1
|
Zou H, Huang X, Xiao W, He H, Liu S, Zeng H. Recent advancements in bacterial anti-phage strategies and the underlying mechanisms altering susceptibility to antibiotics. Microbiol Res 2025; 295:128107. [PMID: 40023108 DOI: 10.1016/j.micres.2025.128107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025]
Abstract
The rapid spread of multidrug-resistant bacteria and the challenges in developing new antibiotics have brought renewed international attention to phage therapy. However, in bacteria-phage co-evolution, the rapid development of bacterial resistance to phage has limited its clinical application. This review consolidates the latest advancements in research on anti-phage mechanisms, encompassing strategies such as systems associated with reduced nicotinamide adenine dinucleotide (NAD+) to halt the propagation of the phage, symbiotic bacteria episymbiont-mediated modulation of gene expression in host bacteria to resist phage infection, and defence-related reverse transcriptase (DRT) encoded by bacteria to curb phage infections. We conduct an in-depth analysis of the underlying mechanisms by which bacteria undergo alterations in antibiotic susceptibility after developing phage resistance. We also discuss the remaining challenges and promising directions for phage-based therapy in the future.
Collapse
Affiliation(s)
- Huanhuan Zou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaoyi Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Wenyue Xiao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Haoxuan He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Shenshen Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Haiyan Zeng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
2
|
Shamsuzzaman M, Kim S, Kim J. Therapeutic Potential of Novel Phages with Antibiotic Combinations Against ESBL-Producing and Carbapenem-Resistant Escherichia coli. J Glob Antimicrob Resist 2025:S2213-7165(25)00077-3. [PMID: 40268052 DOI: 10.1016/j.jgar.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 02/25/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVES The emergence of extended-spectrum β-lactamase (ESBL)-producing E. coli and carbapenem-resistant E. coli (CREC) is a significant global health challenge. This study focuses on isolating and characterizing two novel phages, EC.W1-9 and EC.W15-4, and investigating their efficacy with antibiotics against these resistant E. coli. METHODS In vivo experiments were conducted using BALB/c mice, and E.coli isolates were collected, cultured, and evaluated for antibiotic susceptibility. Phages were isolated from hospital sewage and prepared to infect the E. coli. RESULTS The isolated phages, EC.W1-9 and EC.W15-4, belonged to the Podoviridae and Straboviridae families, and lack integrase or toxin-coding genes, indicating safety for therapeutic use. The combination of these phages significently enhanced their lytic ability, lysing 61.7% of 60 E. coli isolates, compared to 41.6%-55% lysis by individual phages. Furthermore, the phage combination demonstrated 100% susceptibility against different E. coli sequence types, including ST73, ST648, ST2311, ST405, ST7962, ST131, ST13003, and ST167. Additionally, synergy between antibiotics and phage combinations improved susceptibility rates to 73.3% for ESBL producers and 54% for CREC. The combined treatment of isolated phages and antibiotics significantly increased survival rates in BALB/c mice exposed to resistant STs of E.coli, including ST131, ST648, and ST410. Survival rates against ST131 increased by approximately 75% and 50% compared to treatment individual phages. Combined treatment with two phages and antibiotics resulted in 75-100% survival against E. coli ST410 and 100% survival against ST648 CONCLUSIONS: This study highlights the therapeutic importance of phage and phage-antibiotic combinations in combating ESBL-producing E. coli and CREC isolates.
Collapse
Affiliation(s)
- Md Shamsuzzaman
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Republic of Korea; Department of Microbiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Shukho Kim
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Republic of Korea; Department of Microbiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jungmin Kim
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Republic of Korea; Department of Microbiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
3
|
Fong SA, Bouras G, Houtak G, Nepal R, Feizi S, Morales S, Psaltis AJ, Wormald PJ, Vreugde S. Genomic variation in Pseudomonas aeruginosa clinical respiratory isolates with de novo resistance to a bacteriophage cocktail. Microbiol Spectr 2025:e0214924. [PMID: 40162801 DOI: 10.1128/spectrum.02149-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/20/2025] [Indexed: 04/02/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that can cause sinus infections and pneumonia in cystic fibrosis (CF) patients. Bacteriophage therapy is being investigated as a treatment for antibiotic-resistant P. aeruginosa infections. Although virulent bacteriophages have shown promise in treating P. aeruginosa infections, the development of bacteriophage-insensitive mutants (BIMs) in the presence of bacteriophages has been described. The aim of this study was to examine the genetic changes associated with the BIM phenotype. Biofilms of three genetically distinct P. aeruginosa strains, including PAO1 (ATCC 15692), and two clinical respiratory isolates (one CF and one non-CF) were grown for 7 days and treated with either a cocktail of four bacteriophages or a vehicle control for 7 consecutive days. BIMs isolated from the biofilms were detected by streak assays, and resistance to the phage cocktail was confirmed using spot test assays. Comparison of whole genome sequencing between the recovered BIMs and their respective vehicle control-treated phage-sensitive isolates revealed structural variants in two strains, and several small variants in all three strains. These variations involved a TonB-dependent outer membrane receptor in one strain, and mutations in lipopolysaccharide synthesis genes in two strains. Prophage deletion and induction were also noted in two strains, as well as mutations in several genes associated with virulence factors. Mutations in genes involved in susceptibility to conventional antibiotics were also identified in BIMs, with both decreased and increased antibiotic sensitivity to various antibiotics being observed. These findings may have implications for future applications of lytic phage therapy.IMPORTANCELytic bacteriophages are viruses that infect and kill bacteria and can be used to treat difficult-to-treat bacterial infections, including biofilm-associated infections and multidrug-resistant bacteria. Pseudomonas aeruginosa is a bacterium that can cause life-threatening infections. Lytic bacteriophage therapy has been trialed in the treatment of P. aeruginosa infections; however, sometimes bacteria develop resistance to the bacteriophages. This study sheds light on the genetic mechanisms of such resistance, and how this might be harnessed to restore the sensitivity of multidrug-resistant P. aeruginosa to conventional antibiotics.
