1
|
Choi D, Ryu S, Kong M. Phage-derived proteins: Advancing food safety through biocontrol and detection of foodborne pathogens. Compr Rev Food Sci Food Saf 2025; 24:e70124. [PMID: 39898971 DOI: 10.1111/1541-4337.70124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/01/2025] [Accepted: 01/10/2025] [Indexed: 02/04/2025]
Abstract
The emergence of antimicrobial-resistant foodborne pathogens poses a continuous health risk and economic burden as they can easily spread through contaminated food. Therefore, the demand for new antimicrobial agents to address this problem is steadily increasing. Similarly, the development of rapid, sensitive, and accurate pathogen detection tools is a prerequisite for ensuring food safety. Phage-derived proteins have become innovative tools for combating these pathogens because of their potent antimicrobial activity and host specificity. Phage proteins are relatively free from regulation compared to phages per se, and there are no concerns about the transduction of harmful genes. With recent progress in next-generation sequencing technology, the analysis of phage genomes has become more accessible, and numerous phage proteins with potential for biocontrol and detection have been identified. This review provides a comprehensive overview of phage protein research on food safety from 2006 to the present, a pivotal period marked by the certification of phages as Generally Recognized As Safe (GRAS). Emphasizing recent advancements, we investigated the diverse applications of various phage proteins for biocontrol and detection purposes. While highlighting the successful implementation of these proteins, we also address the current bottlenecks and propose strategies to overcome these challenges. By summarizing the current state of research on phage-derived proteins, this review contributes to a deeper understanding of their potential as effective antimicrobial agents and tools for detecting foodborne pathogens.
Collapse
Affiliation(s)
- Dahee Choi
- Department of Food Science and Biotechnology, Institute of Food and Biotechnology, Seoul National University of Science and Technology, Seoul, South Korea
| | - Sangryeol Ryu
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| | - Minsuk Kong
- Department of Food Science and Biotechnology, Institute of Food and Biotechnology, Seoul National University of Science and Technology, Seoul, South Korea
| |
Collapse
|
2
|
Yang P, Li J, Ma X, Hu N, Song Z, Chen B, Li S. Novel delivery systems for phages and lysins in the topical management of wound infections: a narrative review. Front Microbiol 2025; 16:1526096. [PMID: 39931378 PMCID: PMC11808012 DOI: 10.3389/fmicb.2025.1526096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025] Open
Abstract
Currently, multidrug-resistant (MDR) bacterial wound infections (WIs) are an extremely challenging clinical problem for physicians. Recently, compared to traditional single liquid delivery drugs, the study of five novel drug delivery systems (i.e., hydrogel, liposomes, electrospun fibers, nanoparticles and nanoemulsion) for phages and their encoded lysins in WI management has become a hot topic. To assess the current landscape of these emerging technologies, we conducted a comprehensive literature search across PubMed, Scopus and Web of Science up to July 2024, using terms such as "phage," "lysin," "wound," "hydrogel," "liposomes," "fibers," "nanoparticles," and "nanoemulsion." The criteria included original studies of five novel delivery systems for phages and lysins in WI management. The findings highlighted the positive effects of the five novel delivery systems for phages and lysins in WI management, significantly reducing wound bacterial populations, and accelerating healing at the injury site. However, the available literature on novel delivery systems for phages and lysins remains limited, particularly for lysins. In conclusion, the application of novel drug delivery systems for phages and lysins showed great potential in combating MDR bacterial WIs.
Collapse
Affiliation(s)
- Pan Yang
- Postdoctoral Research Station, Guangzhou Bay Area Institute of Biomedicine, Guangzhou, China
| | - Jing Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiumei Ma
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Nan Hu
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhangyong Song
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Bin Chen
- Postdoctoral Research Station, Guangzhou Bay Area Institute of Biomedicine, Guangzhou, China
| | - Shizhu Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Hülpüsch C, Rohayem R, Reiger M, Traidl-Hoffmann C. Exploring the skin microbiome in atopic dermatitis pathogenesis and disease modification. J Allergy Clin Immunol 2024; 154:31-41. [PMID: 38761999 DOI: 10.1016/j.jaci.2024.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 03/04/2024] [Accepted: 04/24/2024] [Indexed: 05/20/2024]
Abstract
Inflammatory skin diseases such as atopic eczema (atopic dermatitis [AD]) affect children and adults globally. In AD, the skin barrier is impaired on multiple levels. Underlying factors include genetic, chemical, immunologic, and microbial components. Increased skin pH in AD is part of the altered microbial microenvironment that promotes overgrowth of the skin microbiome with Staphylococcus aureus. The secretion of virulence factors, such as toxins and proteases, by S aureus further aggravates the skin barrier deficiency and additionally disrupts the balance of an already skewed immune response. Skin commensal bacteria, however, can inhibit the growth and pathogenicity of S aureus through quorum sensing. Therefore, restoring a healthy skin microbiome could contribute to remission induction in AD. This review discusses direct and indirect approaches to targeting the skin microbiome through modulation of the skin pH; UV treatment; and use of prebiotics, probiotics, and postbiotics. Furthermore, exploratory techniques such as skin microbiome transplantation, ozone therapy, and phage therapy are discussed. Finally, we summarize the latest findings on disease and microbiome modification through targeted immunomodulatory systemic treatments and biologics. We believe that targeting the skin microbiome should be considered a crucial component of successful AD treatment in the future.
Collapse
Affiliation(s)
- Claudia Hülpüsch
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Institute of Environmental Medicine, Helmholtz Center Munich-German Research Center for Environmental Health, Augsburg, Germany; Christine-Kühne Center for Allergy Research and Education, Davos, Switzerland
| | - Robin Rohayem
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Christine-Kühne Center for Allergy Research and Education, Davos, Switzerland; Dermatology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Matthias Reiger
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Institute of Environmental Medicine, Helmholtz Center Munich-German Research Center for Environmental Health, Augsburg, Germany
| | - Claudia Traidl-Hoffmann
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Chair of Environmental Medicine, Technical University of Munich, Munich, Germany; Institute of Environmental Medicine, Helmholtz Center Munich-German Research Center for Environmental Health, Augsburg, Germany; Christine-Kühne Center for Allergy Research and Education, Davos, Switzerland; ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany.
| |
Collapse
|
4
|
Tham EH, Chia M, Riggioni C, Nagarajan N, Common JE, Kong HH. The skin microbiome in pediatric atopic dermatitis and food allergy. Allergy 2024; 79:1470-1484. [PMID: 38308490 PMCID: PMC11142881 DOI: 10.1111/all.16044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/03/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
The skin microbiome is an extensive community of bacteria, fungi, mites, viruses and archaea colonizing the skin. Fluctuations in the composition of the skin microbiome have been observed in atopic dermatitis (AD) and food allergy (FA), particularly in early life, established disease, and associated with therapeutics. However, AD is a multifactorial disease characterized by skin barrier aberrations modulated by genetics, immunology, and environmental influences, thus the skin microbiome is not the sole feature of this disease. Future research should focus on mechanistic understanding of how early-life skin microbial shifts may influence AD and FA onset, to guide potential early intervention strategies or as microbial biomarkers to identify high-risk infants who may benefit from possible microbiome-based biotherapeutic strategies. Harnessing skin microbes as AD biotherapeutics is an emerging field, but more work is needed to investigate whether this approach can lead to sustained clinical responses.
