1
|
Lu J, Wu H, Wu S, Wang S, Fan H, Ruan H, Qiao J, Caiyin Q, Wen M. Salmonella: Infection mechanism and control strategies. Microbiol Res 2024; 292:128013. [PMID: 39675139 DOI: 10.1016/j.micres.2024.128013] [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: 08/22/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
Salmonella is a foodborne pathogen that predominantly resides in the intestinal tract of humans and animals. Infections caused by Salmonella can lead to various illnesses, including gastroenteritis, bacteremia, septicemia, and focal infections, with severe cases potentially resulting in host mortality. The mechanisms by which Salmonella invades host cells and disseminates throughout the body are partly understood, but there are still many scientific questions to be solved. This review aims to synthesize existing research on the interactions between Salmonella and hosts, detailing a comprehensive infection mechanism from adhesion and invasion to intracellular propagation and systemic spread. Overuse of antibiotics contributes to the emergence of drug-resistant Salmonella strains. An in-depth analysis of the mechanism of Salmonella infection will provide a theoretical basis for the development of novel Salmonella control strategies. These innovative control strategies include antibiotic adjuvants, small molecules, phages, attenuated vaccines, and probiotic therapies, which show huge potential in controlling Salmonella infection.
Collapse
Affiliation(s)
- Juane Lu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Hao Wu
- Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China; School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Shengbo Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China
| | - Shengli Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China
| | - Hongfei Fan
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300072, China
| | - Haihua Ruan
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300072, China
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Qinggele Caiyin
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China.
| | - Mingzhang Wen
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China; Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.
| |
Collapse
|
2
|
Duarte J, Trindade D, Oliveira V, Gomes NCM, Calado R, Pereira C, Almeida A. Isolation and Characterization of Infection of Four New Bacteriophages Infecting a Vibrio parahaemolyticus Strain. Antibiotics (Basel) 2024; 13:1086. [PMID: 39596779 PMCID: PMC11591531 DOI: 10.3390/antibiotics13111086] [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: 10/01/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Bacteria from genus Vibrio continue to be one of the most common threats to aquaculture sustainability. Vibrio spp. have been associated with infectious outbreaks in fish, shrimp, bivalves and even algae farms worldwide. Moreover, several Vibrio spp. are also pathogens that impact human health and are a threat to public health when transferred to consumers through contaminated seafood products. The use of bacteriophages is an evolving technology that could be applied in the treatment of Vibrio spp. either to protect aquaculture farms or to decontaminate seafood, namely bivalves during their depuration. In the present study, bacteriophages vB_VpS_LMAVpS1 (S1) vB_VpS_LMAVpVPP (VPP), vB_VpS_LMAVpSH (SH) and vB_VpS_LMAVpH (H) infecting V. parahaemolyticus were isolated and characterized. All phages presented fast adsorption rates and were able to control V. parahaemolyticus at all multiplicity of infections (MOIs) tested (MOI of 1, 10 and 100), with reductions of more than 4 log CFU/mL being recorded, but only in the presence of divalent cation calcium. The rate of emergence of phage-resistant mutants was very low (1.8 × 10-6 to 3.1 × 10-6). Bacterial phage resistance was not permanent and led to a loss of bacterial fitness. All four phages presented with lysins encoded in their genomes. The results presented provide valuable insights for future studies in the application of these bacteriophages in different scenarios to control, decontaminate or treat bacterial infections or contaminations of V. parahaemolyticus.
Collapse
Affiliation(s)
- João Duarte
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (J.D.); (D.T.); (V.O.); (N.C.M.G.)
| | - David Trindade
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (J.D.); (D.T.); (V.O.); (N.C.M.G.)
| | - Vanessa Oliveira
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (J.D.); (D.T.); (V.O.); (N.C.M.G.)
| | - Newton C. M. Gomes
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (J.D.); (D.T.); (V.O.); (N.C.M.G.)
| | - Ricardo Calado
- Laboratory for Innovation and Sustainability of Marine Biological Resources of the University of Aveiro (ECOMARE), Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Carla Pereira
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (J.D.); (D.T.); (V.O.); (N.C.M.G.)
| | - Adelaide Almeida
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal; (J.D.); (D.T.); (V.O.); (N.C.M.G.)
| |
Collapse
|
3
|
Costa P, Pereira C, Romalde JL, Almeida A. A game of resistance: War between bacteria and phages and how phage cocktails can be the solution. Virology 2024; 599:110209. [PMID: 39186863 DOI: 10.1016/j.virol.2024.110209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024]
Abstract
While phages hold promise as an antibiotic alternative, they encounter significant challenges in combating bacterial infections, primarily due to the emergence of phage-resistant bacteria. Bacterial defence mechanisms like superinfection exclusion, CRISPR, and restriction-modification systems can hinder phage effectiveness. Innovative strategies, such as combining different phages into cocktails, have been explored to address these challenges. This review delves into these defence mechanisms and their impact at each stage of the infection cycle, their challenges, and the strategies phages have developed to counteract them. Additionally, we examine the role of phage cocktails in the evolving landscape of antibacterial treatments and discuss recent studies that highlight the effectiveness of diverse phage cocktails in targeting essential bacterial receptors and combating resistant strains.
Collapse
Affiliation(s)
- Pedro Costa
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Carla Pereira
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Jesús L Romalde
- Department of Microbiology and Parasitology, CRETUS & CIBUS - Faculty of Biology, University of Santiago de Compostela, CP 15782 Santiago de Compostela, Spain.
| | - Adelaide Almeida
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
4
|
Ibne Shoukani H, Nisa S, Bibi Y, Ishfaq A, Ali A, Alharthi S, Kubra KT, Zia M. Green synthesis of polyethylene glycol coated, ciprofloxacin loaded CuO nanoparticles and its antibacterial activity against Staphylococcus aureus. Sci Rep 2024; 14:21246. [PMID: 39261712 PMCID: PMC11390890 DOI: 10.1038/s41598-024-72322-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024] Open
Abstract
Antibacterial resistance requires an advanced strategy to increase the efficacy of current therapeutics in addition to the synthesis of new generations of antibiotics. In this study, copper oxide nanoparticles (CuO-NPs) were green synthesized using Moringa oleifera root extract. CuO-NPs fabricated into a form of aspartic acid-ciprofloxacin-polyethylene glycol coated copper oxide-nanotherapeutics (CIP-PEG-CuO) to improve the antibacterial activity of NPs and the efficacy of the drug with controlled cytotoxicity. These NPs were charachterized by Fourier transform infrared spectroscopy (FTIR), x-rays diffraction spectroscopy (XRD), scanning electron microscopy (SEM) and energy-dispersive spectroscopy (EDS). Antibacterial screening and bacterial chemotaxis investigations demonstrated that CIP-PEG-CuO NPs show enhanced antibacterial potential against Gram-positive and Gram-negative clinically isolated pathogenic bacterial strains as compared to CuO-NPs. In ex-vivo cytotoxicity CIP-PEG-CuO-nano-formulates revealed 88% viability of Baby Hamster Kidney 21 cell lines and 90% RBCs remained intact with nano-formulations during hemolysis assay. An in-vivo studies on animal models show that Staphylococcus aureus were eradicated by this newly developed formulate from the infected skin and showed wound-healing properties. By using specially designed nanoparticles that are engineered to precisely transport antimicrobial agents, these efficient nano-drug delivery systems can target localized infections, ensure targeted delivery, enhance efficacy through increased drug penetration through physical barriers, and reduce systemic side effects for more effective treatment.
Collapse
Affiliation(s)
| | - Sobia Nisa
- Department of Microbiology, The University of Haripur, Haripur, 22620, Pakistan.
| | - Yamin Bibi
- Department of Botany, Rawalpindi Women University, Rawalpindi, 4600, Pakistan
| | - Afsheen Ishfaq
- Department of Medicine, FRPMC/PAF Hospital Faisal, Karachi, 75350, Pakistan
| | - Ashraf Ali
- Department of Chemistry, Faculty of Physical & Applied Sciences, The University of Haripur, Haripur, 22780, Pakistan.
- School of Chemistry & Chemical Engineering , Henan University of Technology, Zhengzhou, 450001, China.
| | - Sarah Alharthi
- Department of Chemistry, College of Science, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
- Research Center of Basic Sciences, Engineering and High Altitude, Taif University, 21944, Taif, Saudi Arabia
| | - Khudija Tul Kubra
- Department of Microbiology, The University of Haripur, Haripur, 22620, Pakistan
| | - Muhammad Zia
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| |
Collapse
|
5
|
Martinho I, Braz M, Duarte J, Brás A, Oliveira V, Gomes NCM, Pereira C, Almeida A. The Potential of Phage Treatment to Inactivate Planktonic and Biofilm-Forming Pseudomonas aeruginosa. Microorganisms 2024; 12:1795. [PMID: 39338470 PMCID: PMC11433742 DOI: 10.3390/microorganisms12091795] [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: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Pseudomonas aeruginosa is a common cause of hospital-acquired infections and exhibits a strong resistance to antibiotics. An alternative treatment option for bacterial infections is the use of bacteriophages (or phages). In this study, two distinct phages, VB_PaD_phPA-G (phPA-G) and VB_PaN_phPA-Intesti (phPA-Intesti), were used as single suspensions or in a phage cocktail to inactivate the planktonic cells and biofilms of P. aeruginosa. Preliminary experiments in culture medium showed that phage phPA-Intesti (reductions of 4.5-4.9 log CFU/mL) outperformed phPA-G (reductions of 0.6-2.6 log CFU/mL) and the phage cocktail (reduction of 4.2 log CFU/mL). Phage phPA-Intesti caused a maximum reduction of 5.5 log CFU/cm2 in the P. aeruginosa biofilm in urine after 4 h of incubation. The combination of phage phPA-Intesti and ciprofloxacin did not improve the efficacy of bacterial inactivation nor reduce the development of resistant mutants. However, the development of resistant bacteria was lower in the combined treatment with the phage and the antibiotic compared to treatment with the antibiotic alone. This phage lacks known toxins, virulence, antibiotic resistance, and integrase genes. Overall, the results suggest that the use of phage phPA-Intesti could be a potential approach to control urinary tract infections (UTIs), namely those caused by biofilm-producing and multidrug-resistant strains of P. aeruginosa.
