Schomburg
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany.
| | - Rüdiger Hauck
- Department of Veterinary Medicine, Institute for Poultry Diseases, Free University Berlin, Berlin, Germany.
| | - Hafez M Hafez
- Department of Veterinary Medicine, Institute for Poultry Diseases, Free University Berlin, Berlin, Germany.
| | - Petra Tielen
- Institute for Microbiology, Technische Universität Braunschweig, Braunschweig, Germany.
| | - Dieter Jahn
- Institute for Microbiology, Technische Universität Braunschweig, Braunschweig, Germany.
| | - Margrete Solheim
- Laboratory of Microbial Gene Technology and Food Microbiology, The Norwegian University of Life Sciences, Ås, Norway.
| | - Ewa Sadowy
- National Medicines Institute, Warsaw, Poland.
| | | | - Lars B Jensen
- Division of Microbiology, National Food Institute, Danish Technical University, Copenhagen, Denmark.
| | | | - Dianelys Quiñones Pérez
- Instituto de Medicina Tropical Pedro Kourí, Servicio de Bacteriología-Micología, La Habana, Cuba.
| | - Theresa Mikalsen
- Department of Medical Biology, Faculty of Health Sciences, Research Group for Host Microbe Interactions, University of Tromsø, Tromsø, Norway.
| | - Jennifer Bender
- Division of Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Burgstr. 37, D-38855, Wernigerode, Germany.
| | - Matthias Steglich
- Division of Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Burgstr. 37, D-38855, Wernigerode, Germany.
| | - Ulrich Nübel
- Division of Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Burgstr. 37, D-38855, Wernigerode, Germany. .,Leibniz-Institut DSMZ - Deutsche Sammlung von Mikrorganismen und Zellkulturen GmbH, Braunschweig, Germany.
| | - Wolfgang Witte
- Division of Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Burgstr. 37, D-38855, Wernigerode, Germany.
| | - Guido Werner
- Division of Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Burgstr. 37, D-38855, Wernigerode, Germany.
| |
Collapse
|