Collapse
Affiliation(s)
- Elizabeth Huiwen Tham
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System (NUHS), Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| | - Minghao Chia
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Carmen Riggioni
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| | - Niranjan Nagarajan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - John E.A. Common
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Heidi H. Kong
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
5
|
Ho M, Nguyen HN, Van Hoang M, Bui TTT, Vu BQ, Dinh THT, Vo HTM, Blaydon DC, Eldirany SA, Bunick CG, Bui CB. Altered skin microbiome, inflammation, and JAK/STAT signaling in Southeast Asian ichthyosis patients. Hum Genomics 2024; 18:38. [PMID: 38627868 PMCID: PMC11022333 DOI: 10.1186/s40246-024-00603-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 04/01/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Congenital ichthyosis (CI) is a collective group of rare hereditary skin disorders. Patients present with epidermal scaling, fissuring, chronic inflammation, and increased susceptibility to infections. Recently, there is increased interest in the skin microbiome; therefore, we hypothesized that CI patients likely exhibit an abnormal profile of epidermal microbes because of their various underlying skin barrier defects. Among recruited individuals of Southeast Asian ethnicity, we performed skin meta-genomics (i.e., whole-exome sequencing to capture the entire multi-kingdom profile, including fungi, protists, archaea, bacteria, and viruses), comparing 36 CI patients (representing seven subtypes) with that of 15 CI age-and gender-matched controls who had no family history of CI. RESULTS This case-control study revealed 20 novel and 31 recurrent pathogenic variants. Microbiome meta-analysis showed distinct microbial populations, decreases in commensal microbiota, and higher colonization by pathogenic species associated with CI; these were correlated with increased production of inflammatory cytokines and Th17- and JAK/STAT-signaling pathways in peripheral blood mononuclear cells. In the wounds of CI patients, we identified specific changes in microbiota and alterations in inflammatory pathways, which are likely responsible for impaired wound healing. CONCLUSIONS Together, this research enhances our understanding of the microbiological, immunological, and molecular properties of CI and should provide critical information for improving therapeutic management of CI patients.
Collapse
Affiliation(s)
- Minh Ho
- Department of Dermatology and Program in Translational Biomedicine, Yale University, New Haven, CT, USA
| | - Huynh-Nga Nguyen
- Microbial Genomics DNA Medical Technology, Ho Chi Minh, Vietnam
- Department of Biology, Dalat University, Da Lat, Lam Dong, Vietnam
| | - Minh Van Hoang
- Vietnam Vascular Anomalies Center, University Medical Center 3, Ho Chi Minh, Vietnam
| | | | - Bao-Quoc Vu
- Microbial Genomics DNA Medical Technology, Ho Chi Minh, Vietnam
- Department of Biology, Dalat University, Da Lat, Lam Dong, Vietnam
| | - Truc Huong Thi Dinh
- Department of Pathophysiology and Immunology, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Hoa Thi My Vo
- Oxford University Clinical Research Unit, Ho Chi Minh, Vietnam
| | - Diana C Blaydon
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London, UK
| | - Sherif A Eldirany
- Department of Dermatology and Program in Translational Biomedicine, Yale University, New Haven, CT, USA
| | - Christopher G Bunick
- Department of Dermatology and Program in Translational Biomedicine, Yale University, New Haven, CT, USA.
| | - Chi-Bao Bui
- Microbial Genomics DNA Medical Technology, Ho Chi Minh, Vietnam.
- Department of Microbiology, City Children's Hospital, Ho Chi Minh, Vietnam.
- School of Medicine, Vietnam National University, Ho Chi Minh, Vietnam.
| |
Collapse
|
6
|
Liu K, Wang C, Zhou X, Guo X, Yang Y, Liu W, Zhao R, Song H. Bacteriophage therapy for drug-resistant Staphylococcus aureus infections. Front Cell Infect Microbiol 2024; 14:1336821. [PMID: 38357445 PMCID: PMC10864608 DOI: 10.3389/fcimb.2024.1336821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
Drug-resistant Staphylococcus aureus stands as a prominent pathogen in nosocomial and community-acquired infections, capable of inciting various infections at different sites in patients. This includes Staphylococcus aureus bacteremia (SaB), which exhibits a severe infection frequently associated with significant mortality rate of approximately 25%. In the absence of better alternative therapies, antibiotics is still the main approach for treating infections. However, excessive use of antibiotics has, in turn, led to an increase in antimicrobial resistance. Hence, it is imperative that new strategies are developed to control drug-resistant S. aureus infections. Bacteriophages are viruses with the ability to infect bacteria. Bacteriophages, were used to treat bacterial infections before the advent of antibiotics, but were subsequently replaced by antibiotics due to limited theoretical understanding and inefficient preparation processes at the time. Recently, phages have attracted the attention of many researchers again because of the serious problem of antibiotic resistance. This article provides a comprehensive overview of phage biology, animal models, diverse clinical case treatments, and clinical trials in the context of drug-resistant S. aureus phage therapy. It also assesses the strengths and limitations of phage therapy and outlines the future prospects and research directions. This review is expected to offer valuable insights for researchers engaged in phage-based treatments for drug-resistant S. aureus infections.
Collapse
Affiliation(s)
- Kaixin Liu
- College of Public Health, Zhengzhou University, Zhengzhou, China
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Chao Wang
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Xudong Zhou
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
- College of Public Health, China Medical University, Shenyang, China
| | - Xudong Guo
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Yi Yang
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Wanying Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Rongtao Zhao
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Hongbin Song
- College of Public Health, Zhengzhou University, Zhengzhou, China
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
- College of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
7
|
Sidbury R, Alikhan A, Bercovitch L, Cohen DE, Darr JM, Drucker AM, Eichenfield LF, Frazer-Green L, Paller AS, Schwarzenberger K, Silverberg JI, Singh AM, Wu PA, Davis DMR. Guidelines of care for the management of atopic dermatitis in adults with topical therapies. J Am Acad Dermatol 2023; 89:e1-e20. [PMID: 36641009 DOI: 10.1016/j.jaad.2022.12.029] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND New evidence has emerged since the 2014 guidelines that further informs the management of atopic dermatitis (AD) with topical therapies. These guidelines update the 2014 recommendations for management of AD with topical therapies. OBJECTIVE To provide evidence-based recommendations related to management of AD in adults using topical treatments. METHODS A multidisciplinary workgroup conducted a systematic review and applied the GRADE (Grading of Recommendations, Assessment, Development, and Evaluations) approach for assessing the certainty of evidence and formulating and grading recommendations. RESULTS The workgroup developed 12 recommendations on the management of AD in adults with topical therapies, including nonprescription agents and prescription topical corticosteroids (TCS), calcineurin inhibitors (TCIs), Janus kinase (JAK) inhibitors, phosphodiesterase-4 inhibitors (PDE-4), antimicrobials, and antihistamines. LIMITATIONS The pragmatic decision to limit the literature review to English-language randomized trials may have excluded data published in other languages and relevant long-term follow-up data. CONCLUSIONS Strong recommendations are made for the use of moisturizers, TCIs, TCS, and topical PDE-4 and JAK inhibitors. Conditional recommendations are made for the use of bathing and wet wrap therapy and against the use of topical antimicrobials, antiseptics, and antihistamines.