Collapse
Affiliation(s)
- Inês Martinho
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Márcia Braz
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João Duarte
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Ana Brás
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Vanessa Oliveira
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Newton C M Gomes
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Carla Pereira
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Adelaide Almeida
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
6
|
Mirha H, Ali SH, Aamar H, Sadiq M, Tharwani ZH, Habib Z, Malikzai A. The impact of antibiotic resistance on the rampant spread of infectious diseases in Pakistan: Insights from a narrative review. Health Sci Rep 2024; 7:e2050. [PMID: 38655423 PMCID: PMC11035969 DOI: 10.1002/hsr2.2050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 01/28/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Background and Aims Antibiotic resistance (ABR) is a global public health emergency which has seen an uptick in low- to middle-income countries in recent times due to a plethora of aggravating factors and has led to a whole host of setting-specific pathogens registering high rates of resistance, causing outbreaks with graver mortality and morbidity. This review analyzes available literature to determine the causes and effects of ABR and recommend solutions to the problem in a Pakistani setting. Methods Sources for this narrative review were identified via electronic databases using keyword search methods. The information was retrieved using databases such as PubMed and Science Direct. Additionally, websites such as CDC and World Health Organization were used to attain pertinent information. All the sources were selected as per their relevance and appropriateness toward the purpose of this review. Results This review details the causes by dividing them into three primary strata, namely (1) under-regulation, (2) over-prescription and self-medication, and (3) lack of medical stewardship. This is made much graver when the COVID-19 pandemic and the subsequent erratic treatment response is considered, with the pandemic augmenting already high levels of consumption. These factors have led a cascade of effects including, but not limited to, a considerable increase in ABR in pathogens to first-line drugs. Conclusion ABR is a serious and growing issue which will result in undesirable personal, local, and national consequences if unchecked. Mitigation and reversal of this trend is necessary by developing existing programs and investing in novel therapies and pharmaceutical research and strengthening regulatory policies and mechanisms.
Collapse
Affiliation(s)
- Hania‐Tul Mirha
- CMH Lahore Medical CollegeNational University of Medical SciencesLahorePakistan
| | - Syed H. Ali
- Dow Medical College, Faculty of MedicineDow University of Health SciencesKarachiPakistan
| | - Humna Aamar
- Faculty of Medicine, Sindh Medical CollegeJinnah Sindh Medical UniversityKarachiPakistan
| | - Mahnoor Sadiq
- Dow Medical College, Faculty of MedicineDow University of Health SciencesKarachiPakistan
| | - Zoaib H. Tharwani
- Dow Medical College, Faculty of MedicineDow University of Health SciencesKarachiPakistan
| | | | | |
Collapse
|
7
|
Duarte J, Máximo C, Costa P, Oliveira V, Gomes NCM, Romalde JL, Pereira C, Almeida A. Potential of an Isolated Bacteriophage to Inactivate Klebsiella pneumoniae: Preliminary Studies to Control Urinary Tract Infections. Antibiotics (Basel) 2024; 13:195. [PMID: 38391581 PMCID: PMC10885952 DOI: 10.3390/antibiotics13020195] [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: 12/31/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024] Open
Abstract
Urinary tract infections (UTIs) caused by resistant Klebsiella pneumoniae can lead to severe clinical complications and even death. An alternative treatment option for infected patients is using bacteriophages. In the present study, we isolated phage VB_KPM_KP1LMA (KP1LMA) from sewage water using a K. pneumoniae strain as a host. Whole-genome analysis indicated that the genome was a double-stranded linear 176,096-bp long DNA molecule with 41.8% GC content and did not contain virulence or antibiotic resistance genes. The inactivation potential of phage KP1LMA was assessed in broth at an MOI of 1 and 10, and a maximum inactivation of 4.9 and 5.4 log CFU/mL, respectively, was observed after 9 h. The efficacy at an MOI of 10 was also assessed in urine to evaluate the phage's performance in an acidic environment. A maximum inactivation of 3.8 log CFU/mL was observed after 9 h. The results suggest that phage KP1LMA could potentially control a UTI caused by this strain of K. pneumoniae, indicating that the same procedure can be used to control UTIs caused by other strains if new specific phages are isolated. Although phage KP1LMA has a narrow host range, in the future, efforts can be made to expand its spectrum of activity and also to combine this phage with others, potentially enabling its use against other K. pneumoniae strains involved in UTIs.
Collapse
Affiliation(s)
- João Duarte
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Carolina Máximo
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Pedro Costa
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Vanessa Oliveira
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Newton C M Gomes
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Jesús L Romalde
- Department of Microbiology and Parasitology, CRETUS & CIBUS, Faculty of Biology, University of Santiago de Compostela, CP 15782 Santiago de Compostela, Spain
| | - Carla Pereira
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Adelaide Almeida
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
8
|
Anyaegbunam ZKG, Mba IE, Doowuese Y, Anyaegbunam NJ, Mba T, Aina FA, Chigor VN, Nweze EI, Eze EA. Antimicrobial resistance containment in Africa: Moving beyond surveillance. BIOSAFETY AND HEALTH 2024; 6:50-58. [DOI: 10.1016/j.bsheal.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024] Open
|
9
|
Carratalá JV, Arís A, Garcia-Fruitós E, Ferrer-Miralles N. Design strategies for positively charged endolysins: Insights into Artilysin development. Biotechnol Adv 2023; 69:108250. [PMID: 37678419 DOI: 10.1016/j.biotechadv.2023.108250] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Endolysins are bacteriophage-encoded enzymes that can specifically degrade the peptidoglycan layer of bacterial cell wall, making them an attractive tool for the development of novel antibacterial agents. The use of genetic engineering techniques for the production and modification of endolysins offers the opportunity to customize their properties and activity against specific bacterial targets, paving the way for the development of personalized therapies for bacterial infections. Gram-negative bacteria possess an outer membrane that can hinder the action of recombinantly produced endolysins. However, certain endolysins are capable of crossing the outer membrane by virtue of segments that share properties resembling those of cationic peptides. These regions increase the affinity of the endolysin towards the bacterial surface and assist in the permeabilization of the membrane. In order to improve the bactericidal effectiveness of endolysins, approaches have been implemented to increase their net charge, including the development of Artilysins containing positively charged amino acids at one end. At present, there are no specific guidelines outlining the steps for implementing these modifications. There is an ongoing debate surrounding the optimal location of positive charge, the need for a linker region, and the specific amino acid composition of peptides for modifying endolysins. The aim of this study is to provide clarity on these topics by analyzing and comparing the most effective modifications found in previous literature.
Collapse
Affiliation(s)
- Jose Vicente Carratalá
- Department of Ruminant Production, Institute of Agriculture and Agrifood Research and Technology (IRTA), Caldes de Montbui, 08140 Barcelona, Spain; Institute for Biotechnology and Biomedicine, Autonomous University of Barcelona, Bellaterra, 08193 Barcelona, Spain; Department of Genetics and Microbiology, Autonomous University of Barcelona, Bellaterra, 08193 Barcelona, Spain; Bioengineering, Biomaterials and Nanomedicine Networking Biomedical Research Centre (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain.