Collapse
Affiliation(s)
- Stephanie A Fong
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - George Bouras
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Ghais Houtak
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Roshan Nepal
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Sholeh Feizi
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Sandra Morales
- AmpliPhi Australia, Brookvale, New South Wales, Australia
| | - Alkis J Psaltis
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Peter-John Wormald
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Sarah Vreugde
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| |
Collapse
|
4
|
Ciudin A, Padulles B, Manasia P, Alcoberro J, Toma C, Popescu R, Persu C, Aguilar A. Reshaping Resistance: How Autovaccine Therapy Alters the Course of Recurrent Multidrug-Resistant Urinary Tract Infections. Life (Basel) 2025; 15:50. [PMID: 39859990 PMCID: PMC11766521 DOI: 10.3390/life15010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Urinary tract infections (UTIs) caused by multidrug-resistant (MDR) bacteria pose a considerable challenge due to high treatment failure rates and associated healthcare costs. This pioneering study evaluates the effectiveness of personalized autovaccine therapy in managing recurrent UTIs in patients with MDR bacteria, aiming to offer an innovative treatment that reduces antibiotic resistance and hospitalizations. METHODS In this prospective, single-center study, 40 patients with recurrent MDR UTIs received personalized sublingual autovaccines derived from their own bacterial isolates. The study assessed UTI recurrence rates, changes in antibiotic use, and hospitalization days over 12 months. RESULTS The autovaccine therapy significantly reduced UTI recurrence, with 67.5% of patients experiencing fewer infections. Antibiotic usage decreased by 74.4%, and total hospitalization days annually reduced from 400 to 216. A significant shift was observed from MDR to multi-susceptible bacterial profiles among participants. CONCLUSIONS This study is the first to demonstrate that autovaccine therapy is a safe and effective approach for managing recurrent UTIs caused by MDR bacteria, significantly lowering infection frequency, antibiotic needs, and hospitalization. These findings support integrating autovaccine therapy into standard UTI management to combat antibiotic resistance and lessen healthcare burdens.
Collapse
Affiliation(s)
- Alexandru Ciudin
- Urology Department, Hospital Universitari de Mollet, 08100 Barcelona, Spain; (B.P.); (P.M.); (J.A.); (A.A.)
| | - Bernat Padulles
- Urology Department, Hospital Universitari de Mollet, 08100 Barcelona, Spain; (B.P.); (P.M.); (J.A.); (A.A.)
| | - Pasqualino Manasia
- Urology Department, Hospital Universitari de Mollet, 08100 Barcelona, Spain; (B.P.); (P.M.); (J.A.); (A.A.)
| | - Josep Alcoberro
- Urology Department, Hospital Universitari de Mollet, 08100 Barcelona, Spain; (B.P.); (P.M.); (J.A.); (A.A.)
| | - Cristian Toma
- Urology Department, Spitalul Clinic “Prof. Dr. Th. Burghele”, 061344 Bucuresti, Romania; (R.P.); (C.P.)
| | - Răzvan Popescu
- Urology Department, Spitalul Clinic “Prof. Dr. Th. Burghele”, 061344 Bucuresti, Romania; (R.P.); (C.P.)
| | - Cristian Persu
- Urology Department, Spitalul Clinic “Prof. Dr. Th. Burghele”, 061344 Bucuresti, Romania; (R.P.); (C.P.)
| | - Antonio Aguilar
- Urology Department, Hospital Universitari de Mollet, 08100 Barcelona, Spain; (B.P.); (P.M.); (J.A.); (A.A.)
| |
Collapse
|
5
|
Sarkodie-Addo P, Osman AH, Aglomasa BC, Donkor ES. Phage therapy in the management of respiratory and pulmonary infections: a systematic review. Ther Adv Infect Dis 2025; 12:20499361241307841. [PMID: 39866829 PMCID: PMC11760135 DOI: 10.1177/20499361241307841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 11/27/2024] [Indexed: 01/28/2025] Open
Abstract
Background Lower respiratory tract infections (LRTIs) pose a significant threat to global health, causing more than 2 million deaths worldwide. This menace is intensified by the alarming increase in drug resistance, which limits the availability of effective antibiotics for bacterial respiratory infections. Consequently, there is an urgent demand for alternative therapeutic options. Phage therapy (PT) has re-emerged as a promising therapeutic approach and as an adjunct to antibiotic treatment. Objective This systematic review synthesises the application of PT for LRTIs in humans, providing unified and updated data on the evaluation of the safety and efficacy of PT for LRTIs. Design Systematic review. Data sources and methods Following the PRISMA guidelines, a comprehensive search strategy was carried out (spanning January 2000 - February 2024) in four databases: PubMed, Scopus, ScienceDirect and Web of Science to retrieve published records of PT for LRTIs in humans only. The reference list of each included study was evaluated for possible inclusion of other relevant articles. Results Among the 18 records that fulfilled the inclusion criteria, 70 patients were administered PT. Microbiologically, 71.42% (n = 50/70) of the patients improved; with either the eradication of the pathogen or a decrease in bacterial load, whilst 15.71% (n = 11/70) did not record any improvement. About 5.71% (n = 4/70) recorded a partial/incomplete improvement, whilst 7.14% (n = 5/70) of the patients microbiological outcomes were unspecified. Clinically, up to 74.29% (n = 52/70) of the patients improved, whilst 10.00% (n = 7/70) of the patients showed no improvement. Another 2.86% (n = 2/70) recorded partial/incomplete improvement, whilst 12.86% (n = 9/70) were uncategorized. Phage titres that yielded positive outcomes ranged from 105 to 1012 PFU/mL. Studies that achieved a substantial phage titre at the site of infection frequently observed notable improvements. Regarding the safety of PT, 77.78% (N = 14/18) of the studies did not record any adverse effects after PT was administered, whilst 16.66% (n = 3/18) of the studies reported adverse effects. Conclusion Based on recently published data originating mainly from observational studies, PT has shown considerable efficacy and safety in the treatment of LRTIs. However, there is a lack of uniform methodologies and protocols across different PT cases in the management of LRTIs. Consequently, there is a need for additional clinical studies to establish standardised pharmacokinetic elements and an overall protocol for PT. By doing so, we can fully unlock the potential of PT in effectively managing clinical bacterial infections, including LRTIs.
Collapse
Affiliation(s)
| | - Abdul-Halim Osman
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Bill Clinton Aglomasa
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Eric S. Donkor
- Department of Medical Microbiology, University of Ghana Medical School, P.O. Box KB 4236, Accra, Ghana
| |
Collapse
|
6
|
Gil-Gil T, Laborda P, Martínez JL, Hernando-Amado S. Use of adjuvants to improve antibiotic efficacy and reduce the burden of antimicrobial resistance. Expert Rev Anti Infect Ther 2025; 23:31-47. [PMID: 39670956 DOI: 10.1080/14787210.2024.2441891] [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: 06/26/2024] [Revised: 10/28/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION The increase in antibiotic resistance, together with the absence of novel antibiotics, makes mandatory the introduction of novel strategies to optimize the use of existing antibiotics. Among these strategies, the use of molecules that increase their activity looks promising. AREAS COVERED Different categories of adjuvants have been reviewed. Anti-resistance adjuvants increase the activity of antibiotics by inhibiting antibiotic resistance determinants. Anti-virulence approaches focus on the infection process itself; reducing virulence in combination with an antibiotic can improve therapeutic efficacy. Combination of phages with antibiotics can also be useful, since they present different mechanisms of action and targets. Finally, combining antibiotics with adjuvants in the same molecule may serve to improve antibiotics' efficacy and to overcome potential problems of differential pharmacokinetics/pharmacodynamics. EXPERT OPINION The successful combination of inhibitors of β-lactamases with β-lactams has shown that adjuvants can improve the efficacy of current antibiotics. In this sense, novel anti-resistance adjuvants able to inhibit efflux pumps are still needed, as well as anti-virulence compounds that improve the efficacy of antibiotics by interfering with the infection process. Although adjuvants may present different pharmacodynamics/pharmacokinetics than antibiotics, conjugates containing both compounds can solve this problem. Finally, already approved drugs can be a promising source of antibiotic adjuvants.