Collapse
Affiliation(s)
- Robert Sidbury
- Division of Dermatology, Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, Washington
| | - Ali Alikhan
- Department of Dermatology, Sutter Medical Foundation, Sacramento, California
| | - Lionel Bercovitch
- Department of Dermatology, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - David E Cohen
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, New York
| | - Jennifer M Darr
- Department of Pediatrics, National Jewish Health, Denver, Colorado
| | - Aaron M Drucker
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
| | - Lawrence F Eichenfield
- University of California San Diego and Division of Pediatric and Adolescent Dermatology, Rady Children's Hospital, San Diego, California
| | | | - Amy S Paller
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Jonathan I Silverberg
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Anne Marie Singh
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Peggy A Wu
- Department of Dermatology, University of California, Davis, Sacramento, California
| | - Dawn M R Davis
- Departments of Dermatology and Pediatrics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
8
|
Liu H, Hu Z, Li M, Yang Y, Lu S, Rao X. Therapeutic potential of bacteriophage endolysins for infections caused by Gram-positive bacteria. J Biomed Sci 2023; 30:29. [PMID: 37101261 PMCID: PMC10131408 DOI: 10.1186/s12929-023-00919-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/14/2023] [Indexed: 04/28/2023] Open
Abstract
Gram-positive (G+) bacterial infection is a great burden to both healthcare and community medical resources. As a result of the increasing prevalence of multidrug-resistant G+ bacteria such as methicillin-resistant Staphylococcus aureus (MRSA), novel antimicrobial agents must urgently be developed for the treatment of infections caused by G+ bacteria. Endolysins are bacteriophage (phage)-encoded enzymes that can specifically hydrolyze the bacterial cell wall and quickly kill bacteria. Bacterial resistance to endolysins is low. Therefore, endolysins are considered promising alternatives for solving the mounting resistance problem. In this review, endolysins derived from phages targeting G+ bacteria were classified based on their structural characteristics. The active mechanisms, efficacy, and advantages of endolysins as antibacterial drug candidates were summarized. Moreover, the remarkable potential of phage endolysins in the treatment of G+ bacterial infections was described. In addition, the safety of endolysins, challenges, and possible solutions were addressed. Notwithstanding the limitations of endolysins, the trends in development indicate that endolysin-based drugs will be approved in the near future. Overall, this review presents crucial information of the current progress involving endolysins as potential therapeutic agents, and it provides a guideline for biomaterial researchers who are devoting themselves to fighting against bacterial infections.
Collapse
Affiliation(s)
- He Liu
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China
| | - Zhen Hu
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China
| | - Mengyang Li
- Department of Microbiology, School of Medicine, Chongqing University, Chongqing, 400044, China
| | - Yi Yang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China
| | - Shuguang Lu
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
9
|
Eichenseher F, Herpers BL, Badoux P, Leyva-Castillo JM, Geha RS, van der Zwart M, McKellar J, Janssen F, de Rooij B, Selvakumar L, Röhrig C, Frieling J, Offerhaus M, Loessner MJ, Schmelcher M. Linker-Improved Chimeric Endolysin Selectively Kills Staphylococcus aureus In Vitro, on Reconstituted Human Epidermis, and in a Murine Model of Skin Infection. Antimicrob Agents Chemother 2022; 66:e0227321. [PMID: 35416713 PMCID: PMC9112974 DOI: 10.1128/aac.02273-21] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/16/2022] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus causes a broad spectrum of diseases in humans and animals. It is frequently associated with inflammatory skin disorders such as atopic dermatitis, where it aggravates symptoms. Treatment of S. aureus-associated skin infections with antibiotics is discouraged due to their broad-range deleterious effect on healthy skin microbiota and their ability to promote the development of resistance. Thus, novel S. aureus-specific antibacterial agents are desirable. We constructed two chimeric cell wall-lytic enzymes, Staphefekt SA.100 and XZ.700, which are composed of functional domains from the bacteriophage endolysin Ply2638 and the bacteriocin lysostaphin. Both enzymes specifically killed S. aureus and were inactive against commensal skin bacteria such as Staphylococcus epidermidis, with XZ.700 proving more active than SA.100 in multiple in vitro activity assays. When surface-attached mixed staphylococcal cultures were exposed to XZ.700 in a simplified microbiome model, the enzyme selectively removed S. aureus and retained S. epidermidis. Furthermore, XZ.700 did not induce resistance in S. aureus during repeated rounds of exposure to sublethal concentrations. Finally, we demonstrated that XZ.700 formulated as a cream is effective at killing S. aureus on reconstituted human epidermis and that an XZ.700-containing gel significantly reduces bacterial numbers compared to an untreated control in a mouse model of S. aureus-induced skin infection.
Collapse
Affiliation(s)
- Fritz Eichenseher
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
- Micreos GmbH, Wädenswil, Switzerland
| | - Bjorn L. Herpers
- Regional Public Health Laboratory Kennemerland, Haarlem, The Netherlands
| | - Paul Badoux
- Regional Public Health Laboratory Kennemerland, Haarlem, The Netherlands
| | | | - Raif S. Geha
- Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Ferd Janssen
- Micreos Human Health B.V., Bilthoven, The Netherlands
| | - Bob de Rooij
- Micreos Human Health B.V., Bilthoven, The Netherlands
| | | | | | | | | | - Martin J. Loessner
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Mathias Schmelcher
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
- Micreos GmbH, Wädenswil, Switzerland
| |
Collapse
|
10
|
Valente LG, Le ND, Pitton M, Chiffi G, Grandgirard D, Jakob SM, Cameron DR, Resch G, Que YA, Leib SL. Efficacy assessment of a novel endolysin PlyAZ3aT for the treatment of ceftriaxone-resistant pneumococcal meningitis in an infant rat model. PLoS One 2022; 17:e0266928. [PMID: 35472061 PMCID: PMC9041855 DOI: 10.1371/journal.pone.0266928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/29/2022] [Indexed: 11/21/2022] Open
Abstract
Background Treatment failure in pneumococcal meningitis due to antibiotic resistance is an increasing clinical challenge and alternatives to antibiotics warrant investigation. Phage-derived endolysins efficiently kill gram-positive bacteria including multi-drug resistant strains, making them attractive therapeutic candidates. The current study assessed the therapeutic potential of the novel endolysin PlyAZ3aT in an infant rat model of ceftriaxone-resistant pneumococcal meningitis. Methods Efficacy of PlyAZ3aT was assessed in a randomized, blinded and controlled experimental study in infant Wistar rats. Meningitis was induced by intracisternal infection with 5 x 107 CFU/ml of a ceftriaxone-resistant clinical strain of S. pneumoniae, serotype 19A. Seventeen hours post infection (hpi), animals were randomized into 3 treatment groups and received either (i) placebo (phosphate buffered saline [PBS], n = 8), (ii) 50 mg/kg vancomycin (n = 10) or (iii) 400 mg/kg PlyAZ3aT (n = 8) via intraperitoneal injection. Treatments were repeated after 12 h. Survival at 42 hpi was the primary outcome; bacterial loads in cerebrospinal fluid (CSF) and blood were secondary outcomes. Additionally, pharmacokinetics of PlyAZ3aT in serum and CSF was assessed. Results PlyAZ3aT did not improve survival compared to PBS, while survival for vancomycin treated animals was 70% which is a significant improvement when compared to PBS or PlyAZ3aT (p<0.05 each). PlyAZ3aT was not able to control the infection, reflected by the inability to reduce bacterial loads in the CSF, whereas Vancomycin sterilized the CSF and within 25 h. Pharmacokinetic studies indicated that PlyAZ3aT did not cross the blood brain barrier (BBB). In support, PlyAZ3aT showed a peak concentration of 785 μg/ml in serum 2 h after intraperitoneal injection but could not be detected in CSF. Conclusion In experimental pneumococcal meningitis, PlyAZ3aT failed to cure the infection due to an inability to reach the CSF. Optimization of the galenic formulation e.g. using liposomes might enable crossing of the BBB and improve treatment efficacy.