| | - Anna Arís
- Department of Ruminant Production, Institute of Agriculture and Agrifood Research and Technology (IRTA), Caldes de Montbui, 08140 Barcelona, Spain
| | - Elena Garcia-Fruitós
- Department of Ruminant Production, Institute of Agriculture and Agrifood Research and Technology (IRTA), Caldes de Montbui, 08140 Barcelona, Spain
| | - Neus Ferrer-Miralles
- Institute for Biotechnology and Biomedicine, Autonomous University of Barcelona, Bellaterra, 08193 Barcelona, Spain; Department of Genetics and Microbiology, Autonomous University of Barcelona, Bellaterra, 08193 Barcelona, Spain; Bioengineering, Biomaterials and Nanomedicine Networking Biomedical Research Centre (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
| |
Collapse
|
10
|
Silva EC, Rodrigues LMR, Vila MMDC, Balcão VM. Newly isolated phages preying on Pseudomonas syringae pv. garcae: In vitro and ex vivo inactivation studies in coffee plant leafs. Enzyme Microb Technol 2023; 171:110325. [PMID: 37716050 DOI: 10.1016/j.enzmictec.2023.110325] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/18/2023] [Accepted: 09/10/2023] [Indexed: 09/18/2023]
Abstract
Coffee canker, or bacterial halo blight (BHB) of coffee, is a disease caused by the phytopathogenic bacterium Pseudomonas syringae pv. garcae (Psg), having been found for the first time in 1955, in the Garça region (State of São Paulo), and which has stood out in the Brazilian coffee plantations in recent years, leading to severe economic losses that seriously affect coffee trade. The treatments available are still scarce, involving frequent spraying of coffee plantations with either copper derivatives or the antibiotic kasugamycin. However, these compounds should be avoided due to environmental toxicity and the development of bacterial resistances. Herein we report the isolation and physical/biological characterisation of two novel lytic phages and their efficacy in the control of Psg. Phages ph002F and ph004F were isolated from coffee plant leaves in Brazil (Sorocaba/SP and Itu/SP cities), using Psg IBSBF-158 as the host. According to the transmission electron microscopy analyses, both phages belong to the class Caudoviricetes and present myovirus-like morphotypes. Phages ph002F and ph004F showed eclipse times of 5 min and 20 min, respectively, and a burst size of 123 PFU/host cell and 12 PFU/host cell, respectively, allowing to conclude they replicate well in Psg IBSBF-158 with latency periods of 50 min. Phage ph002F (reduction of 4.59 log CFU/mL, compared to uninfected culture) was more effective in inactivating Psg than phage ph004F (reduction of 3.85 log CFU/mL) after 10 h of incubation at a MOI of 10. As a cocktail, the two phages were highly effective in reducing the bacterial load (reduction of 5.26 log CFU/mL at a MOI of 0.1 or reduction of 5.03 log CFU/mL at a MOI of 10, relative to untreated culture), after 12 h of treatment. This study provides evidence that the isolated phages are promising candidates against the causative agent of BHB in coffee plants.
Collapse
Affiliation(s)
- Erica C Silva
- VBlab - Laboratory of Bacterial Viruses, University of Sorocaba, 18023-000 Sorocaba, SP, Brazil
| | - Lucas M R Rodrigues
- VBlab - Laboratory of Bacterial Viruses, University of Sorocaba, 18023-000 Sorocaba, SP, Brazil; Agronomic Institute of Campinas (IAC), Centro de café Alcides Carvalho, Campinas, SP, Brazil
| | - Marta M D C Vila
- VBlab - Laboratory of Bacterial Viruses, University of Sorocaba, 18023-000 Sorocaba, SP, Brazil
| | - Victor M Balcão
- VBlab - Laboratory of Bacterial Viruses, University of Sorocaba, 18023-000 Sorocaba, SP, Brazil; Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, P-3810-193 Aveiro, Portugal.
| |
Collapse
|
11
|
Gundersen MS, Fiedler AW, Bakke I, Vadstein O. The impact of phage treatment on bacterial community structure is minor compared to antibiotics. Sci Rep 2023; 13:21032. [PMID: 38030754 PMCID: PMC10687242 DOI: 10.1038/s41598-023-48434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/27/2023] [Indexed: 12/01/2023] Open
Abstract
Phage treatment is suggested as an alternative to antibiotics; however, there is limited knowledge of how phage treatment impacts resident bacterial community structure. When phages induce bacterial lysis, resources become available to the resident community. Therefore, the density of the target bacterium is essential to consider when investigating the effect of phage treatment. This has never been studied. Thus, we invaded microcosms containing a lake-derived community with Flavobacterium columnare strain Fc7 at no, low or high densities, and treated them with either the bacteriophage FCL-2, the antibiotic Penicillin or kept them untreated (3 × 3 factorial design). The communities were sampled over the course of one week, and bacterial community composition and density were examined by 16S rDNA amplicon sequencing and flow cytometry. We show that phage treatment had minor impacts on the resident community when the host F. columnare Fc7 of the phage was present, as it caused no significant differences in bacterial density α- and β-diversity, successional patterns, and community assembly. However, a significant change was observed in community composition when the phage host was absent, mainly driven by a substantial increase in Aquirufa. In contrast, antibiotics induced significant changes in all community characteristics investigated. The most crucial finding was a bloom of γ-proteobacteria and a shift from selection to ecological drift dominating community assembly. This study investigated whether the amount of a bacterial host impacted the effect of phage treatment on community structure. We conclude that phage treatment did not significantly affect the diversity or composition of the bacterial communities when the phage host was present, but introduced changes when the host was absent. In contrast, antibiotic treatment was highly disturbing to community structure. Moreover, higher amounts of the bacterial host of the phage increased the contribution of stochastic community assembly and resulted in a feast-famine like response in bacterial density in all treatment groups. This finding emphasises that the invader density used in bacterial invasion studies impacts the experimental reproducibility. Overall, this study supports that phage treatment is substantially less disturbing to bacterial communities than antibiotic treatments.
Collapse
Affiliation(s)
- Madeleine S Gundersen
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Alexander W Fiedler
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ingrid Bakke
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Olav Vadstein
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
12
|
Islam MS, Fan J, Pan F. The power of phages: revolutionizing cancer treatment. Front Oncol 2023; 13:1290296. [PMID: 38033486 PMCID: PMC10684691 DOI: 10.3389/fonc.2023.1290296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Cancer is a devastating disease with a high global mortality rate and is projected to increase further in the coming years. Current treatment options, such as chemotherapy and radiation therapy, have limitations including side effects, variable effectiveness, high costs, and limited availability. There is a growing need for alternative treatments that can target cancer cells specifically with fewer side effects. Phages, that infect bacteria but not eukaryotic cells, have emerged as promising cancer therapeutics due to their unique properties, including specificity and ease of genetic modification. Engineered phages can transform cancer treatment by targeting cancer cells while sparing healthy ones. Phages exhibit versatility as nanocarriers, capable of delivering therapeutic agents like gene therapy, immunotherapy, and vaccines. Phages are extensively used in vaccine development, with filamentous, tailed, and icosahedral phages explored for different antigen expression possibilities. Engineered filamentous phages bring benefits such as built in adjuvant properties, cost-effectiveness, versatility in multivalent formulations, feasibility of oral administration, and stability. Phage-based vaccines stimulate the innate immune system by engaging pattern recognition receptors on antigen-presenting cells, enhancing phage peptide antigen presentation to B-cells and T-cells. This review presents recent phage therapy advances and challenges in cancer therapy, exploring its versatile tools and vaccine potential.
Collapse
Affiliation(s)
- Md. Sharifull Islam
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jie Fan
- Department of Cardiology, Handan Central Hospital, Handan, Hebei, China
| | - Fan Pan
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
13
|
Graczyk S, Pasławski R, Grzeczka A, Pasławska U, Świeczko-Żurek B, Malisz K, Popat K, Sionkowska A, Golińska P, Rai M. Antimicrobial and Antiproliferative Coatings for Stents in Veterinary Medicine-State of the Art and Perspectives. MATERIALS (BASEL, SWITZERLAND) 2023; 16:6834. [PMID: 37959431 PMCID: PMC10649059 DOI: 10.3390/ma16216834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/15/2023]
Abstract
Microbial colonization in veterinary stents poses a significant and concerning issue in veterinary medicine. Over time, these pathogens, particularly bacteria, can colonize the stent surfaces, leading to various complications. Two weeks following the stent insertion procedure, the colonization becomes observable, with the aggressiveness of bacterial growth directly correlating with the duration of stent placement. Such microbial colonization can result in infections and inflammations, compromising the stent's efficacy and, subsequently, the animal patient's overall well-being. Managing and mitigating the impact of these pathogens on veterinary stents is a crucial challenge that veterinarians and researchers are actively addressing to ensure the successful treatment and recovery of their animal patients. In addition, irritation of the tissue in the form of an inserted stent can lead to overgrowth of granulation tissue, leading to the closure of the stent lumen, as is most often the case in the trachea. Such serious complications after stent placement require improvements in the procedures used to date. In this review, antibacterial or antibiofilm strategies for several stents used in veterinary medicine have been discussed based on the current literature and the perspectives have been drawn. Various coating strategies such as coating with hydrogel, antibiotic, or other antimicrobial agents have been reviewed.