Collapse
Affiliation(s)
- Teresa Gil-Gil
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Pablo Laborda
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| | | | | |
Collapse
|
7
|
Teymouri S, Yousefi MH, Heidari S, Farokhi S, Afkhami H, Kashfi M. Beyond antibiotics: mesenchymal stem cells and bacteriophages-new approaches to combat bacterial resistance in wound infections. Mol Biol Rep 2024; 52:64. [PMID: 39699690 DOI: 10.1007/s11033-024-10163-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
Wound management is a major global health problem. With the rising incidence of diabetic wounds, accidents, and other injuries, the demand for prompt wound treatment has become increasingly critical. Millions of people suffer from serious, large wounds resulting from major accidents, surgeries, and wars. These wounds require considerable time to heal and are susceptible to infection. Furthermore, chronic wounds, particularly in elderly and diabetic patients, often require frequent medical interventions to prevent complications. Consequently, wound management imposes a significant economic burden worldwide. The complications arising from wound infections can vary from localized issues to systemic effects. The most severe local complication of wound infection is the non-healing, which results from the disruption of the wound-healing process. This often leads to significant pain, discomfort, and psychological trauma for the patient. Systemic complications may include cellulitis, osteomyelitis, and septicemia. Mesenchymal stem cells are characterized by their high capacity for division, making them suitable candidates for the treatment of tissue damage. Additionally, they produce antimicrobial peptides and various cytokines, which enhance their antimicrobial activity. Evidence shows that phages are effective in treating wound-related infections, and phage therapy has proven to be highly effective for patients when administered correctly. The purpose of this article is to explore the use of bacteriophages and mesenchymal stem cells in wound healing and infection management.
Collapse
Affiliation(s)
- Samane Teymouri
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | | | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
| | - Mojtaba Kashfi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Fellowship in Clinical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Olorundare OO, Zrelovs N, Kabantiyok D, Svanberga K, Jansons J, Kazaks A, Agada GO, Agu CG, Morenikeji OR, Oluwapelumi OA, Dung T, Pewan SB. Isolation and Characterization of a Novel Jumbo Phage HPP-Temi Infecting Pseudomonas aeruginosa Pa9 and Increasing Host Sensitivity to Ciprofloxacin. Antibiotics (Basel) 2024; 13:1006. [PMID: 39596701 PMCID: PMC11591403 DOI: 10.3390/antibiotics13111006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Pseudomonas aeruginosa is a bacteria responsible for many hospital-acquired infections. Phages are promising alternatives for treating P. aeruginosa infections, which are often intrinsically resistant. The combination of phage and antibiotics in clearing bacterial infection holds promise due to increasing reports of enhanced effectiveness when both are used together. The aim of the study is to isolate and characterize a novel P. aeruginosa phage and determine its effectiveness in in vitro combination with antibiotics in controlling P. aeruginosa. In this study, a novel jumbo myophage HPP-Temi infecting P. aeruginosa Pa9 (PP334386) was isolated from household sewage. Electron micrographs of the phage were obtained to determine the morphological features of HPP-Temi virions. Complete genome analysis and a combination of Pseudomonas phage HPP-Temi with antibiotics were examined. The phage HPP-Temi was able to productively infect P. aeruginosa ATCC 9027 but was unable to infect a closely related genus. The phage was stable at 4-37 °C, 0.5% NaCl, and pH 8 for at least one hour. The HPP-Temi genome is a 302,719-bp-long dsDNA molecule with a GC content of 46.46%. The genome was predicted to have 436 ORFs and 7 tRNA genes. No virulence factor-related genes, antimicrobial resistance, or temperate lifestyle-associated genes were found in the phage HPP-Temi genome. Phage HPP-Temi is most closely related to the known or tentative representatives of the Pawinskivirus genus and can be proposed as a representative for the creation of a novel phage species in that genus. The phage and antibiotics (Ciprofloxacin) combination at varying phage titers (103, 106, 109) were used against P. aeruginosa Pa9 (PP334386) at 3.0 × 108 CFU/mL, which was carried out in triplicate. The result showed that combining antibiotics with phage significantly reduced the bacteria count at 103 and 106 titers, while no growth was observed at 109 PFU/mL. This suggests that the effect of phage HPP-Temi in combination with antibiotics is a potential and promising agent for the control of P. aeruginosa infections.
Collapse
Affiliation(s)
- Olufunke Olufunmilola Olorundare
- Department of Medical Microbiology, Clinical Sciences, University of Jos, Jos 930105, Nigeria
- Forest Research Institute of Nigeria, Federal College of Forestry, Jos 930105, Nigeria
| | - Nikita Zrelovs
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (N.Z.); (K.S.); (J.J.); (A.K.)
| | - Dennis Kabantiyok
- Fleming Sentinel Lab for AMR, National Veterinary Research Institute NVRI, Vom 930001, Nigeria; (D.K.); (G.O.A.); (O.A.O.); (T.D.)
| | - Karina Svanberga
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (N.Z.); (K.S.); (J.J.); (A.K.)
| | - Juris Jansons
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (N.Z.); (K.S.); (J.J.); (A.K.)
| | - Andris Kazaks
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (N.Z.); (K.S.); (J.J.); (A.K.)
| | - Godwin Ojonugwa Agada
- Fleming Sentinel Lab for AMR, National Veterinary Research Institute NVRI, Vom 930001, Nigeria; (D.K.); (G.O.A.); (O.A.O.); (T.D.)
| | - Chibuzor Gerald Agu
- Bacteria Research National Veterinary Research Institute NVRI, Vom 930001, Nigeria;
| | - Oluwatoyin Ruth Morenikeji
- West African Centre for Emerging Infectious Diseases, Jos University Teaching Hospital, Jos 930241, Nigeria;
| | - Ogundeji Alice Oluwapelumi
- Fleming Sentinel Lab for AMR, National Veterinary Research Institute NVRI, Vom 930001, Nigeria; (D.K.); (G.O.A.); (O.A.O.); (T.D.)
| | - Thomas Dung
- Fleming Sentinel Lab for AMR, National Veterinary Research Institute NVRI, Vom 930001, Nigeria; (D.K.); (G.O.A.); (O.A.O.); (T.D.)