Collapse
Affiliation(s)
- Luca G. Valente
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Ngoc Dung Le
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Melissa Pitton
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Gabriele Chiffi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Denis Grandgirard
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Stephan M. Jakob
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - David R. Cameron
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Grégory Resch
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital, Lausanne, Switzerland
| | - Yok-Ai Que
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Stephen L. Leib
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
11
|
Miyano T, Irvine AD, Tanaka RJ. Model-based meta-analysis to optimise S. aureus-targeted therapies for atopic dermatitis. JID INNOVATIONS 2022; 2:100110. [PMID: 35757782 PMCID: PMC9214323 DOI: 10.1016/j.xjidi.2022.100110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/29/2022] Open
Abstract
Several clinical trials of Staphylococcus aureus (S. aureus)‒targeted therapies for atopic dermatitis (AD) have shown conflicting results about whether they improve AD severity scores. This study performs a model-based meta-analysis to investigate the possible causes of these conflicting results and suggests how to improve the efficacies of S. aureus‒targeted therapies. We developed a mathematical model that describes systems-level AD pathogenesis involving dynamic interactions between S. aureus and coagulase-negative Staphylococcus (CoNS). Our model simulation reproduced the clinically observed detrimental effects of the application of S. hominis A9 and flucloxacillin on AD severity and showed that these effects disappeared if the bactericidal activity against CoNS was removed. A hypothetical (modeled) eradication of S. aureus by 3.0 log10 colony-forming unit per cm2 without killing CoNS achieved Eczema Area and Severity Index 75 comparable with that of dupilumab. This efficacy was potentiated if dupilumab was administered in conjunction with S. aureus eradication (Eczema Area and Severity Index 75 at week 16) (S. aureus eradication: 66.7%, dupilumab 61.6% and combination 87.8%). The improved efficacy was also seen for virtual dupilumab poor responders. Our model simulation suggests that killing CoNS worsens AD severity and that S. aureus‒specific eradication without killing CoNS could be effective for patients with AD, including dupilumab poor responders. This study will contribute to designing promising S. aureus‒targeted therapy.
Collapse
Affiliation(s)
- Takuya Miyano
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Alan D. Irvine
- Pediatric Dermatology, Children’s Health Ireland at Crumlin, Dublin, Ireland
- Clinical Medicine, College of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Reiko J. Tanaka
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Correspondence: Reiko J. Tanaka, Department of Bioengineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom.
| |
Collapse
|
12
|
Özal D, Arndt A, Thomé M. Bacteriophages and related endolysins for reduction of microorganisms in the human body - a systematic review. GMS HYGIENE AND INFECTION CONTROL 2022; 17:Doc01. [PMID: 35111563 PMCID: PMC8780682 DOI: 10.3205/dgkh000404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background: In recent years, resistance to antibiotics has become a global threat, and alternatives to antibiotics have become an area of research. The main alternative methods are briefly described in this review. However, the main focus is bacteriophage-related therapy. Bacteriophages are viruses which, due to the production of the enzyme endolysin, are able to kill bacterial host cells. Bacteriophage therapies have a long tradition. Their potential to function as antibiotics lies in their bactericidal activity and specificity in killing bacteria without infecting or affecting eukaryotic cells. Objective: To systematically review the outcomes of bacteriophage therapy in patients with bacterial infections. Methods: The MEDLINE, EMBASE, Web of Science and CENTRAL databases were searched electronically using search terms referring to bacteriophages, endolysins and antimicrobial resistance. After the literature was screened for their titles and abstracts, full-text reviews considering inclusion/exclusion criteria were performed. Data concerning patients with bacterial infections, treatment with either bacteriophages or its enzyme endolysin and their outcomes were extracted and analysed. Results: Thirteen publications were identified that met all inclusion criteria. Data extraction shows that bacteriophages or endolysins have the potential to combat bacterial infections and significantly reduce inflammatory mediators. However, 3 out of 4 randomized controlled trials revealed that there was no significant difference between phage/endolysin treated patients and control group. Significant clinical improvements were seen in cohort and case studies. A few minor side effects were reported. Conclusions: Although there are countries in which bacteriophages are prescribed as an alternative to established antibiotics, this valuable experience has yet to be examined sufficiently in clinical trials conducted to modern standards. Despite improvements in symptoms shown in the reviewed clinical trials, the infection and the bacteria themselves were rarely completely eradicated. Therefore, no definite answer can be given as to effectiveness, and further clinical trials are necessary.
Collapse
Affiliation(s)
- Dilara Özal
- Kassel School of Medicine, University of Southampton, Southampton, UK,*To whom correspondence should be addressed: Dilara Özal, Kassel School of Medicine, University of Southampton, Southampton, UK, E-mail:
| | | | - Marcus Thomé
- Kassel School of Medicine, University of Southampton, Southampton, UK,Department of Microbiology, Klinikum Kassel, Kassel, Germany
| |
Collapse
|
13
|
Abstract
The skin microbiome is a key component of pathogenesis in atopic dermatitis (AD). The skin of AD patients is characterized by microbial dysbiosis, with a reduction of microbial diversity and overrepresentation of pathogenic Staphylococcus aureus (S. aureus). Recent exciting studies have elucidated an importance of establishing an appropriate immune response to microbes in early life and uncovered the new mechanisms of microbial community dynamics in modulating our skin microbiome. Several microbes are associated with AD pathogenesis, with proposed pathogenic effects from S. aureus and Malassezia. The complex relationships between microbes within the skin microbiome consortia includes various species, such as Staphylococcal, Roseomonas and Cutibacterium strains, that can inhibit S. aureus and are potential probiotics for AD skin. Numerous microbes are now also reported to modulate host response via communication with keratinocytes, specialized immune cells and adipocytes to improve skin health and barrier function. This increased understanding of skin microbiota bioactives has led to new biotherapeutic approaches that target the skin surface microenvironment for AD treatment.
Collapse
|
14
|
Danis-Wlodarczyk KM, Wozniak DJ, Abedon ST. Treating Bacterial Infections with Bacteriophage-Based Enzybiotics: In Vitro, In Vivo and Clinical Application. Antibiotics (Basel) 2021; 10:1497. [PMID: 34943709 PMCID: PMC8698926 DOI: 10.3390/antibiotics10121497] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Over the past few decades, we have witnessed a surge around the world in the emergence of antibiotic-resistant bacteria. This global health threat arose mainly due to the overuse and misuse of antibiotics as well as a relative lack of new drug classes in development pipelines. Innovative antibacterial therapeutics and strategies are, therefore, in grave need. For the last twenty years, antimicrobial enzymes encoded by bacteriophages, viruses that can lyse and kill bacteria, have gained tremendous interest. There are two classes of these phage-derived enzymes, referred to also as enzybiotics: peptidoglycan hydrolases (lysins), which degrade the bacterial peptidoglycan layer, and polysaccharide depolymerases, which target extracellular or surface polysaccharides, i.e., bacterial capsules, slime layers, biofilm matrix, or lipopolysaccharides. Their features include distinctive modes of action, high efficiency, pathogen specificity, diversity in structure and activity, low possibility of bacterial resistance development, and no observed cross-resistance with currently used antibiotics. Additionally, and unlike antibiotics, enzybiotics can target metabolically inactive persister cells. These phage-derived enzymes have been tested in various animal models to combat both Gram-positive and Gram-negative bacteria, and in recent years peptidoglycan hydrolases have entered clinical trials. Here, we review the testing and clinical use of these enzymes.
Collapse
Affiliation(s)
| | - Daniel J. Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA;
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA;
| | - Stephen T. Abedon
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA;
| |
Collapse
|
15
|
Abedon ST, Danis-Wlodarczyk KM, Alves DR. Phage Therapy in the 21st Century: Is There Modern, Clinical Evidence of Phage-Mediated Efficacy? Pharmaceuticals (Basel) 2021; 14:1157. [PMID: 34832939 PMCID: PMC8625828 DOI: 10.3390/ph14111157] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022] Open
Abstract
Many bacteriophages are obligate killers of bacteria. That this property could be medically useful was first recognized over one hundred years ago, with 2021 being the 100-year anniversary of the first clinical phage therapy publication. Here we consider modern use of phages in clinical settings. Our aim is to answer one question: do phages serve as effective anti-bacterial infection agents when used clinically? An important emphasis of our analyses is on whether phage therapy-associated anti-bacterial infection efficacy can be reasonably distinguished from that associated with often coadministered antibiotics. We find that about half of 70 human phage treatment reports-published in English thus far in the 2000s-are suggestive of phage-mediated anti-bacterial infection efficacy. Two of these are randomized, double-blinded, infection-treatment studies while 14 of those studies, in our opinion, provide superior evidence of a phage role in observed treatment successes. Roughly three-quarters of these potentially phage-mediated outcomes are based on microbiological as well as clinical results, with the rest based on clinical success. Since many of these phage treatments are of infections for which antibiotic therapy had not been successful, their collective effectiveness is suggestive of a valid utility in employing phages to treat otherwise difficult-to-cure bacterial infections.