Collapse
Affiliation(s)
- Szymon Graczyk
- Institute of Veterinary Medicine, Department of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Torun, Poland; (R.P.); (A.G.); (U.P.)
| | - Robert Pasławski
- Institute of Veterinary Medicine, Department of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Torun, Poland; (R.P.); (A.G.); (U.P.)
| | - Arkadiusz Grzeczka
- Institute of Veterinary Medicine, Department of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Torun, Poland; (R.P.); (A.G.); (U.P.)
| | - Urszula Pasławska
- Institute of Veterinary Medicine, Department of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Torun, Poland; (R.P.); (A.G.); (U.P.)
| | - Beata Świeczko-Żurek
- Department of Biomaterials Technology, Faculty of Mechanical Engineering and Ship Technology, Gdansk University of Technology, Gabriela Narutowicza 11/12, 80-229 Gdansk, Poland; (B.Ś.-Ż.); (K.M.)
| | - Klaudia Malisz
- Department of Biomaterials Technology, Faculty of Mechanical Engineering and Ship Technology, Gdansk University of Technology, Gabriela Narutowicza 11/12, 80-229 Gdansk, Poland; (B.Ś.-Ż.); (K.M.)
| | - Ketul Popat
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Alina Sionkowska
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland
| | - Patrycja Golińska
- Department of Microbiology, Nicolaus Copernicus University, ul. Lwowska 1, 87-100 Torun, Poland;
| | - Mahendra Rai
- Department of Chemistry, Federal University of Piaui (UFPI), Teresina 64049-550, Brazil;
| |
Collapse
|
14
|
Huang W, Chen W, Chen Y, Fang S, Huang T, Chang P, Chang Y. Salmonella YqiC exerts its function through an oligomeric state. Protein Sci 2023; 32:e4749. [PMID: 37555831 PMCID: PMC10503411 DOI: 10.1002/pro.4749] [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: 04/27/2023] [Revised: 07/18/2023] [Accepted: 08/07/2023] [Indexed: 08/10/2023]
Abstract
Protein oligomerization occurs frequently both in vitro and in vivo, with specific functionalities associated with different oligomeric states. The YqiC protein from Salmonella Typhimurium forms a homotrimer through its C-terminal coiled-coil domain, and the protein is closely linked to the colonization and invasion of the bacteria to the host cells. To elucidate the importance of the oligomeric state of YqiC in vivo and its relation with bacterial infection, we mutated crucial residues in YqiC's coiled-coil region and confirmed the loss of trimer formation using chemical crosslinking and size exclusion chromatography coupled with multiple angle light scattering (SEC-MALS) techniques. The yqiC-knockout strain complemented with mutant YqiC showed significantly reduced colonization and invasion of Salmonella to host cells, demonstrating the critical role of YqiC oligomerization in bacterial pathogenesis. Furthermore, we conducted a protein-protein interaction study of YqiC using a pulled-down assay coupled with mass spectrometry analysis to investigate the protein's role in bacterial virulence. The results reveal that YqiC interacts with subunits of Complex II of the electron transport chain (SdhA and SdhB) and the β-subunit of F0 F1 -ATP synthase. These interactions suggest that YqiC may modulate the energy production of Salmonella and subsequently affect the assembly of crucial virulence factors, such as flagella. Overall, our findings provide new insights into the molecular mechanisms of YqiC's role in S. Typhimurium pathogenesis and suggest potential therapeutic targets for bacterial infections.
Collapse
Affiliation(s)
- Wei‐Chun Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Wai‐Ting Chen
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Yueh‐Chen Chen
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Shiuh‐Bin Fang
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho HospitalTaipei Medical UniversityTaipeiTaiwan
- Department of Pediatrics, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Master Program for Clinical Genomics and Proteomics, College of PharmacyTaipei Medical UniversityTaipeiTaiwan
| | - Tzu‐Wen Huang
- Department of Microbiology and Immunology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Pei‐Ru Chang
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho HospitalTaipei Medical UniversityTaipeiTaiwan
- Department of Pediatrics, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Yu‐Chu Chang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- International PhD Program in Cell Therapy and Regenerative Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
15
|
Asadi M, Taheri-Anganeh M, Ranjbar M, Khatami SH, Maleksabet A, Mostafavi-Pour Z, Ghasemi Y, Keshavarzi A, Savardashtaki A. LYZ2-SH3b as a novel and efficient enzybiotic against methicillin-resistant Staphylococcus aureus. BMC Microbiol 2023; 23:257. [PMID: 37704938 PMCID: PMC10500863 DOI: 10.1186/s12866-023-03002-9] [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: 01/12/2023] [Accepted: 09/04/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Enzybiotics are promising alternatives to conventional antibiotics for drug-resistant infections. Exolysins, as a class of enzybiotics, show antibacterial effects against methicillin-resistant Staphylococcus aureus (MRSA). This study evaluated a novel exolysin containing an SH3b domain for its antibacterial activity against MRSA. METHODS This study designed a chimeric exolysin by fusing the Cell-binding domain (SH3b) from Lysostaphin with the lytic domain (LYZ2) from the gp61 enzyme. Subsequently, LYZ2-SH3b was cloned and expressed in Escherichia coli (E. coli). Finally, the antibacterial effects of LYZ2-SH3b compared with LYZ2 and vancomycin against reference and clinical isolates of MRSA were measured using the disc diffusion method, the minimal inhibitory concentration (MIC), and the minimal bactericidal concentration (MBC) assays. RESULTS Analysis of bioinformatics showed that LYZ2-SH3b was stable, soluble, and non-allergenic. Protein purification was performed with a 0.8 mg/ml yield for LYZ2-SH3b. The plate lysis assay results indicated that, at the same concentrations, LYZ2-SH3b has a more inhibitory effect than LYZ2. The MICs of LYZ2 were 4 µg/mL (ATCC 43,300) and 8 µg/mL (clinical isolate ST239), whereas, for LYZ2-SH3b, they were 2 µg/mL (ATCC 43,300) and 4 µg/mL (clinical isolate ST239). This suggests a higher efficiency of LYZ2-SH3b compared to LYZ2. Furthermore, the MBCs of LYZ2 were 4 µg/mL (ATCC 43,300) and 8 µg/mL (clinical isolate ST239), whereas, for LYZ2-SH3b, they were 2 µg/mL (ATCC 43,300) and 4 µg/mL (clinical isolate ST239), thus confirming the superior lytic activity of LYZ2-SH3b over LYZ2. CONCLUSIONS The study suggests that phage endolysins, such as LYZ2-SH3b, may represent a promising new approach to treating MRSA infections, particularly in cases where antibiotic resistance is a concern. But further studies are needed.
Collapse
Affiliation(s)
- Marzieh Asadi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences Shiraz, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Ranjbar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences Shiraz, Shiraz, Iran
| | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Maleksabet
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zohreh Mostafavi-Pour
- Recombinant Protein Laboratory, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | | | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences Shiraz, Shiraz, Iran.
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
16
|
Pereira AO, Barros NMA, Guerrero BR, Emencheta SC, Baldo DÂ, Oliveira JM, Vila MMDC, Balcão VM. An Edible Biopolymeric Microcapsular Wrapping Integrating Lytic Bacteriophage Particles for Salmonella enterica: Potential for Integration into Poultry Feed. Antibiotics (Basel) 2023; 12:988. [PMID: 37370307 DOI: 10.3390/antibiotics12060988] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
This research work aimed at developing an edible biopolymeric microcapsular wrapping (EBMW) integrating lytic bacteriophage particles for Salmonella enterica, with potential application in poultry feed for biocontrol of that pathogen. This pathogen is known as one of the main microorganisms responsible for contamination in the food industry and in foodstuff. The current techniques for decontamination and pathogen control in the food industry can be very expensive, not very selective, and even outdated, such as the use of broad-spectrum antibiotics that end up selecting resistant bacteria. Hence, there is a need for new technologies for pathogen biocontrol. In this context, bacteriophage-based biocontrol appears as a potential alternative. As a cocktail, both phages were able to significantly reduce the bacterial load after 12 h of treatment, at either multiplicity of infection (MOI) 1 and 10, by 84.3% and 87.6%, respectively. Entrapment of the phage virions within the EBMW matrix did not exert any deleterious effect upon their lytic activity. The results obtained showed high promise for integration in poultry feed aiming at controlling Salmonella enterica, since the edible biopolymeric microcapsular wrapping integrating lytic bacteriophage particles developed was successful in maintaining lytic phage viability while fully stabilizing the phage particles.
Collapse
Affiliation(s)
- Arthur O Pereira
- PhageLab-Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, SP, Brazil
| | - Nicole M A Barros
- PhageLab-Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, SP, Brazil
| | - Bruna R Guerrero
- PhageLab-Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, SP, Brazil
| | - Stephen C Emencheta
- PhageLab-Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, SP, Brazil
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Enugu, Nigeria
| | - Denicezar  Baldo
- LaFiNAU-Laboratory of Applied Nuclear Physics, University of Sorocaba, Sorocaba 18023-000, SP, Brazil
| | - José M Oliveira
- LaFiNAU-Laboratory of Applied Nuclear Physics, University of Sorocaba, Sorocaba 18023-000, SP, Brazil
| | - Marta M D C Vila
- PhageLab-Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, SP, Brazil
| | - Victor M Balcão
- PhageLab-Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, SP, Brazil
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, P-3810-193 Aveiro, Portugal
| |
Collapse
|
17
|
Zhang H, Zhang X, Liang S, Wang J, Zhu Y, Zhang W, Liu S, Schwarz S, Xie F. Bactericidal synergism between phage endolysin Ply2660 and cathelicidin LL-37 against vancomycin-resistant Enterococcus faecalis biofilms. NPJ Biofilms Microbiomes 2023; 9:16. [PMID: 37024490 PMCID: PMC10078070 DOI: 10.1038/s41522-023-00385-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 03/23/2023] [Indexed: 04/08/2023] Open
Abstract
Antibiotic resistance and the ability to form biofilms of Enterococcus faecalis have compromised the choice of therapeutic options, which triggered the search for new therapeutic strategies, such as the use of phage endolysins and antimicrobial peptides. However, few studies have addressed the synergistic relationship between these two promising options. Here, we investigated the combination of the phage endolysin Ply2660 and the antimicrobial peptide LL-37 to target drug-resistant biofilm-producing E. faecalis. In vitro bactericidal assays were used to demonstrate the efficacy of the Ply2660-LL-37 combination against E. faecalis. Larger reductions in viable cell counts were observed when Ply2660 and LL-37 were applied together than after individual treatment with either substance. Transmission electron microscopy revealed that the Ply2660-LL-37 combination could lead to severe cell lysis of E. faecalis. The mode of action of the Ply2660-LL-37 combination against E. faecalis was that Ply2660 degrades cell wall peptidoglycan, and subsequently, LL-37 destroys the cytoplasmic membrane. Furthermore, Ply2660 and LL-37 act synergistically to inhibit the biofilm formation of E. faecalis. The Ply2660-LL-37 combination also showed a synergistic effect for the treatment of established biofilm, as biofilm killing with this combination was superior to each substance alone. In a murine peritoneal septicemia model, the Ply2660-LL-37 combination distinctly suppressed the dissemination of E. faecalis isolates and attenuated organ injury, being more effective than each treatment alone. Altogether, our findings indicate that the combination of a phage endolysin and an antimicrobial peptide may be a potential antimicrobial strategy for combating E. faecalis.