| | | |
Collapse
|
9
|
Miller IP, Laney AG, Zahn G, Sheehan BJ, Whitley KV, Kuddus RH. Isolation and preliminary characterization of a novel bacteriophage vB_KquU_φKuK6 that infects the multidrug-resistant pathogen Klebsiella quasipneumoniae. Front Microbiol 2024; 15:1472729. [PMID: 39479209 PMCID: PMC11524547 DOI: 10.3389/fmicb.2024.1472729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Background Klebsiella quasipneumoniae (previously known as K. pneumoniae K6) strains are among the multidrug-resistant hypervirulent bacterial pathogens. Phage therapy can help treat infections caused by such pathogens. Here we report some aspects of virology and therapeutic potentials of vB_KquU_φKuK6, a bacteriophage that infects Klebsiella quasipneumoniae. Methods K. quasipneumoniae (ATCC 700603) was used to screen wastewater lytic phages. The isolate vB_KquU_φKuK6 that consistently created large clear plaques was characterized using standard virological and molecular methods. Results vB_KquU_φKuK6 has a complex capsid with an icosahedral head (~60 nm) and a slender tail (~140 nm × 10 nm). The phage has a 51% AT-rich linear dsDNA genome (51,251 bp) containing 121 open reading frames. The genome contains genes encoding spanin, endolysin, and holin proteins necessary for lytic infection and a recombinase gene possibly involved in lysogenic infection. vB_KquU_φKuK6 is stable at -80 to +67°C, pH 4-9, and brief exposure to one volume percent of chloroform. vB_KquU_φKuK6 has a narrow host range. Its lytic infection cycle involves a latency of 20 min and a burst size of 435 plaque-forming units. The phage can cause lysogenic infection, and the resulting lysogens are resistant to lytic infection by vB_KquU_φKuK6. vB_KquU_φKuK6 reduces the host cells' ability to form biofilm but fails to eliminate that ability. vB_KquU_φKuK6 demonstrates phage-antibiotic synergy and reduces the minimum inhibitory concentration of chloramphenicol and neomycin sulfate by about 8 folds. Conclusion vB_KquU_φKuK6 cannot be directly used for phage therapy because it is a temperate bacteriophage. However, genetically modified strains of vB_KquU_φKuK6 alone or combined with antibiotics or other lytic Klebsiella phages can have therapeutic utilities in treating K. quasipneumoniae infections.
Collapse
Affiliation(s)
| | | | | | | | | | - Ruhul H. Kuddus
- Department of Biology, Utah Valley University, Orem, UT, United States
| |
Collapse
|
10
|
Teymouri S, Pourhajibagher M, Bahador A. A review of the fighting Acinetobacter baumannii on three fronts: antibiotics, phages, and nanoparticles. Mol Biol Rep 2024; 51:1044. [PMID: 39377967 DOI: 10.1007/s11033-024-09979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/30/2024] [Indexed: 02/06/2025]
Abstract
In the current era of antibiotic resistance, researchers are exploring alternative ways to treat bacterial infections that are resistant to multiple drugs. Acinetobacter baumannii (A. baumannii) is a bacterium that is commonly encountered in clinical settings and is known to be resistant to several drugs. Due to the increase in drug-resistant infections caused by this bacteria, there is an urgent need to investigate alternative treatment options such as phage therapy and combination therapy. Despite the success of phages in some cases, there are some limitations in their clinical application that can be overcome by combining phages with other substrates such as nanoparticles to improve their function. The integration of nanotechnology with phage therapy against A. baumannii promises to overcome antibiotic resistance. By exploiting the targeted delivery and controlled release capabilities of nanoparticles, we can enhance the therapeutic potential of phages while minimizing their limitations. Continued research in this field will undoubtedly pave the way for more effective and precise treatments against A. baumannii infections and provide hope in the fight against antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Samane Teymouri
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Pourhajibagher
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Abbas Bahador
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Fellowship in Clinical Laboratory Sciences, BioHealth Lab, Tehran, Iran.
| |
Collapse
|
11
|
Cui L, Watanabe S, Miyanaga K, Kiga K, Sasahara T, Aiba Y, Tan XE, Veeranarayanan S, Thitiananpakorn K, Nguyen HM, Wannigama DL. A Comprehensive Review on Phage Therapy and Phage-Based Drug Development. Antibiotics (Basel) 2024; 13:870. [PMID: 39335043 PMCID: PMC11428490 DOI: 10.3390/antibiotics13090870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Phage therapy, the use of bacteriophages (phages) to treat bacterial infections, is regaining momentum as a promising weapon against the rising threat of multidrug-resistant (MDR) bacteria. This comprehensive review explores the historical context, the modern resurgence of phage therapy, and phage-facilitated advancements in medical and technological fields. It details the mechanisms of action and applications of phages in treating MDR bacterial infections, particularly those associated with biofilms and intracellular pathogens. The review further highlights innovative uses of phages in vaccine development, cancer therapy, and as gene delivery vectors. Despite its targeted and efficient approach, phage therapy faces challenges related to phage stability, immune response, and regulatory approval. By examining these areas in detail, this review underscores the immense potential and remaining hurdles in integrating phage-based therapies into modern medical practices.
Collapse
Affiliation(s)
- Longzhu Cui
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Shinya Watanabe
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Kazuhiko Miyanaga
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Kotaro Kiga
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Teppei Sasahara
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Yoshifumi Aiba
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Xin-Ee Tan
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Srivani Veeranarayanan
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Kanate Thitiananpakorn
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Huong Minh Nguyen
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Dhammika Leshan Wannigama
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
| |
Collapse
|
12
|
Vasileiadis A, Bozidis P, Konstantinidis K, Kesesidis N, Potamiti L, Kolliopoulou A, Beloukas A, Panayiotidis MI, Havaki S, Gorgoulis VG, Gartzonika K, Karakasiliotis I. A Novel Dhillonvirus Phage against Escherichia coli Bearing a Unique Gene of Intergeneric Origin. Curr Issues Mol Biol 2024; 46:9312-9329. [PMID: 39329903 PMCID: PMC11430396 DOI: 10.3390/cimb46090551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
Antibiotics resistance is expanding amongst pathogenic bacteria. Phage therapy is a revived concept for targeting bacteria with multiple antibiotics resistances. In the present study, we isolated and characterized a novel phage from hospital treatment plant input, using Escherichia coli (E. coli) as host bacterium. Phage lytic activity was detected by using soft agar assay. Whole-genome sequencing of the phage was performed by using Next-Generation Sequencing (NGS). Host range was determined using other species of bacteria and representative genogroups of E. coli. Whole-genome sequencing of the phage revealed that Escherichia phage Ioannina is a novel phage within the Dhillonvirus genus, but significantly diverged from other Dhillonviruses. Its genome is a 45,270 bp linear double-stranded DNA molecule that encodes 61 coding sequences (CDSs). The coding sequence of CDS28, a putative tail fiber protein, presented higher similarity to representatives of other phage families, signifying a possible recombination event. Escherichia phage Ioannina lytic activity was broad amongst the E. coli genogroups of clinical and environmental origin with multiple resistances. This phage may present in the future an important therapeutic tool against bacterial strains with multiple antibiotic resistances.