Collapse
Affiliation(s)
- Stephen T. Abedon
- Department of Microbiology, The Ohio State University, Mansfield, OH 44906, USA;
| | | | - Diana R. Alves
- Department of Microbiology, The Ohio State University, Mansfield, OH 44906, USA;
| |
Collapse
|
16
|
Castillo CR, Alishahedani ME, Gough P, Chaudhary PP, Yadav M, Matriz J, Myles IA. Assessing the effects of common topical exposures on skin bacteria associated with atopic dermatitis. SKIN HEALTH AND DISEASE 2021; 1. [PMID: 34723253 PMCID: PMC8555759 DOI: 10.1002/ski2.41] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Background While patients and families struggling with atopic dermatitis (AD) have documented concerns for a contributory role of skin care products in AD pathology, nearly all the skin microbiome studies to date have asked participants to avoid topical products (such as soaps or select medications) for the preceding days to weeks prior to sample collection. Thus, given the established role of the microbiome in AD, the interactions between topical exposures, dysbiosis and AD remains underrepresented in the academic literature. Objectives To address this knowledge gap, we expanded our previous evaluations to test the toxicological effects of a broader range of common chemicals, AD treatment lotions, creams and ointments using both health- and AD-associated strains of Roseomonas mucosa and Staphylococcus spp. Methods Use of in vitro culture techniques and mouse models were deployed to identify chemicals with dysbiotic or pre-biotic potential. A proof-of-concept study was subsequently performed in healthy volunteers to assess global microbiome shifts after exposure to select chemicals using dermatologic patch testing. Results Numerous chemicals possessed antibiotic properties, including many not marketed as anti-microbials. Through targeted combination of potentially beneficial chemicals, we identified combinations which promoted the growth of health-associated isolates over disease-associated strains in bacterial culture and enhanced microbe-specific outcomes in an established mouse model of AD; the most promising of which was the combination of citral and colophonium (often sold as lemon myrtle oil and pine tar). Additional studies would likely further optimize the combination of ingredients use. Similar results were seen in the proof-of-concept human studies. Conclusions Our results could offer a systematic, multiplex approach to identify which products carry dysbiotic potential and thus may guide formulation of new topicals to benefit patients with AD.
Collapse
Affiliation(s)
- C R Castillo
- Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - M E Alishahedani
- Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - P Gough
- Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - P P Chaudhary
- Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - M Yadav
- Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - J Matriz
- Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - I A Myles
- Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
17
|
Nomura T, Kabashima K. Advances in Atopic Dermatitis in 2019-2020: Endotypes from skin barrier, ethnicity, properties of antigen, cytokine profiles, microbiome, and engagement of immune cells. J Allergy Clin Immunol 2021; 148:1451-1462. [PMID: 34756922 DOI: 10.1016/j.jaci.2021.10.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022]
Abstract
Key research advances in atopic dermatitis (AD) suggest the complexity of its endotypes. A comprehensive serum biomarker panel revealed at least four types of AD. Some represent classic TH2-dominant AD with filaggrin mutations commonly reported in Europeans, a simultaneously activated multipolar axes of cytokines often reported in Asians, and an intrinsic type characterized by TH2-inferiority. Innate lymphoid cells, including NK cells, NKT cells, and fibroblasts, play a role in AD development and heterogeneity. Here, we discuss the endotypes of AD from the perspective of antigen types (hapten vs. protein antigens), barrier function, and a novel set of immune cells. Endotypic stratification of AD may lead to the development of customized therapeutic strategies in the future.
Collapse
Affiliation(s)
- Takashi Nomura
- Department of Dermatology, Faculty of Medicine, Kyoto University 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kenji Kabashima
- Department of Dermatology, Faculty of Medicine, Kyoto University 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
18
|
Gutiérrez D, Rodríguez-Rubio L, Ruas-Madiedo P, Fernández L, Campelo AB, Briers Y, Nielsen MW, Pedersen K, Lavigne R, García P, Rodríguez A. Design and Selection of Engineered Lytic Proteins With Staphylococcus aureus Decolonizing Activity. Front Microbiol 2021; 12:723834. [PMID: 34594314 PMCID: PMC8477017 DOI: 10.3389/fmicb.2021.723834] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022] Open
Abstract
Staphylococcus aureus causes various infections in humans and animals, the skin being the principal reservoir of this pathogen. The widespread occurrence of methicillin-resistant S. aureus (MRSA) limits the elimination and treatment of this pathogen. Phage lytic proteins have been proven as efficient antimicrobials against S. aureus. Here, a set of 12 engineered proteins based on endolysins were conceptualized to select the most optimal following a stepwise funnel approach assessing parameters including turbidity reduction, minimum inhibitory concentration (MIC), time-kill curves, and antibiofilm assays, as well as testing their stability in a broad range of storage conditions (pH, temperature, and ionic strength). The engineered phage lysins LysRODIΔAmi and ClyRODI-H5 showed the highest specific lytic activity (5 to 50 times higher than the rest), exhibited a shelf-life up to 6 months and remained stable at temperatures up to 50°C and in a pH range from 3 to 9. LysRODIΔAmi showed the lower MIC values against all staphylococcal strains tested. Both proteins were able to kill 6 log units of the strain S. aureus Sa9 within 5 min and could remove preformed biofilms (76 and 65%, respectively). Moreover, LysRODIΔAmi could prevent biofilm formation at low protein concentrations (0.15–0.6 μM). Due to its enhanced antibiofilm properties, LysRODIΔAmi was selected to effectively remove S. aureus contamination in both intact and disrupted keratinocyte monolayers. Notably, this protein did not demonstrate any toxicity toward human keratinocytes, even at high concentrations (22.1 μM). Finally, a pig skin ex vivo model was used to evaluate treatment of artificially contaminated pig skin using LysRODIΔAmi (16.5 μg/cm2). Following an early reduction of S. aureus, a second dose of protein completely eradicated S. aureus. Overall, our results suggest that LysRODIΔAmi is a suitable candidate as antimicrobial agent to prevent and treat staphylococcal skin infections.
Collapse
Affiliation(s)
- Diana Gutiérrez
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Asturias, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Lorena Rodríguez-Rubio
- Laboratory of Gene Technology, Department of Biosystems, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Patricia Ruas-Madiedo
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Asturias, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Lucía Fernández
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Asturias, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Ana Belén Campelo
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Asturias, Spain
| | - Yves Briers
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Martin Weiss Nielsen
- Department of Microbiology and Production, National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Karl Pedersen
- Department of Microbiology and Production, National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Rob Lavigne
- Laboratory of Gene Technology, Department of Biosystems, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Pilar García
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Asturias, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Ana Rodríguez
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Asturias, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
19
|
Ho MKY, Zhang P, Chen X, Xia J, Leung SSY. Bacteriophage endolysins against gram-positive bacteria, an overview on the clinical development and recent advances on the delivery and formulation strategies. Crit Rev Microbiol 2021; 48:303-326. [PMID: 34478359 DOI: 10.1080/1040841x.2021.1962803] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Facing the increasing threat of multi-drug antimicrobial resistance (AMR), humans strive to search for antibiotic drug candidates and antibacterial alternatives from all possible places, from soils in remote areas to deep in the sea. In this "gold rush for antibacterials," researchers turn to the natural enemy of bacterial cells, bacteriophage (phages), and find them a rich source of weapons for AMR bacteria. Endolysins (lysins), the enzymes phages use to break the bacterial cells from within, have been shown to be highly selective and efficient in killing their target bacteria from outside while maintaining a low occurrence of bacterial resistance. In this review, we start with the structures and mechanisms of action of lysins against Gram-positive (GM+) bacteria. The developmental history of lysins is also outlined. Then, we detail the latest preclinical and clinical research on their safety and efficacy against GM+ bacteria, focusing on the formulation strategies of these enzymes. Finally, the challenges and potential hurdles are discussed. Notwithstanding these limitations, the trends in development indicate that the first, approved lysin drugs will be available soon in the near future. Overall, this review presents a timely summary of the current progress on lysins as antibacterial enzymes for AMR GM+ bacteria, and provides a guidebook for biomaterial researchers who are dedicating themselves to the battle against bacterial infections.