Collapse
Affiliation(s)
- Huihui Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xinyuan Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Siyu Liang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jing Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yao Zhu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Wanjiang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Siguo Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Stefan Schwarz
- Institute of Microbiology and Epizootics, Centre for Infection Medicine, School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, 14163, Berlin, Germany.
| | - Fang Xie
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| |
Collapse
|
18
|
Choudhary P, Kraatz HB, Lévesque CM, Gong SG. Microencapsulation of Probiotic Streptococcus salivarius LAB813. ACS OMEGA 2023; 8:12011-12018. [PMID: 37033842 PMCID: PMC10077535 DOI: 10.1021/acsomega.2c07721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 03/13/2023] [Indexed: 06/19/2023]
Abstract
Probiotics are living microorganisms that confer a health benefit on the host when administered in adequate amounts. Streptococcus salivarius, a commensal bacterium found in the oral cavity, has been shown to secrete antimicrobial peptides and can be used as probiotics. This study aimed to develop a delivery system for the probiotic LAB813, a novel S. salivarius strain first identified in the laboratory. Probiotics can be delivered and protected through the encapsulation of biomaterials such as polysaccharides. Their biocompatibility, biodegradability, user-friendliness, and ease of access make polysaccharides useful for encapsulating probiotics. Alginate (Alg) and chitosan (Ch) are naturally obtained polysaccharides and, hence, tested for LAB813 encapsulation. An extrusion method of encapsulation was performed to form Alg microcapsules (Alg-LAB813), some of which were coated with Ch (Alg-LAB813-Ch) to provide dual-layered protection. Inhibitory assays of the Alg-LAB813 and Alg-LAB813-Ch microcapsules were assayed against an indicator strain. Alg-LAB813-Ch microcapsules showed superior antibacterial properties compared to Alg-LAB813 microcapsules over 24 h and when subject to temperatures ranging from 4 to 68 °C. In addition, Alg-LAB813-Ch microcapsules retained antibacterial activity for up to 28 days of storage at 4 °C. The strong and sustained inhibitory activities of Ch-coated Alg encapsulated LAB813 signify the potential for their use to improve oral health.
Collapse
Affiliation(s)
| | - Heinz-Bernhard Kraatz
- Department
of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada
| | - Céline M. Lévesque
- Faculty
of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
| | - Siew-Ging Gong
- Faculty
of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
| |
Collapse
|
19
|
Islam MS, Rahman MT. A Comprehensive Review on Bacterial Vaccines Combating Antimicrobial Resistance in Poultry. Vaccines (Basel) 2023; 11:vaccines11030616. [PMID: 36992200 DOI: 10.3390/vaccines11030616] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Bacterial vaccines have become a crucial tool in combating antimicrobial resistance (AMR) in poultry. The overuse and misuse of antibiotics in poultry farming have led to the development of AMR, which is a growing public health concern. Bacterial vaccines are alternative methods for controlling bacterial diseases in poultry, reducing the need for antibiotics and improving animal welfare. These vaccines come in different forms including live attenuated, killed, and recombinant vaccines, and they work by stimulating the immune system to produce a specific response to the target bacteria. There are many advantages to using bacterial vaccines in poultry, including reduced use of antibiotics, improved animal welfare, and increased profitability. However, there are also limitations such as vaccine efficacy and availability. The use of bacterial vaccines in poultry is regulated by various governmental bodies and there are economic considerations to be taken into account, including costs and return on investment. The future prospects for bacterial vaccines in poultry are promising, with advancements in genetic engineering and vaccine formulation, and they have the potential to improve the sustainability of the poultry industry. In conclusion, bacterial vaccines are essential in combating AMR in poultry and represent a crucial step towards a more sustainable and responsible approach to poultry farming.
Collapse
Affiliation(s)
- Md Saiful Islam
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Md Tanvir Rahman
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| |
Collapse
|
20
|
Cagnetta GE, Martínez SR, Ibarra LE, Gallastegui A, Martucci JF, Palacios RE, Chesta CA, Gómez ML. Reusable antimicrobial antibiotic-free dressings obtained by photopolymerization. BIOMATERIALS ADVANCES 2023; 149:213399. [PMID: 37011423 DOI: 10.1016/j.bioadv.2023.213399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023]
Abstract
In recent years significant efforts have been made to develop new materials for wound dressing with improved healing properties. However, the synthesis methods usually employed to this end are often complex or require several steps. We describe here the synthesis and characterization of antimicrobial reusable dermatological wound dressings based on N-isopropylacrylamide co-polymerized with [2-(Methacryloyloxy) ethyl] trimethylammonium chloride hydrogels (NIPAM-co-METAC). The dressings were obtained with a very efficient single-step synthesis procedure based on visible light (455 nm) by photopolymerization. To this end, F8BT nanoparticles of the conjugated polymer (poly(9,9-dioctylfluorene-alt-benzothiadiazole) - F8BT) were used as macro-photoinitiators, and a modified silsesquioxane was employed as crosslinker. Dressings obtained by this simple and gentle method show antimicrobial and wound healing properties, without the incorporation of antibiotics or any other additives. The physical and mechanical properties of these hydrogel-based dressings were evaluated, as well as their microbiological properties, through in vitro experiments. Results show that dressings with a molar ratio of METAC of 0.5 or higher exhibit high swelling capacity, appropriate water vapor transmission rate values, stability and thermal response, high ductility and adhesiveness. In addition, biological tests showed that the dressings have significant antimicrobial capacity. The best inactivation performance was found for hydrogels synthesized with the highest METAC content. The dressings were tested several times with fresh bacterial cultures, showing a bacterial kill efficiency of 99.99 % even after three repetitions in a row, employing the same dressing, demonstrating the intrinsic bactericidal property of the materials and their reusability. In addition, the gels show low hemolytic effect, high dermal biocompatibility and noticeable wound healing effects. Overall results demonstrate that some specific hydrogel formulations have potential application as dermatological dressings for wound healing and disinfection.
Collapse
Affiliation(s)
- Gonzalo E Cagnetta
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Campus Universitario, 5800 Río Cuarto, Argentina
| | - Sol R Martínez
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Campus Universitario, 5800 Río Cuarto, Argentina
| | - Luis E Ibarra
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Campus Universitario, 5800 Río Cuarto, Argentina
| | - Antonela Gallastegui
- POLYMAT, University of the Basque Country UPV/EHU, Avenida Tolosa 72, Donostia-San Sebastian 20018, Gipuzkoa, Spain
| | - Josefa F Martucci
- Instituto de Investigaciones en Ciencias y Tecnología de los Materiales (INTEMA), Universidad Nacional de Mar del Plata, Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Av. Colón 10850, 7600 Mar del Plata, Argentina
| | - Rodrigo E Palacios
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Campus Universitario, 5800 Río Cuarto, Argentina
| | - Carlos A Chesta
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Campus Universitario, 5800 Río Cuarto, Argentina
| | - María L Gómez
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Campus Universitario, 5800 Río Cuarto, Argentina.
| |
Collapse
|
21
|
Khambhati K, Bhattacharjee G, Gohil N, Dhanoa GK, Sagona AP, Mani I, Bui NL, Chu D, Karapurkar JK, Jang SH, Chung HY, Maurya R, Alzahrani KJ, Ramakrishna S, Singh V. Phage engineering and phage-assisted CRISPR-Cas delivery to combat multidrug-resistant pathogens. Bioeng Transl Med 2023; 8:e10381. [PMID: 36925687 PMCID: PMC10013820 DOI: 10.1002/btm2.10381] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/30/2022] [Accepted: 07/16/2022] [Indexed: 12/13/2022] Open
Abstract
Antibiotic resistance ranks among the top threats to humanity. Due to the frequent use of antibiotics, society is facing a high prevalence of multidrug resistant pathogens, which have managed to evolve mechanisms that help them evade the last line of therapeutics. An alternative to antibiotics could involve the use of bacteriophages (phages), which are the natural predators of bacterial cells. In earlier times, phages were implemented as therapeutic agents for a century but were mainly replaced with antibiotics, and considering the menace of antimicrobial resistance, it might again become of interest due to the increasing threat of antibiotic resistance among pathogens. The current understanding of phage biology and clustered regularly interspaced short palindromic repeats (CRISPR) assisted phage genome engineering techniques have facilitated to generate phage variants with unique therapeutic values. In this review, we briefly explain strategies to engineer bacteriophages. Next, we highlight the literature supporting CRISPR-Cas9-assisted phage engineering for effective and more specific targeting of bacterial pathogens. Lastly, we discuss techniques that either help to increase the fitness, specificity, or lytic ability of bacteriophages to control an infection.