Collapse
Affiliation(s)
- Anastasios Vasileiadis
- Laboratory of Biology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (A.V.); (K.K.); (N.K.)
- Department of Microbiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45332 Ioannina, Greece; (P.B.); (K.G.)
| | - Petros Bozidis
- Department of Microbiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45332 Ioannina, Greece; (P.B.); (K.G.)
| | - Konstantinos Konstantinidis
- Laboratory of Biology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (A.V.); (K.K.); (N.K.)
| | - Nikolaos Kesesidis
- Laboratory of Biology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (A.V.); (K.K.); (N.K.)
| | - Louiza Potamiti
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.P.); (M.I.P.)
| | - Anna Kolliopoulou
- Molecular Microbiology and Immunology Laboratory, Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece; (A.K.); (A.B.)
| | - Apostolos Beloukas
- Molecular Microbiology and Immunology Laboratory, Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece; (A.K.); (A.B.)
| | - Mihalis I. Panayiotidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.P.); (M.I.P.)
| | - Sophia Havaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.H.); (V.G.G.)
| | - Vassilis G. Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.H.); (V.G.G.)
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4GJ, UK
- Faculty of Health and Medical Sciences, University of Surrey, Surrey GU2 7YH, UK
| | - Konstantina Gartzonika
- Department of Microbiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45332 Ioannina, Greece; (P.B.); (K.G.)
| | - Ioannis Karakasiliotis
- Laboratory of Biology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (A.V.); (K.K.); (N.K.)
| |
Collapse
|
13
|
Bin Yahia NM, Shan M, Zhu Y, Yang Y, Zhang S, Yang Y. From crisis to cure: harnessing the potential of mycobacteriophages in the battle against tuberculosis. J Appl Microbiol 2024; 135:lxae208. [PMID: 39134510 DOI: 10.1093/jambio/lxae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/26/2024] [Accepted: 08/10/2024] [Indexed: 08/30/2024]
Abstract
Tuberculosis (TB) is a serious and fatal disease caused by Mycobacterium tuberculosis (Mtb). The World Health Organization reported an estimated 1.30 million TB-related deaths in 2022. The escalating prevalence of Mtb strains classified as being multi-, extensively, extremely, or totally drug resistant, coupled with the decreasing efficacies of conventional therapies, necessitates the development of novel treatments. As viruses that infect Mycobacterium spp., mycobacteriophages may represent a strategy to combat and eradicate drug-resistant TB. More exploration is needed to provide a comprehensive understanding of mycobacteriophages and their genome structure, which could pave the way toward a definitive treatment for TB. This review focuses on the properties of mycobacteriophages, their potential in diagnosing and treating TB, the benefits and drawbacks of their application, and their use in human health. Specifically, we summarize recent research on mycobacteriophages targeted against Mtb infection and newly developed mycobacteriophage-based tools to diagnose and treat diseases caused by Mycobacterium spp. We underscore the urgent need for innovative approaches and highlight the potential of mycobacteriophages as a promising avenue for developing effective diagnosis and treatment to combat drug-resistant Mycobacterium strains.
Collapse
Affiliation(s)
- Noura M Bin Yahia
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 P.R. China
| | - Minghai Shan
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 P.R. China
- General Hospital of Ningxia Medical University, Yinchuan, 750004 P.R. China
| | - Yue Zhu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 P.R. China
| | - Yuma Yang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 P.R. China
| | - Sihan Zhang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 P.R. China
| | - Yanhui Yang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 P.R. China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004 P.R. China
| |
Collapse
|
14
|
Sivarajan K, Ravindhiran R, Sekar JN, Murugesan R, Chidambaram K, Dhandapani K. Deciphering the impact of Acinetobacter baumannii on human health, and exploration of natural compounds as efflux pump inhibitors to treat multidrug resistance. J Med Microbiol 2024; 73. [PMID: 39212030 DOI: 10.1099/jmm.0.001867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Acinetobacter baumannii is an ESKAPE pathogen and threatens human health by generating infections with high fatality rates. A. baumannii leads to a spectrum of infections such as skin and wound infections, endocarditis, meningitis pneumonia, septicaemia and urinary tract infections. Recently, strains of A. baumannii have emerged as multidrug-resistant (MDR), meaning they are resistant to at least three different classes of antibiotics. MDR development is primarily intensified by widespread antibiotic misuse and inadequate stewardship. The World Health Organization (WHO) declared A. baumannii a precarious MDR species. A. baumannii maintains the MDR phenotype via a diverse array of antimicrobial metabolite-hydrolysing enzymes, efflux of antibiotics, impermeability and antibiotic target modification, thereby complicating treatment. Hence, a deeper understanding of the resistance mechanisms employed by MDR A. baumannii can give possible approaches to treat antimicrobial resistance. Resistance-nodulation-cell division (RND) efflux pumps have been identified as the key contributors to MDR determinants, owing to their capacity to force a broad spectrum of chemical substances out of the bacterial cell. Though synthetic inhibitors have been reported previously, their efficacy and safety are of debate. As resistance-modifying agents, phytochemicals are ideal choices. These natural compounds could eliminate the bacteria or interact with pathogenicity events and reduce the bacteria's ability to evolve resistance. This review aims to highlight the mechanism behind the multidrug resistance in A. baumannii and elucidate the utility of natural compounds as efflux pump inhibitors to deal with the infections caused by A. baumannii.
Collapse
Affiliation(s)
- Karthiga Sivarajan
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | - Ramya Ravindhiran
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | - Jothi Nayaki Sekar
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | - Rajeswari Murugesan
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | - Kumarappan Chidambaram
- Department of Pharmacology and Toxicology, School of Pharmacy, King Khalid University, Abha 652529, Saudi Arabia
| | - Kavitha Dhandapani
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| |
Collapse
|
15
|
Cebani L, Mvubu NE. Can We Exploit Inflammasomes for Host-Directed Therapy in the Fight against Mycobacterium tuberculosis Infection? Int J Mol Sci 2024; 25:8196. [PMID: 39125766 PMCID: PMC11311975 DOI: 10.3390/ijms25158196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (M. tb), is a major global health issue, with around 10 million new cases annually. Advances in TB immunology have improved our understanding of host signaling pathways, leading to innovative therapeutic strategies. Inflammasomes, protein complexes organized by cytosolic pattern recognition receptors (PRRs), play a crucial role in the immune response to M. tb by activating caspase 1, which matures proinflammatory cytokines IL1β and IL18. While inflammation is necessary to fight infection, excessive or dysregulated inflammation can cause tissue damage, highlighting the need for precise inflammasome regulation. Drug-resistant TB strains have spurred research into adjunctive host-directed therapies (HDTs) that target inflammasome pathways to control inflammation. Canonical and non-canonical inflammasome pathways can trigger excessive inflammation, leading to immune system exhaustion and M. tb spread. Novel HDT interventions can leverage precision medicine by tailoring treatments to individual inflammasome responses. Studies show that medicinal plant derivatives like silybin, andrographolide, and micheliolide and small molecules such as OLT1177, INF39, CY-09, JJ002, Ac-YVAD-cmk, TAK-242, and MCC950 can modulate inflammasome activation. Molecular tools like gene silencing and knockouts may also be used for severe TB cases. This review explores these strategies as potential adjunctive HDTs in fighting TB.