Collapse
Affiliation(s)
- Marco Kai Yuen Ho
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, China
| | - Pengfei Zhang
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, China
| | - Xi Chen
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
20
|
Nainu F, Permana AD, Djide NJN, Anjani QK, Utami RN, Rumata NR, Zhang J, Emran TB, Simal-Gandara J. Pharmaceutical Approaches on Antimicrobial Resistance: Prospects and Challenges. Antibiotics (Basel) 2021; 10:981. [PMID: 34439031 PMCID: PMC8388863 DOI: 10.3390/antibiotics10080981] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023] Open
Abstract
The rapid increase in pathogenic microorganisms with antimicrobial resistant profiles has become a significant public health problem globally. The management of this issue using conventional antimicrobial preparations frequently results in an increase in pathogen resistance and a shortage of effective antimicrobials for future use against the same pathogens. In this review, we discuss the emergence of AMR and argue for the importance of addressing this issue by discovering novel synthetic or naturally occurring antibacterial compounds and providing insights into the application of various drug delivery approaches, delivered through numerous routes, in comparison with conventional delivery systems. In addition, we discuss the effectiveness of these delivery systems in different types of infectious diseases associated with antimicrobial resistance. Finally, future considerations in the development of highly effective antimicrobial delivery systems to combat antimicrobial resistance are presented.
Collapse
Affiliation(s)
- Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Sulawesi Selatan, Indonesia; (A.D.P.); (N.J.N.D.); (Q.K.A.); (R.N.U.); (N.R.R.)
| | - Andi Dian Permana
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Sulawesi Selatan, Indonesia; (A.D.P.); (N.J.N.D.); (Q.K.A.); (R.N.U.); (N.R.R.)
| | - Nana Juniarti Natsir Djide
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Sulawesi Selatan, Indonesia; (A.D.P.); (N.J.N.D.); (Q.K.A.); (R.N.U.); (N.R.R.)
| | - Qonita Kurnia Anjani
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Sulawesi Selatan, Indonesia; (A.D.P.); (N.J.N.D.); (Q.K.A.); (R.N.U.); (N.R.R.)
- Medical Biology Centre, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK
| | - Rifka Nurul Utami
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Sulawesi Selatan, Indonesia; (A.D.P.); (N.J.N.D.); (Q.K.A.); (R.N.U.); (N.R.R.)
- Institute of Pharmaceutical Science, King’s College of London, London SE1 9NH, UK
| | - Nur Rahma Rumata
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Sulawesi Selatan, Indonesia; (A.D.P.); (N.J.N.D.); (Q.K.A.); (R.N.U.); (N.R.R.)
- Sekolah Tinggi Ilmu Farmasi Makassar, Makassar 90242, Sulawesi Selatan, Indonesia
| | - Jianye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China;
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo–Ourense Campus, E32004 Ourense, Spain
| |
Collapse
|
21
|
Hwang J, Thompson A, Jaros J, Blackcloud P, Hsiao J, Shi VY. Updated understanding of Staphylococcus aureus in atopic dermatitis: From virulence factors to commensals and clonal complexes. Exp Dermatol 2021; 30:1532-1545. [PMID: 34293242 DOI: 10.1111/exd.14435] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/18/2021] [Accepted: 07/20/2021] [Indexed: 12/22/2022]
Abstract
Atopic dermatitis (AD) is a common inflammatory dermatosis that has multiple contributing factors including genetic, immunologic and environmental. Staphylococcus aureus (SA) has long been associated with exacerbation of AD. SA produces many virulence factors that interact with the human skin and immune system. These superantigens and toxins have been shown to contribute to adhesion, inflammation and skin barrier destruction. Recent advances in genome sequencing techniques have led to a broadened understanding of the multiple ways SA interacts with the cutaneous environment in AD hosts. For example, temporal shifts in the microbiome, specifically in clonal complexes of SA, have been identified during AD flares and remission. Herein, we review mechanisms of interaction between the cutaneous microbiome and SA and highlight known differences in SA clonal complexes that contribute to AD pathogenesis. Detailed knowledge of the genetic strains of SA and cutaneous dysbiosis is becoming increasingly relevant in paving the way for microbiome-modulating and precision therapies for AD.
Collapse
Affiliation(s)
- Jonwei Hwang
- University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Alyssa Thompson
- College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Joanna Jaros
- John H. Stroger Hospital Cook County Health Dermatology, Chicago, Illinois, USA
| | - Paul Blackcloud
- Division of Dermatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Jennifer Hsiao
- Division of Dermatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Vivian Y Shi
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
22
|
Walsh L, Johnson CN, Hill C, Ross RP. Efficacy of Phage- and Bacteriocin-Based Therapies in Combatting Nosocomial MRSA Infections. Front Mol Biosci 2021; 8:654038. [PMID: 33996906 PMCID: PMC8116899 DOI: 10.3389/fmolb.2021.654038] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is a pathogen commonly found in nosocomial environments where infections can easily spread - especially given the reduced immune response of patients and large overlap between personnel in charge of their care. Although antibiotics are available to treat nosocomial infections, the increased occurrence of antibiotic resistance has rendered many treatments ineffective. Such is the case for methicillin resistant S. aureus (MRSA), which has continued to be a threat to public health since its emergence. For this reason, alternative treatment technologies utilizing antimicrobials such as bacteriocins, bacteriophages (phages) and phage endolysins are being developed. These antimicrobials provide an advantage over antibiotics in that many have narrow inhibition spectra, enabling treatments to be selected based on the target (pathogenic) bacterium while allowing for survival of commensal bacteria and thus avoiding collateral damage to the microbiome. Bacterial resistance to these treatments occurs less frequently than with antibiotics, particularly in circumstances where combinatory antimicrobial therapies are used. Phage therapy has been well established in Eastern Europe as an effective treatment against bacterial infections. While there are no Randomized Clinical Trials (RCTs) to our knowledge examining phage treatment of S. aureus infections that have completed all trial phases, numerous clinical trials are underway, and several commercial phage preparations are currently available to treat S. aureus infections. Bacteriocins have primarily been used in the food industry for bio-preservation applications. However, the idea of repurposing bacteriocins for human health is an attractive one considering their efficacy against many bacterial pathogens. There are concerns about the ability of bacteriocins to survive the gastrointestinal tract given their proteinaceous nature, however, this obstacle may be overcome by altering the administration route of the therapy through encapsulation, or by bioengineering protease-resistant variants. Obstacles such as enzymatic digestion are less of an issue for topical/local administration, for example, application to the surface of the skin. Bacteriocins have also shown impressive synergistic effects when used in conjunction with other antimicrobials, including antibiotics, which may allow antibiotic-based therapies to be used more sparingly with less resistance development. This review provides an updated account of known bacteriocins, phages and phage endolysins which have demonstrated an impressive ability to kill S. aureus strains. In particular, examples of antimicrobials with the ability to target MRSA strains and their subsequent use in a clinical setting are outlined.