Collapse
Affiliation(s)
- Khushal Khambhati
- Department of Biosciences, School of ScienceIndrashil UniversityRajpurMehsanaGujaratIndia
| | - Gargi Bhattacharjee
- Department of Biosciences, School of ScienceIndrashil UniversityRajpurMehsanaGujaratIndia
| | - Nisarg Gohil
- Department of Biosciences, School of ScienceIndrashil UniversityRajpurMehsanaGujaratIndia
| | - Gurneet K. Dhanoa
- School of Life SciencesUniversity of Warwick, Gibbet Hill CampusCoventryUnited Kindgom
| | - Antonia P. Sagona
- School of Life SciencesUniversity of Warwick, Gibbet Hill CampusCoventryUnited Kindgom
| | - Indra Mani
- Department of MicrobiologyGargi College, University of DelhiNew DelhiIndia
| | - Nhat Le Bui
- Center for Biomedicine and Community HealthInternational School, Vietnam National UniversityHanoiVietnam
| | - Dinh‐Toi Chu
- Center for Biomedicine and Community HealthInternational School, Vietnam National UniversityHanoiVietnam
- Faculty of Applied SciencesInternational School, Vietnam National UniversityHanoiVietnam
| | | | - Su Hwa Jang
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoulSouth Korea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulSouth Korea
| | - Hee Yong Chung
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoulSouth Korea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulSouth Korea
- College of MedicineHanyang UniversitySeoulSouth Korea
| | - Rupesh Maurya
- Department of Biosciences, School of ScienceIndrashil UniversityRajpurMehsanaGujaratIndia
| | - Khalid J. Alzahrani
- Department of Clinical Laboratories SciencesCollege of Applied Medical Sciences, Taif UniversityTaifSaudi Arabia
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoulSouth Korea
- College of MedicineHanyang UniversitySeoulSouth Korea
| | - Vijai Singh
- Department of Biosciences, School of ScienceIndrashil UniversityRajpurMehsanaGujaratIndia
| |
Collapse
|
22
|
Liu P, Dong X, Cao X, Xie Q, Huang X, Jiang J, Dai H, Tang Z, Lin Y, Feng S, Luo K. Identification of Three Campylobacter Lysins and Enhancement of Their Anti-Escherichia coli Efficacy Using Colicin-Based Translocation and Receptor-Binding Domain Fusion. Microbiol Spectr 2023; 11:e0451522. [PMID: 36749047 PMCID: PMC10100823 DOI: 10.1128/spectrum.04515-22] [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: 11/06/2022] [Accepted: 01/24/2023] [Indexed: 02/08/2023] Open
Abstract
The emergence of multidrug-resistant Escherichia coli, which poses a major threat to public health, has motivated the development of numerous alternative antimicrobials. Lysins are bacteriophage- and bacterium-derived peptidoglycan hydrolases that represent a new antibiotic treatment targeting bacterial cell walls. However, the bactericidal effect of native lysins on Gram-negative bacteria is restricted by the presence of an outer membrane. Here, we first evaluated the antibacterial activity of three Campylobacter-derived lysins (Clysins) against E. coli. To improve their transmembrane ability and antibacterial activities, six engineered Clysins were constructed by fusing with the translocation and receptor-binding (TRB) domains from two types of colicins (colicin A [TRBA] and colicin K [TRBK]), and their biological activities were determined. Notably, engineered lysin TRBK-Cly02 exhibited the highest bactericidal activity against the E. coli BL21 strain, with a reduction of 6.22 ± 0.34 log units of cells at a concentration of 60.1 μg/mL, and formed an observable inhibition zone even at a dose of 6.01 μg. Moreover, TRBK-Cly02 killed E. coli dose dependently and exhibited the strongest bactericidal activity at pH 6. It also exhibited potential bioactivity against multidrug-resistant E. coli clinical isolates. In summary, this study identified three lysins from Campylobacter strains against E. coli, and the enhancement of their antibacterial activities by TRB domains fusion may allow them to be developed as potential alternatives to antibiotics. IMPORTANCE Three lysins from Campylobacter, namely, Clysins, were investigated, and their antibacterial activities against E. coli were determined for the first time. To overcome the restriction of the outer membrane of Gram-negative bacteria, we combined the TRB domains of colicins with these Clysins. Moreover, we discovered that the Clysins fused with TRB domains from colicin K (TRBK) killed E. coli more effectively, and this provides a new foundation for the development of novel bioengineered lysins by employing TRBK constructs that target outer membrane receptor/transport systems. One of the designed lysins, TRBK-Cly02, exhibited potent bactericidal efficacy against E. coli strains and may be used for control of multidrug-resistant clinical isolates. The results suggest that TRBK-Cly02 can be considered a potential antibacterial agent against pathogenic E. coli.
Collapse
Affiliation(s)
- Peiqi Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xinying Dong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xuewei Cao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qianmei Xie
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiuqin Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jinfei Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Huilin Dai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zheng Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yizhen Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Saixiang Feng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Kaijian Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
23
|
Fiorillo A, Frezza D, Di Lallo G, Visconti S. A Phage Therapy Model for the Prevention of Pseudomonas syringae pv. actinidiae Infection of Kiwifruit Plants. PLANT DISEASE 2023; 107:267-271. [PMID: 35939740 DOI: 10.1094/pdis-02-22-0348-sc] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Great efforts have been made with chemicals and pesticides to contain the spread of Pseudomonas syringae pv. actinidiae (Psa) responsible for kiwifruit canker. Unfortunately, only partial results were obtained for this bacterial pandemic, and alternative remedies were proposed to avoid soil pollution and the onset of antibiotic resistance. Among these, phage therapy represents a possible tool with low environmental impact and high specificity. Several phages have been isolated and tested for the capacity to kill Psa in vitro, but experiments to verify their efficacy in vivo are still lacking. In the present study, we demonstrated that the phage φPSA2 (previously characterized) contains the spread of Psa inside plant tissue and reduces the symptoms of the disease. Our data are a strong indication for the efficiency of this phage and open the possibility of developing a phage therapy based on φPSA2 to counteract the bacterial canker of kiwifruit.
Collapse
Affiliation(s)
- Anna Fiorillo
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Domenico Frezza
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Gustavo Di Lallo
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Sabina Visconti
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
24
|
Chatupheeraphat C, Peamchai J, Luk-in S, Eiamphungporn W. Synergistic effect and antibiofilm activity of the antimicrobial peptide K11 with conventional antibiotics against multidrug-resistant and extensively drug-resistant Klebsiella pneumoniae. Front Cell Infect Microbiol 2023; 13:1153868. [PMID: 37113135 PMCID: PMC10126264 DOI: 10.3389/fcimb.2023.1153868] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction Infections caused by drug-resistant Klebsiella pneumoniae are now a serious problem for public health, associated with high morbidity and mortality due to limited treatment options. Therefore, new antibacterial agents or a combination of agents as the first line of treatment are urgently needed. K11 is a novel antimicrobial peptide (AMP) that has demonstrated in vitro antimicrobial activity against several types of bacteria. Additionally, K11 has previously shown no hemolytic activity. Herein, the antibacterial activity, the synergistic action of K11 in combination with different conventional antibiotics and the antibiofilm activity of K11 against multidrug-resistant (MDR) and extensively drug-resistant (XDR) K. pneumoniae were investigated. Meanwhile, the stability and ability to induce the bacterial resistance of K11 were also tested. Methods Fifteen clinical isolates of MDR/XDR K. pneumoniae were used in this study. The minimum inhibitory concentration (MIC) of K11 against these isolates was determined by the broth microdilution method. In vitro synergy between K11 and antibiotics was evaluated using the checkerboard methodology. The antibiofilm activity of K11 against K. pneumoniae strong biofilm producers were explored by the crystal violet staining. The stability in different environments and resistance induction of K11 were evaluated by MIC determination. Results The MIC values of K11 against MDR/XDR K. pneumoniae isolates were 8-512 μg/mL. Intriguingly, the synergistic effects were clearly observed for K11 in combination with chloramphenicol, meropenem, rifampicin, or ceftazidime, whereas no synergy was observed when K11 was combined with colistin. Besides, K11 effectively prevented biofilm formation against K. pneumoniae strong biofilm producers in a concentration-dependent manner starting at 0.25×MIC and exerted an enhancing effect when administered in combination with meropenem, chloramphenicol, or rifampicin. Additionally, K11 demonstrated high thermal and wide pH stability along with good stability in serum and physiological salts. Significantly, K. pneumoniae showed no induction of resistance even after prolonged exposure to a sub-inhibitory concentration of K11. Conclusion These findings indicate that K11 is a promising candidate with potent antibacterial and antibiofilm activities without inducing resistance and acts synergistically with conventional antibiotics against drug-resistant K. pneumoniae.