Collapse
Affiliation(s)
| | - Nontobeko E. Mvubu
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa;
| |
Collapse
|
16
|
Subramanian A. Emerging roles of bacteriophage-based therapeutics in combating antibiotic resistance. Front Microbiol 2024; 15:1384164. [PMID: 39035437 PMCID: PMC11257900 DOI: 10.3389/fmicb.2024.1384164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/06/2024] [Indexed: 07/23/2024] Open
Abstract
Amid the growing challenge of antibiotic resistance on a global scale, there has been a notable resurgence in bacteriophage-based treatments, signaling a shift in our approach to managing infections. Bacteriophages (BPs), bacterial predators of nature, present a promising alternative for tackling infections caused by antibiotic-resistant pathogens. This review delves into the intricate relationship between bacteriophages and resistant bacteria, exploring various treatment strategies. Drawing upon both preclinical and clinical studies, the review highlights the effectiveness of bacteriophage therapy, particularly when integrated synergistically with conventional antibiotics. It discusses various treatment approaches for systemic and localized infections, demonstrating the adaptability of bacteriophage therapy across different clinical scenarios. Furthermore, the formulation and delivery of bacteriophages shed light on the various methods used to encapsulate and administer them effectively. It also acknowledges the challenge of bacterial resistance to bacteriophages and the ongoing efforts to overcome this hurdle. In addition, this review highlights the importance of the bacteriophage sensitivity profile (phagogram), which helps tailor treatment regimens to individual patients and specific pathogens. By surpassing the limitations of traditional antibiotics, bacteriophage-based therapies offer a personalized and potent solution against antibiotic resistance, promising to reshape the future of infectious disease management.
Collapse
|
17
|
Osman AH, Darkwah S, Kotey FCN, Odoom A, Hotor P, Dayie NTKD, Donkor ES. Reservoirs of Nosocomial Pathogens in Intensive Care Units: A Systematic Review. ENVIRONMENTAL HEALTH INSIGHTS 2024; 18:11786302241243239. [PMID: 38828046 PMCID: PMC11141231 DOI: 10.1177/11786302241243239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/14/2024] [Indexed: 06/05/2024]
Abstract
Background Nosocomial pathogens are known to exacerbate morbidity and mortality in contemporary critical healthcare. Hospital fomites, which include inanimate surfaces, have been identified as "breeding grounds" for pathogens that cause nosocomial infections. This systematic review aimed to deliver incisive insights on nosocomial pathogens in intensive care units (ICUs) and the role of fomites as potential reservoirs for their transmission. Method An extensive exploration of electronic databases, including PubMed and Scopus, from 1990 to 2023, was carried out between 25th and 29th May 2023, per standard PRISMA guidelines. Information were extracted from articles that reported on fomites in the ICU. Studies that did not quantitatively report the fomite contamination, and those that exclusively took samples from patients in the ICU were excluded from the analysis. Results About 40% of the total samples collected on fomites from all the studies yielded microbial growth, with species of Staphylococcus being the most predominant. Other prevalent microbes were Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniae, Candida spp., Enterococcus sp., and Enterobacter sp. The neonatal intensive care unit (NICU) had the highest proportion of contaminated fomites. Among known fomites, the sphygmomanometer exhibited a 100% detection rate of nosocomial pathogens. This included E. aerogenes, Staphylococcus aureus, coagulase-negative Staphylococci (CoNS), E. coli, and K. pneumoniae. Multidrug-resistant (MDR) bacteria, such as methicillin-resistant S. aureus (MRSA), vancomycin-resistant Enterococci (VRE), extended-spectrum beta-lactamase (ESBL)-producing E. coli, and MDR Pseudomonas aeruginosa were commonly isolated on fomites in the ICUs. Conclusion Many fomites that are readily used in patient care in the ICU harbour nosocomial pathogens. The most common fomite appeared to be mobile phones, sphygmomanometers, and stethoscopes, with Staphylococcus being the most common contaminant. Consequently, the need for rigorous disinfection and sterilization protocols on fomites in the ICU cannot be overemphasized. Additionally, heightened awareness on the subject among health professionals is crucial to mitigating the risk and burden of nosocomial infections caused by drug-resistant bacteria.
Collapse
Affiliation(s)
- Abdul-Halim Osman
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Samuel Darkwah
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Fleischer C N Kotey
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Alex Odoom
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Prince Hotor
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Nicholas T K D Dayie
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Eric S Donkor
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| |
Collapse
|
18
|
Buendia P, Fernandez K, Raley C, Rahnavard A, Crandall KA, Castro JG. Hospital antimicrobial stewardship: profiling the oral microbiome after exposure to COVID-19 and antibiotics. Front Microbiol 2024; 15:1346762. [PMID: 38476940 PMCID: PMC10927822 DOI: 10.3389/fmicb.2024.1346762] [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: 11/29/2023] [Accepted: 01/22/2024] [Indexed: 03/14/2024] Open
Abstract
Introduction During the COVID-19 Delta variant surge, the CLAIRE cross-sectional study sampled saliva from 120 hospitalized patients, 116 of whom had a positive COVID-19 PCR test. Patients received antibiotics upon admission due to possible secondary bacterial infections, with patients at risk of sepsis receiving broad-spectrum antibiotics (BSA). Methods The saliva samples were analyzed with shotgun DNA metagenomics and respiratory RNA virome sequencing. Medical records for the period of hospitalization were obtained for all patients. Once hospitalization outcomes were known, patients were classified based on their COVID-19 disease severity and the antibiotics they received. Results Our study reveals that BSA regimens differentially impacted the human salivary microbiome and disease progression. 12 patients died and all of them received BSA. Significant associations were found between the composition of the COVID-19 saliva microbiome and BSA use, between SARS-CoV-2 genome coverage and severity of disease. We also found significant associations between the non-bacterial microbiome and severity of disease, with Candida albicans detected most frequently in critical patients. For patients who did not receive BSA before saliva sampling, our study suggests Staphylococcus aureus as a potential risk factor for sepsis. Discussion Our results indicate that the course of the infection may be explained by both monitoring antibiotic treatment and profiling a patient's salivary microbiome, establishing a compelling link between microbiome and the specific antibiotic type and timing of treatment. This approach can aid with emergency room triage and inpatient management but also requires a better understanding of and access to narrow-spectrum agents that target pathogenic bacteria.