Collapse
Affiliation(s)
- Lauren Walsh
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Crystal N Johnson
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Cork, Ireland
| | - Colin Hill
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Cork, Ireland
| |
Collapse
|
23
|
van Mierlo MMF, Pasmans SGMA, Totté JEE, de Wit J, Herpers BL, Vos MC, Klaassen CHW, Pardo LM. Temporal Variation in Staphylococcus aureus Protein A Genotypes from Nose and Skin in Atopic Dermatitis Patients. Dermatology 2021; 237:506-512. [PMID: 33823508 DOI: 10.1159/000515235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/06/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Staphylococcus aureus colonization is associated with disease severity in patients with atopic dermatitis (AD). OBJECTIVE To investigate temporal variation in S. aureus protein A gene (spa)-types isolated from the nose and lesional skin and the correlation of spa-types with disease severity. RESULTS This study included 96 adult AD patients who were assessed at baseline (T0) and after a strict 2-week follow-up period (T1) in which treatment was standardized with a topical corticosteroid. Fifty-five different spa-types were detected in the nose and skin cultures. Seventy-three patients were colonized with S. aureus in the nasal cavity at both time points (persistent carriership), 59 of whom (81%) had identical spa-types over time. For skin samples, 42 (75%) of the 56 persistent skin carriers had identical spa-types over time. The same spa-type was carried in the nose and skin in 79 and 77% of the patients at T0 and T1, respectively. More severe disease was not associated with specific spa-types or with temporal variation in spa-type. CONCLUSION S. aureus strains in AD are highly heterogeneous between patients. The majority of patients carry the same spa-type in the nose and skin without temporal variation, suggesting clonal colonization within individual patients. No predominant spa-type or temporal variation is associated with increased disease severity.
Collapse
Affiliation(s)
- Minke M F van Mierlo
- Department of Dermatology, Erasmus MC University Medical Center Rotterdam-Sophia Children's Hospital-Center of Pediatric Dermatology, Rotterdam, The Netherlands
| | - Suzanne G M A Pasmans
- Department of Dermatology, Erasmus MC University Medical Center Rotterdam-Sophia Children's Hospital-Center of Pediatric Dermatology, Rotterdam, The Netherlands
| | - Joan E E Totté
- Department of Dermatology, Erasmus MC University Medical Center Rotterdam-Sophia Children's Hospital-Center of Pediatric Dermatology, Rotterdam, The Netherlands.,Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jill de Wit
- Department of Dermatology, Erasmus MC University Medical Center Rotterdam-Sophia Children's Hospital-Center of Pediatric Dermatology, Rotterdam, The Netherlands
| | - Bjorn L Herpers
- Regional Public Health Laboratory Kennemerland, Haarlem, The Netherlands
| | - Margreet C Vos
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Corné H W Klaassen
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Luba M Pardo
- Department of Dermatology, Erasmus MC University Medical Center Rotterdam-Sophia Children's Hospital-Center of Pediatric Dermatology, Rotterdam, The Netherlands
| |
Collapse
|
24
|
Nie T, Meng F, Zhou L, Lu F, Bie X, Lu Z, Lu Y. In Silico Development of Novel Chimeric Lysins with Highly Specific Inhibition against Salmonella by Computer-Aided Design. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:3751-3760. [PMID: 33565867 DOI: 10.1021/acs.jafc.0c07450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Four novel chimeric lysins (P361, P362, P371, and P372), which were the fusion of Salmonella phage lysins and novel antimicrobial peptide LeuA-P, were obtained using bioinformatics analysis and in silico design. The recombinant chimeric lysins were expressed in E. coli BL21(DE3) strain and showed highly specific inhibition against Salmonella. The minimal inhibitory concentrations (MICs) of P362 and P372 to S. typhi CMCC 50071 were 8 and 16 μg/mL, respectively. Both 1 × MIC P362 and P372 could increase the outer membrane permeability and cleave the cell wall peptidoglycan, causing the leakage of intracellular nucleic acids and proteins and ultimately killing Salmonella efficiently without drug resistance. The combination of P362, P372, and potassium sorbate reduced more than 3 log CFU/g counts of microorganisms in contaminated chilled chicken and extended the shelf life by 7 days. The strategy of antimicrobial peptide (AMP)-lysin chimera inspired the inability of phage lysin to specifically inhibit Gram-negative bacteria with dense outer membranes in vitro.
Collapse
Affiliation(s)
- Ting Nie
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Fanqiang Meng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Libang Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Fengxia Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Xiaomei Bie
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Zhaoxin Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, China
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, Jiangsu Province 210023, China
| |
Collapse
|
25
|
Pinto AM, Silva MD, Pastrana LM, Bañobre-López M, Sillankorva S. The clinical path to deliver encapsulated phages and lysins. FEMS Microbiol Rev 2021; 45:6204673. [PMID: 33784387 DOI: 10.1093/femsre/fuab019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
The global emergence of multidrug-resistant pathogens is shaping the current dogma regarding the use of antibiotherapy. Many bacteria have evolved to become resistant to conventional antibiotherapy, representing a health and economic burden for those afflicted. The search for alternative and complementary therapeutic approaches has intensified and revived phage therapy. In recent decades, the exogenous use of lysins, encoded in phage genomes, has shown encouraging effectiveness. These two antimicrobial agents reduce bacterial populations; however, many barriers challenge their prompt delivery at the infection site. Encapsulation in delivery vehicles provides targeted therapy with a controlled compound delivery, surpassing chemical, physical and immunological barriers that can inactivate and eliminate them. This review explores phages and lysins' current use to resolve bacterial infections in the respiratory, digestive, and integumentary systems. We also highlight the different challenges they face in each of the three systems and discuss the advances towards a more expansive use of delivery vehicles.
Collapse
Affiliation(s)
- Ana Mafalda Pinto
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, 4710-057 Braga, Portugal.,INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Maria Daniela Silva
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, 4710-057 Braga, Portugal.,INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Lorenzo M Pastrana
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Manuel Bañobre-López
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Sanna Sillankorva
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| |
Collapse
|
26
|
Schmelcher M, Loessner MJ. Bacteriophage endolysins - extending their application to tissues and the bloodstream. Curr Opin Biotechnol 2020; 68:51-59. [PMID: 33126104 DOI: 10.1016/j.copbio.2020.09.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/25/2022]
Abstract
The rapid emergence of antibiotic-resistant bacteria and the lack of novel antibacterial agents pose a serious threat for patients and healthcare systems. Bacteriophage-encoded peptidoglycan hydrolases (endolysins) represent a promising new class of antimicrobials. Over the past two decades, research on these enzymes has evolved from basic in vitro characterization to sophisticated protein engineering approaches, including advanced preclinical and clinical testing. In recent years, increasingly specific animal models have shown efficacy of endolysins against bacterial infections of various different organs and tissues of the body. Despite these advances, some challenges with regard to systemic application of endolysins remain to be addressed. These include immunogenicity, circulation half-life, and cell and tissue-specific targeting and penetration properties.
Collapse
Affiliation(s)
- Mathias Schmelcher
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland.
| | - Martin J Loessner
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Gutiérrez D, Briers Y. Lysins breaking down the walls of Gram-negative bacteria, no longer a no-go. Curr Opin Biotechnol 2020; 68:15-22. [PMID: 33053478 DOI: 10.1016/j.copbio.2020.08.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 01/24/2023]
Abstract
Phage lysins are one of the most advanced classes of antibacterials under clinical evaluation and have a new mode of action based on peptidoglycan degradation. Lysins were initially excluded from use against Gram-negative pathogens because of their impermeable outer membrane, but are now increasingly developed as effective antibacterials against these critical priority pathogens. Generally, three routes of investigation have been recently explored and advanced to different extents, including the use of lysins that possess intrinsic activity due to a positively charged C-terminus that destabilizes the outer membrane, the use of physical or chemical means to disrupt the outer membrane integrity and protein engineering to equip the lysin with the necessary tools to overcome the outer membrane.