Collapse
Affiliation(s)
- Chawalit Chatupheeraphat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Jiratchaya Peamchai
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Sirirat Luk-in
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Warawan Eiamphungporn
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
- *Correspondence: Warawan Eiamphungporn,
| |
Collapse
|
25
|
Pérez-Aranda M, Pajuelo E, Navarro-Torre S, Pérez-Palacios P, Begines B, Rodríguez-Llorente ID, Torres Y, Alcudia A. Antimicrobial and Antibiofilm Effect of 4,4'-Dihydroxy-azobenzene against Clinically Resistant Staphylococci. Antibiotics (Basel) 2022; 11:antibiotics11121800. [PMID: 36551456 PMCID: PMC9774766 DOI: 10.3390/antibiotics11121800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
The spread of antibiotic resistance among human and animal pathogens is one of the more significant public health concerns. Moreover, the restrictions on the use of particular antibiotics can limit the options for the treatment of infections in veterinary clinical practice. In this context, searching for alternative antimicrobial substances is crucial nowadays. In this study, 4,4'-dihydroxy-azobenzene (DHAB) was tested for its potential in vitro as an antimicrobial agent against two relevant human and animal pathogens, namely Staphylococcus aureus and Staphylococcus pseudintermedius. The values of minimal inhibitory concentration (MIC) were 64 and 32 mg/L respectively, and they comparable to other azo compounds of probed antimicrobial activity. In addition, the minimal bactericidal concentrations (MCB) were 256 and 64 mg/L. The mechanism by which DHAB produces toxicity in staphylococci has been investigated. DHAB caused membrane damage as revealed by the increase in thiobarbituric acid reactive substances (TBARS) such as malondialdehyde. Furthermore, differential induction of the enzymes peroxidases and superoxide dismutase in S. aureus and S. pseudintermedius suggested their prevalent role in ROS-scavenging due to the oxidative burst induced by this compound in either species. In addition, this substance was able to inhibit the formation of biofilms by both bacteria as observed by colorimetric tests and scanning electron microscopy. In order to assess the relevance of DHAB against clinical strains of MRSA, 10 clinical isolates resistant to either methicillin or daptomycin were assayed; 80% of them gave values of CMI and CMB similar to those of the control S. aureus strain. Finally, cutaneous plasters containing a composite formed by an agar base supplemented with DHAB were designed. These plasters were able to inhibit in vitro the growth of S. aureus and S. pseudintermedius, particularly the later, and this suggests that this substance could be a promising candidate as an alternative to antibiotics in the treatment of animal skin infections, as it has been proven that the toxicity of this substance is very low particularly at a dermal level.
Collapse
Affiliation(s)
- María Pérez-Aranda
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, c/Profesor García González, 2, 41012 Sevilla, Spain
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, c/Profesor García González, 2, 41012 Sevilla, Spain
| | - Eloísa Pajuelo
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, c/Profesor García González, 2, 41012 Sevilla, Spain
- Correspondence: (E.P.); (A.A.); Tel.: +34-954556924 (E.P.); +34-954556740 (A.A.)
| | - Salvadora Navarro-Torre
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, c/Profesor García González, 2, 41012 Sevilla, Spain
| | - Patricia Pérez-Palacios
- UGC Enfermedades Infecciosas, Microbiología Clínica y Medicina Preventiva, Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena, CSIC, Universidad de Sevilla, 41009 Seville, Spain
| | - Belén Begines
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, c/Profesor García González, 2, 41012 Sevilla, Spain
| | - Ignacio D. Rodríguez-Llorente
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, c/Profesor García González, 2, 41012 Sevilla, Spain
| | - Yadir Torres
- Departamento de Ingeniería y Ciencia de los Materiales y del Transporte, Escuela Politécnica Superior, Universidad de Sevilla, Virgen de África 7, 41011 Sevilla, Spain
| | - Ana Alcudia
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, c/Profesor García González, 2, 41012 Sevilla, Spain
- Correspondence: (E.P.); (A.A.); Tel.: +34-954556924 (E.P.); +34-954556740 (A.A.)
| |
Collapse
|
26
|
Yao Q, Wu C, Yu X, Chen X, Pan G, Chen B. Current material engineering strategies to prevent catheter encrustation in urinary tracts. Mater Today Bio 2022; 16:100413. [PMID: 36118951 PMCID: PMC9474921 DOI: 10.1016/j.mtbio.2022.100413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/19/2022] Open
Abstract
Catheters and ureteric stents have played a vital role in relieving urinary obstruction in many urological conditions. With the increasing use of urinary catheters/stents, catheter/stent-related complications such as infection and encrustation are also increasing because of their design defects. Long-term use of antibiotics and frequent replacement of catheters not only increase the economic burden on patients but also bring the pain of catheter replacement. This is unfavorable for patients with long indwelling catheters or stents but inconvenient to replace. In recent years, some promising technologies and mechanisms have been used to prevent infection and encrustation, mainly drug loading coatings, functional coatings, biodegradable polymers and metallic materials for urinary devices. Obvious effects in anti-encrustation and anti-infection experiments of the above strategies in vivo or in vitro have been conducted, which is very helpful for further clinical trials. This review mainly introduces catheter/stent technology and mechanisms in the past ten years to address the potential impact of anti-encrustation coating of catheter/stent materials for the prevention of encrustation and to analyze the progress made in this field.
Collapse
Affiliation(s)
- Qin Yao
- Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, Jiangsu, 212001, PR China
| | - Chengshuai Wu
- Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, Jiangsu, 212001, PR China
| | - Xiaoyu Yu
- Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, Jiangsu, 212001, PR China
| | - Xu Chen
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 304 Xuefu Road, Zhenjiang, Jiangsu, 212013, PR China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 304 Xuefu Road, Zhenjiang, Jiangsu, 212013, PR China
| | - Binghai Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, Jiangsu, 212001, PR China
| |
Collapse
|
27
|
The coordination of anti-phage immunity mechanisms in bacterial cells. Nat Commun 2022; 13:7412. [PMID: 36456580 PMCID: PMC9715693 DOI: 10.1038/s41467-022-35203-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/18/2022] [Indexed: 12/02/2022] Open
Abstract
Bacterial cells are equipped with a variety of immune strategies to fight bacteriophage infections. Such strategies include unspecific mechanisms directed against any phage infecting the cell, ranging from the identification and cleavage of the viral DNA by restriction nucleases (restriction-modification systems) to the suicidal death of infected host cells (abortive infection, Abi). In addition, CRISPR-Cas systems generate an immune memory that targets specific phages in case of reinfection. However, the timing and coordination of different antiviral systems in bacterial cells are poorly understood. Here, we use simple mathematical models of immune responses in individual bacterial cells to propose that the intracellular dynamics of phage infections are key to addressing these questions. Our models suggest that the rates of viral DNA replication and cleavage inside host cells define functional categories of phages that differ in their susceptibility to bacterial anti-phage mechanisms, which could give raise to alternative phage strategies to escape bacterial immunity. From this viewpoint, the combined action of diverse bacterial defenses would be necessary to reduce the chances of phage immune evasion. The decision of individual infected cells to undergo suicidal cell death or to incorporate new phage sequences into their immune memory would be determined by dynamic interactions between the host's immune mechanisms and the phage DNA. Our work highlights the importance of within-cell dynamics to understand bacterial immunity, and formulates hypotheses that may inspire future research in this area.
Collapse
|
28
|
Rozman U, Duh D, Cimerman M, Turk SŠ. Hygiene of Medical Devices and Minimum Inhibitory Concentrations for Alcohol-Based and QAC Disinfectants among Isolates from Physical Therapy Departments. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14690. [PMID: 36429408 PMCID: PMC9691081 DOI: 10.3390/ijerph192214690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 06/16/2023]
Abstract
Disinfectants are used intensively to control and prevent healthcare-associated infections. With continuous use and exposure to disinfectants, bacteria may develop reduced susceptibility. The study aimed to check the hygiene of devices in the physiotherapy department. For isolated bacterial strains, we aimed to determine the minimum inhibitory concentration of five different disinfectant wipe products currently in use. Microbiological environmental sampling in four various institutions in four different cities from two counties was performed, followed by CFU calculation and identification using matrix-assisted laser desorption and ionization with time-of-flight analyzer mass spectrometry (MALDI-TOF). The sampling was performed on three different occasions: before patient use, after patient use, and after disinfection. The susceptibility of isolates to three different alcohol-based and three different quaternary ammonium compounds (QAC) disinfectant wipes was examined by determining the minimal inhibitory concentrations (MIC). We identified 27 different bacterial species from 11 different genera. Gram-positive bacteria predominated. The most abundant genera were Staphylococcus, Micrococcus, and Bacillus. The average MIC values of alcohol-based disinfectants range between 66.61 and 148.82 g/L, and those of QAC-based disinfectants range between 2.4 and 3.5 mg/L. Distinctive strains with four-fold increases in MIC values, compared to average values, were identified. The widespread use of disinfectants can induce a reduction in the susceptibility of bacteria against disinfectants and affect the increase in the proportion of antibiotic-resistant bacteria. Therefore, it is urgent to define clear criteria for defining a microorganism as resistant to disinfectants by setting epidemiological cut-off (ECOFF) values and standardizing protocols for testing the resistance of microorganisms against disinfectants.