Collapse
Affiliation(s)
| | | | - Castle Raley
- The George Washington University Genomics Core, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Ali Rahnavard
- Department of Biostatistics and Bioinformatics, Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Keith A. Crandall
- The George Washington University Genomics Core, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
- Department of Biostatistics and Bioinformatics, Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Jose Guillermo Castro
- Division of Infectious Diseases, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
19
|
Boakye-Yiadom E, Odoom A, Osman AH, Ntim OK, Kotey FCN, Ocansey BK, Donkor ES. Fungal Infections, Treatment and Antifungal Resistance: The Sub-Saharan African Context. Ther Adv Infect Dis 2024; 11:20499361241297525. [PMID: 39544852 PMCID: PMC11562003 DOI: 10.1177/20499361241297525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024] Open
Abstract
Fungal pathogens cause a wide range of infections in humans, from superficial to disfiguring, allergic syndromes, and life-threatening invasive infections, affecting over a billion individuals globally. With an estimated 1.5 million deaths annually attributable to them, fungal pathogens are a major cause of mortality in humans, especially people with underlying immunosuppression. The continuous increase in the population of individuals at risk of fungal infections in sub-Saharan Africa, such as HIV patients, tuberculosis patients, intensive care patients, patients with haematological malignancies, transplant (haematopoietic stem cell and organ) recipients and the growing global threat of multidrug-resistant fungal strains, raise the need for an appreciation of the region's perspective on antifungal usage and resistance. In addition, the unavailability of recently introduced novel antifungal drugs in sub-Saharan Africa further calls for regular evaluation of resistance to antifungal agents in these settings. This is critical for ensuring appropriate and optimal use of the limited available arsenal to minimise antifungal resistance. This review, therefore, elaborates on the multifaceted nature of fungal resistance to the available antifungal drugs on the market and further provides insights into the prevalence of fungal infections and the use of antifungal agents in sub-Saharan Africa.
Collapse
Affiliation(s)
- Emily Boakye-Yiadom
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
- Department of Microbiology and Immunology, University of Health and Allied Sciences, Ho, Ghana
| | - Alex Odoom
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Abdul-Halim Osman
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Onyansaniba K. Ntim
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Fleischer C. N. Kotey
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Bright K. Ocansey
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Eric S. Donkor
- Department of Medical Microbiology, University of Ghana Medical School, Accra, P.O. Box KB 4236, Ghana
| |
Collapse
|
20
|
El-Nour SAA, Hammad AA, Fathy R, Eid AS. Application of coliphage as biocontrol agent in combination with gamma irradiation to eliminate multi-drug-resistant E. coli in minimally processed vegetables. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:123907-123924. [PMID: 37995029 PMCID: PMC10746767 DOI: 10.1007/s11356-023-31071-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/12/2023] [Indexed: 11/24/2023]
Abstract
Biofilm formation is a rising concern in the food industry. Escherichia coli (E. coli) is one of the most important food-borne pathogens that can survive in food and food-related environments and eventually produce biofilms. This study suggested that both coliphages used were successful in preventing the creation of new biofilms as well as removing existing ones. Confocal laser scanning microscopy verified these findings. According to the findings, neither coliphage survived at 37 °C, but both remained stable at 4 °C and - 20 °C for extended periods of time. The study revealed that both coliphages demonstrated a greater degree of gamma irradiation resistance when compared to E. coli. The study's results indicate that the implementation of a dual method, which incorporates gamma irradiation (1.5 kGy) and coliphage treatment, on various kinds of vegetables that were infected with E. coli, resulted in a significant reduction in bacterial count (surpassing 99.99%) following a 24-h incubation period. Combining gamma irradiation and the coliphage approach was significantly effective at lowering polysaccharide concentrations and proteins in the biofilm matrix. The results revealed that the pairing of gamma irradiation and coliphages acted in conjunction to cause disruptions in the matrix of biofilm, thereby promoting cell removal compared with either of the individual treatments. Ca+ ions strengthen the weak virion interaction with the relevant bacterial host cell receptors during the adsorption process. In conclusion, use of coliphage in combination with gamma irradiation treatment can be applied to improve fresh produce's microbial safety and enhance its storability in supermarkets.
Collapse
Affiliation(s)
- Salwa A Abou El-Nour
- Radiation Microbiology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Ali A Hammad
- Radiation Microbiology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Reham Fathy
- Radiation Microbiology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt.
| | - Amal S Eid
- Radiation Microbiology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| |
Collapse
|
21
|
Adefisoye MA, Olaniran AO. Antimicrobial resistance expansion in pathogens: a review of current mitigation strategies and advances towards innovative therapy. JAC Antimicrob Resist 2023; 5:dlad127. [PMID: 38089461 PMCID: PMC10712721 DOI: 10.1093/jacamr/dlad127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024] Open
Abstract
The escalating problem of antimicrobial resistance (AMR) proliferation in clinically important pathogens has become one of the biggest threats to human health and the global economy. Previous studies have estimated AMR-associated deaths and disability-adjusted life-years (DALYs) in many countries with a view to presenting a clearer picture of the global burden of AMR-related diseases. Recently, several novel strategies have been advanced to combat resistance spread. These include efflux activity inhibition, closing of mutant selection window (MSW), biofilm disruption, lytic bacteriophage particles, nanoantibiotics, engineered antimicrobial peptides, and the CRISPR-Cas9 gene-editing technique. The single or integrated deployment of these strategies has shown potentialities towards mitigating resistance and contributing to valuable therapeutic outcomes. Correspondingly, the new paradigm of personalized medicine demands innovative interventions such as improved and accurate point-of-care diagnosis and treatment to curtail AMR. The CRISPR-Cas system is a novel and highly promising nucleic acid detection and manipulating technology with the potential for application in the control of AMR. This review thus considers the specifics of some of the AMR-mitigating strategies, while noting their drawbacks, and discusses the advances in the CRISPR-based technology as an important point-of-care tool for tracking and curbing AMR in our fight against a looming 'post-antibiotic' era.