Collapse
Affiliation(s)
- Diana Gutiérrez
- Department of Biotechnology, Ghent University, Valentin Vaerwyckweg 1, 9000 Gent, Belgium
| | - Yves Briers
- Department of Biotechnology, Ghent University, Valentin Vaerwyckweg 1, 9000 Gent, Belgium.
| |
Collapse
|
28
|
Topical anti-microbial peptide omiganan recovers cutaneous dysbiosis but does not improve clinical symptoms in patients with mild-to-moderate atopic dermatitis in a phase 2 randomized controlled trial. J Am Acad Dermatol 2020; 86:854-862. [PMID: 33010325 DOI: 10.1016/j.jaad.2020.08.132] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Dysbiosis and colonization with Staphylococcus aureus is considered to play an important role in the pathogenesis of atopic dermatitis (AD). Recovering this dysbiosis may improve AD symptoms. Omiganan is a synthetic indolicidin analogue antimicrobial peptide with activity against S. aureus and could be a viable new treatment option for AD. OBJECTIVE To explore the tolerability, clinical efficacy and pharmacodynamics of omiganan in mild-to-moderate AD. METHODS Eighty patients were randomized to omiganan 1%, 1.75%, 2.5% or vehicle twice daily for 28 days on all lesions. Weekly visits included clinical scores, and microbiological and pharmacodynamic assessments of one 'target lesion'. RESULTS In all omiganan treatment groups dysbiosis was recovered by reducing Staphylococcus abundance and increasing diversity. A reduction of cultured S. aureus was observed in all omiganan treatment groups, with a significant reduction for omiganan 2.5% compared to vehicle (-93.5%, 95%CI=-99.2%/-28.5% p=0.02). No significant clinical improvement was observed. CONCLUSION Topical administration of omiganan twice daily for up to 28 days in patients with mild-to-moderate AD led to a recovery of dysbiosis, but without clinical improvement. Therefore, a mono-treatment that selectively targets the microbiome does not appear to be a successful treatment strategy in mild-to-moderate AD.
Collapse
|
29
|
Manohar P, Loh B, Athira S, Nachimuthu R, Hua X, Welburn SC, Leptihn S. Secondary Bacterial Infections During Pulmonary Viral Disease: Phage Therapeutics as Alternatives to Antibiotics? Front Microbiol 2020; 11:1434. [PMID: 32733404 PMCID: PMC7358648 DOI: 10.3389/fmicb.2020.01434] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/03/2020] [Indexed: 12/25/2022] Open
Abstract
Secondary bacterial infections manifest during or after a viral infection(s) and can lead to negative outcomes and sometimes fatal clinical complications. Research and development of clinical interventions is largely focused on the primary pathogen, with research on any secondary infection(s) being neglected. Here we highlight the impact of secondary bacterial infections and in particular those caused by antibiotic-resistant strains, on disease outcomes. We describe possible non-antibiotic treatment options, when small molecule drugs have no effect on the bacterial pathogen and explore the potential of phage therapy and phage-derived therapeutic proteins and strategies in treating secondary bacterial infections, including their application in combination with chemical antibiotics.
Collapse
Affiliation(s)
- Prasanth Manohar
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, China.,The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Belinda Loh
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, China
| | - Sudarsanan Athira
- Antibiotic Resistance and Phage Therapy Laboratory, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Ramesh Nachimuthu
- Antibiotic Resistance and Phage Therapy Laboratory, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Xiaoting Hua
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Susan C Welburn
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, China.,Infection Medicine, Biomedical Sciences, Edinburgh Medical School, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Sebastian Leptihn
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, China.,Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Infection Medicine, Biomedical Sciences, Edinburgh Medical School, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
30
|
Tham EH, Koh E, Common JEA, Hwang IY. Biotherapeutic Approaches in Atopic Dermatitis. Biotechnol J 2020; 15:e1900322. [PMID: 32176834 DOI: 10.1002/biot.201900322] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/24/2020] [Indexed: 12/15/2022]
Abstract
The skin microbiome plays a central role in inflammatory skin disorders such as atopic dermatitis (AD). In AD patients, an imbalance between pathogenic Staphylococcus aureus (S. aureus) and resident skin symbionts creates a state of dysbiosis which induces immune dysregulation and impairs skin barrier function. There are now exciting new prospects for microbiome-based interventions for AD prevention. In the hopes of achieving sustained control and management of disease in AD patients, current emerging biotherapeutic strategies aim to harness the skin microbiome associated with health by restoring a more diverse symbiotic skin microbiome, while selectively removing pathogenic S. aureus. Examples of such strategies are demonstrated in skin microbiome transplants, phage-derived anti-S. aureus endolysins, monoclonal antibodies, and quorum sensing (QS) inhibitors. However, further understanding of the skin microbiome and its role in AD pathogenesis is still needed to understand how these biotherapeutics alter the dynamics of the microbiome community; to optimize patient selection, drug delivery, and treatment duration; overcome rapid recolonization upon treatment cessation; and improve efficacy to allow these therapeutic options to eventually reach routine clinical practice.
Collapse
Affiliation(s)
- Elizabeth Huiwen Tham
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, 119074, Singapore
| | - Elvin Koh
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation, National University of Singapore, Singapore, 119228, Singapore
| | - John E A Common
- Skin Research Institute of Singapore, A*STAR, Singapore, 308232, Singapore
| | - In Young Hwang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation, National University of Singapore, Singapore, 119228, Singapore
| |
Collapse
|
31
|
Imanishi I, Uchiyama J, Tsukui T, Hisatsune J, Ide K, Matsuzaki S, Sugai M, Nishifuji K. Therapeutic Potential of an Endolysin Derived from Kayvirus S25-3 for Staphylococcal Impetigo. Viruses 2019; 11:v11090769. [PMID: 31443379 PMCID: PMC6784202 DOI: 10.3390/v11090769] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 12/31/2022] Open
Abstract
Impetigo is a contagious skin infection predominantly caused by Staphylococcus aureus. Decontamination of S. aureus from the skin is becoming more difficult because of the emergence of antibiotic-resistant strains. Bacteriophage endolysins are less likely to invoke resistance and can eliminate the target bacteria without disturbance of the normal microflora. In this study, we investigated the therapeutic potential of a recombinant endolysin derived from kayvirus S25-3 against staphylococcal impetigo in an experimental setting. First, the recombinant S25-3 endolysin required an incubation period of over 15 minutes to exhibit efficient bactericidal effects against S. aureus. Second, topical application of the recombinant S25-3 endolysin decreased the number of intraepidermal staphylococci and the size of pustules in an experimental mouse model of impetigo. Third, treatment with the recombinant S25-3 endolysin increased the diversity of the skin microbiota in the same mice. Finally, we revealed the genus-specific bacteriolytic effect of recombinant S25-3 endolysin against staphylococci, particularly S. aureus, among human skin commensal bacteria. Therefore, topical treatment with recombinant S25-3 endolysin can be a promising disease management procedure for staphylococcal impetigo by efficient bacteriolysis of S. aureus while improving the cutaneous bacterial microflora.
Collapse
Affiliation(s)
- Ichiro Imanishi
- Laboratory of Veterinary Internal Medicine, Division of Animal Life Science, Institute of Agriculture, Graduate School, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Jumpei Uchiyama
- Laboratory of Veterinary Microbiology I, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan
| | - Toshihiro Tsukui
- Nippon Zenyaku Kogyo Co. Ltd., 1-1 Tairanoue, Sasagawa, Asaka-machi, Koriyama, Fukushima 963-0196, Japan
| | - Junzo Hisatsune
- Department of Bacteriology, Graduate school of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Kaori Ide
- Laboratory of Veterinary Internal Medicine, Division of Animal Life Science, Institute of Agriculture, Graduate School, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Shigenobu Matsuzaki
- Department of Ophthalmology and Visual Science, Kochi Medical School, Kochi University, 185-1 Kohasu, Oko-cho, Nankoku, Kochi 783-8505, Japan
| | - Motoyuki Sugai
- Department of Bacteriology, Graduate school of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Koji Nishifuji
- Laboratory of Veterinary Internal Medicine, Division of Animal Life Science, Institute of Agriculture, Graduate School, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan.
| |
Collapse
|