Collapse
Affiliation(s)
- Martins A Adefisoye
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, Westville Campus, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa
- Department of Microbiology, School of Science and Technology, Babcock University, Ilishan-Remo, Nigeria
| | - Ademola O Olaniran
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, Westville Campus, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa
| |
Collapse
|
22
|
Kontoghiorghes GJ. Drug Selection and Posology, Optimal Therapies and Risk/Benefit Assessment in Medicine: The Paradigm of Iron-Chelating Drugs. Int J Mol Sci 2023; 24:16749. [PMID: 38069073 PMCID: PMC10706143 DOI: 10.3390/ijms242316749] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
The design of clinical protocols and the selection of drugs with appropriate posology are critical parameters for therapeutic outcomes. Optimal therapeutic protocols could ideally be designed in all diseases including for millions of patients affected by excess iron deposition (EID) toxicity based on personalised medicine parameters, as well as many variations and limitations. EID is an adverse prognostic factor for all diseases and especially for millions of chronically red-blood-cell-transfused patients. Differences in iron chelation therapy posology cause disappointing results in neurodegenerative diseases at low doses, but lifesaving outcomes in thalassemia major (TM) when using higher doses. In particular, the transformation of TM from a fatal to a chronic disease has been achieved using effective doses of oral deferiprone (L1), which improved compliance and cleared excess toxic iron from the heart associated with increased mortality in TM. Furthermore, effective L1 and L1/deferoxamine combination posology resulted in the complete elimination of EID and the maintenance of normal iron store levels in TM. The selection of effective chelation protocols has been monitored by MRI T2* diagnosis for EID levels in different organs. Millions of other iron-loaded patients with sickle cell anemia, myelodysplasia and haemopoietic stem cell transplantation, or non-iron-loaded categories with EID in different organs could also benefit from such chelation therapy advances. Drawbacks of chelation therapy include drug toxicity in some patients and also the wide use of suboptimal chelation protocols, resulting in ineffective therapies. Drug metabolic effects, and interactions with other metals, drugs and dietary molecules also affected iron chelation therapy. Drug selection and the identification of effective or optimal dose protocols are essential for positive therapeutic outcomes in the use of chelating drugs in TM and other iron-loaded and non-iron-loaded conditions, as well as general iron toxicity.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
23
|
Oluwarinde BO, Ajose DJ, Abolarinwa TO, Montso PK, Du Preez I, Njom HA, Ateba CN. Safety Properties of Escherichia coli O157:H7 Specific Bacteriophages: Recent Advances for Food Safety. Foods 2023; 12:3989. [PMID: 37959107 PMCID: PMC10650914 DOI: 10.3390/foods12213989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Shiga-toxin-producing Escherichia coli (STEC) is typically detected on food products mainly due to cross-contamination with faecal matter. The serotype O157:H7 has been of major public health concern due to the severity of illness caused, prevalence, and management. In the food chain, the main methods of controlling contamination by foodborne pathogens often involve the application of antimicrobial agents, which are now becoming less efficient. There is a growing need for the development of new approaches to combat these pathogens, especially those that harbour antimicrobial resistant and virulent determinants. Strategies to also limit their presence on food contact surfaces and food matrices are needed to prevent their transmission. Recent studies have revealed that bacteriophages are useful non-antibiotic options for biocontrol of E. coli O157:H7 in both animals and humans. Phage biocontrol can significantly reduce E. coli O157:H7, thereby improving food safety. However, before being certified as potential biocontrol agents, the safety of the phage candidates must be resolved to satisfy regulatory standards, particularly regarding phage resistance, antigenic properties, and toxigenic properties. In this review, we provide a general description of the main virulence elements of E. coli O157:H7 and present detailed reports that support the proposals that phages infecting E. coli O157:H7 are potential biocontrol agents. This paper also outlines the mechanism of E. coli O157:H7 resistance to phages and the safety concerns associated with the use of phages as a biocontrol.
Collapse
Affiliation(s)
- Bukola Opeyemi Oluwarinde
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mahikeng 2375, South Africa; (B.O.O.); (D.J.A.); (T.O.A.); (P.K.M.)
- Antimicrobial Resistance and Phage Bio-Control Research Group (AREPHABREG), Department of Microbiology, North-West University, Mahikeng 2735, South Africa
| | - Daniel Jesuwenu Ajose
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mahikeng 2375, South Africa; (B.O.O.); (D.J.A.); (T.O.A.); (P.K.M.)
- Antimicrobial Resistance and Phage Bio-Control Research Group (AREPHABREG), Department of Microbiology, North-West University, Mahikeng 2735, South Africa
| | - Tesleem Olatunde Abolarinwa
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mahikeng 2375, South Africa; (B.O.O.); (D.J.A.); (T.O.A.); (P.K.M.)
- Antimicrobial Resistance and Phage Bio-Control Research Group (AREPHABREG), Department of Microbiology, North-West University, Mahikeng 2735, South Africa
| | - Peter Kotsoana Montso
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mahikeng 2375, South Africa; (B.O.O.); (D.J.A.); (T.O.A.); (P.K.M.)
- Antimicrobial Resistance and Phage Bio-Control Research Group (AREPHABREG), Department of Microbiology, North-West University, Mahikeng 2735, South Africa
| | - Ilse Du Preez
- Centre for Human Metabolomics, North-West University, Potchefstroom 2531, South Africa;
| | - Henry Akum Njom
- Agricultural Research Council, Private Bag X1251, Potchefstroom 2531, South Africa;
| | - Collins Njie Ateba
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mahikeng 2375, South Africa; (B.O.O.); (D.J.A.); (T.O.A.); (P.K.M.)
- Antimicrobial Resistance and Phage Bio-Control Research Group (AREPHABREG), Department of Microbiology, North-West University, Mahikeng 2735, South Africa
| |
Collapse
|
24
|
Abedon ST. Automating Predictive Phage Therapy Pharmacology. Antibiotics (Basel) 2023; 12:1423. [PMID: 37760719 PMCID: PMC10525195 DOI: 10.3390/antibiotics12091423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/02/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Viruses that infect as well as often kill bacteria are called bacteriophages, or phages. Because of their ability to act bactericidally, phages increasingly are being employed clinically as antibacterial agents, an infection-fighting strategy that has been in practice now for over one hundred years. As with antibacterial agents generally, the development as well as practice of this phage therapy can be aided via the application of various quantitative frameworks. Therefore, reviewed here are considerations of phage multiplicity of infection, bacterial likelihood of becoming adsorbed as a function of phage titers, bacterial susceptibility to phages also as a function of phage titers, and the use of Poisson distributions to predict phage impacts on bacteria. Considered in addition is the use of simulations that can take into account both phage and bacterial replication. These various approaches can be automated, i.e., by employing a number of online-available apps provided by the author, the use of which this review emphasizes. In short, the practice of phage therapy can be aided by various mathematical approaches whose implementation can be eased via online automation.
Collapse
Affiliation(s)
- Stephen T Abedon
- Department of Microbiology, The Ohio State University, Mansfield, OH 44906, USA
| |
Collapse
|