Collapse
Affiliation(s)
- Urška Rozman
- Faculty of Health Sciences, University of Maribor, Žitna ulica 15, 2000 Maribor, Slovenia
| | - Darja Duh
- Chemicals Office of the Republic of Slovenia, Ajdovščina 4, 1000 Ljubljana, Slovenia
| | - Mojca Cimerman
- National Laboratory of Health, Environment and Food, Prvomajska ulica 1, 2000 Maribor, Slovenia
| | - Sonja Šostar Turk
- Faculty of Health Sciences, University of Maribor, Žitna ulica 15, 2000 Maribor, Slovenia
| |
Collapse
|
29
|
Balcão VM, Belline BG, Silva EC, Almeida PFFB, Baldo DÂ, Amorim LRP, Oliveira Júnior JM, Vila MMDC, Del Fiol FS. Isolation and Molecular Characterization of Two Novel Lytic Bacteriophages for the Biocontrol of Escherichia coli in Uterine Infections: In Vitro and Ex Vivo Preliminary Studies in Veterinary Medicine. Pharmaceutics 2022; 14:2344. [PMID: 36365162 PMCID: PMC9692438 DOI: 10.3390/pharmaceutics14112344] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 09/18/2023] Open
Abstract
E. coli is one of the etiological agents responsible for pyometra in female dogs, with conventional treatment involving ovariohysterectomy. Here, we report the isolation and full characterization of two novel lytic phages, viz. vB_EcoM_Uniso11 (ph0011) and vB_EcoM_Uniso21 (ph0021). Both phages belong to the order Caudovirales and present myovirus-like morphotypes, with phage ph0011 being classified as Myoviridae genus Asteriusvirus and phage ph0021 being classified as Myoviridae genus Tequatrovirus, based on their complete genome sequences. The 348,288 bp phage ph0011 and 165,222 bp phage ph0021 genomes do not encode toxins, integrases or antimicrobial resistance genes neither depolymerases related sequences. Both phages were shown to be effective against at least twelve E. coli clinical isolates in in vitro antibacterial activity assays. Based on their features, both phages have potential for controlling pyometra infections caused by E. coli. Phage ph0011 (reduction of 4.24 log CFU/mL) was more effective than phage ph0021 (reduction of 1.90 log CFU/mL) after 12 h of incubation at MOI 1000. As a cocktail, the two phages were highly effective in reducing the bacterial load (reduction of 5.57 log CFU/mL) at MOI 100, after 12 h of treatment. Both phages were structurally and functionally stabilized in vaginal egg formulations.
Collapse
Affiliation(s)
- Victor M. Balcão
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
- Department of Biology and CESAM, Campus Universitário de Santiago, University of Aveiro, P-3810-193 Aveiro, Portugal
| | - Bianca G. Belline
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| | - Erica C. Silva
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| | - Pablo F. F. B. Almeida
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| | - Denicezar Â. Baldo
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| | - Lara R. P. Amorim
- Department of Education, Faculty of Sciences, University of Porto, P-4169-007 Porto, Portugal
| | - José M. Oliveira Júnior
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| | - Marta M. D. C. Vila
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| | - Fernando S. Del Fiol
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| |
Collapse
|
30
|
Co-Expression of Pig IL-2 and Fusion Bovine Cathelicidin Gene by Recombinant Plasmids in Yeast and Their Promotion of Mouse Antibacterial Defense. BIOLOGY 2022; 11:biology11101491. [PMID: 36290395 PMCID: PMC9598770 DOI: 10.3390/biology11101491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/27/2022] [Accepted: 10/06/2022] [Indexed: 11/07/2022]
Abstract
In order to develop an effective and safe immunomodulator to enhance the antimicrobial bioactivity and immunity of animals against infectious bacterial diseases, a recombinant plasmid pGAPZαA-IL2-B co-expressing pig interleukin-2 (PIL-2) and fused bovine cathelicidin (FBC) genes were constructed using the 2A self-cleavage technique. After being expressed in Pichia pastoris strain SMD1168, the recombinant yeast was administered orally to 5-week-old female ICR mice. The control mice were similarly dosed with P. pastoris with a blank plasmid or FBC recombinant plasmid alone. At 28 days post-treatment, the mice were challenged intraperitoneally with virulent strains of either E. coli or S. aureus. Compared with the control groups, the mice that received recombinant yeast co-expressing PIL-2/FBC manifested significant increases in the number of leukocytes, CD4+ and CD8+ T cells, IgG, and the gene expressions of TLRs(TLR1,4,6,9), antimicrobial peptides(CRP4 and CRAMP) and cytokines (IL-2, 4, 6, 7, 12, 15, 23, IFN-γ, and TNF-α) in the blood. Furthermore, the treated mice displayed significantly higher survival than the other two control groups after the challenge. These results suggest that the antimicrobial activity and immunity of animals can be effectively enhanced by the in vivo co-expression of IL-2 and the FBS gene, which can facilitate the development of new immunopotentiation molecules to overcome the infection of antibiotic-resistant bacteria.
Collapse
|
31
|
Saini P, Bari SS, Yadav P, Khullar S, Mandal SK, Bhalla A. Synthesis of
C2
‐Formamide(thiophene)pyrazolyl‐
C4
’‐carbaldehyde and their Transformation to Schiff's Bases and Stereoselective
trans
‐β‐Lactams: Mechanistic and Theoretical Insights. ChemistrySelect 2022. [DOI: 10.1002/slct.202202172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Preety Saini
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University Chandigarh 160014 India
| | - Shamsher S. Bari
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University Chandigarh 160014 India
| | - Pooja Yadav
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University Chandigarh 160014 India
| | - Sadhika Khullar
- Department of Chemistry Dr. B. R. Ambedkar National Institute of Technology Jalandhar 144011 Punjab India
| | - Sanjay K. Mandal
- Department of Chemical Sciences Indian Institute of Science Education and Research Mohali 140306 Punjab India
| | - Aman Bhalla
- Department of Chemistry and Centre of Advanced Studies in Chemistry Panjab University Chandigarh 160014 India
| |
Collapse
|
32
|
Kielholz T, Walther M, Jung N, Windbergs M. Electrospun fibers loaded with antimicrobial peptides for treatment of wound infections. Eur J Pharm Biopharm 2022; 179:246-255. [PMID: 36150615 DOI: 10.1016/j.ejpb.2022.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/23/2022] [Accepted: 09/15/2022] [Indexed: 11/04/2022]
Abstract
The widespread resistance of clinically relevant bacteria against established antibiotics emphasizes the urgent need for novel therapeutics. In this context, wound infections constitute a specific challenge, as most systemically applied antibiotics are insufficiently available at the site of infection. Therefore, the local treatment of infected wounds poses a particular challenge regarding the appropriate release kinetics of actives and their residence time in the wound bed. Consequently, design and development of novel, drug-loaded wound dressings constitute a major research focus for the effective treatment of wound infections. In this study, we employed electrospinning to design drug-loaded wound dressings, incorporating the therapeutically promising antimicrobial peptide tyrothricin. By parallel electrospinning, we combined different ratios of water-soluble polyvinyl pyrrolidone and water-insoluble methacrylate copolymer (EudragitE), in order to take advantage of their specific mechanical stability and dissolution properties. We fabricated fiber mats constituting mechanically stable wound dressings with a controlled drug release profile, combining an initial burst release above the minimal inhibitory concentration of known wound pathogens and a subsequent prolonged antimicrobial effect of the active ingredient. Antimicrobial activity against Staphylococcusaureus and Staphylococcusepidermidis was successfully proven, thereby introducing our tyrothricin-loaded fiber mats as a promising prospective therapy against typical wound-associated pathogens.
Collapse
Affiliation(s)
- Tobias Kielholz
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Marcel Walther
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Nathalie Jung
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Maike Windbergs
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
33
|
Zhang H, Dong M, Yuan S, Jin W. Oral glucagon-like peptide 1 analogue ameliorates glucose intolerance in db/db mice. Biotechnol Lett 2022; 44:1149-1162. [PMID: 36006576 DOI: 10.1007/s10529-022-03288-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 08/04/2022] [Indexed: 11/02/2022]
Abstract
OBJECTIVES We constructed a recombinant oral GLP-1 analogue in Lactococcus lactis (L. lactis) and evaluated its physiological functions. RESULTS In silico docking suggested the alanine at position 8 substituted with serine (A8SGLP-1) reduced binding of DPP4, which translated to reduced cleavage by DPP4 with minimal changes in stability. This was further confirmed by an in vitro enzymatic assay which showed that A8SGLP-1 significantly increased half-life upon DPP4 treatment. In addition, recombinant L. lactis (LL-A8SGLP-1) demonstrated reduced fat mass with no changes in body weight, significant improvement of random glycemic control and reduced systemic inflammation compared with WT GLP-1 in db/db mice. CONCLUSION LL-A8SGLP-1 adopted in live biotherapeutic products reduce blood glucose in db/db mice without affecting its function.
Collapse
Affiliation(s)
- Hanlin Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, 1 West Beichen Rd. No. 5, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Meng Dong
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, 1 West Beichen Rd. No. 5, Beijing, 100101, China
| | - Shouli Yuan
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, 1 West Beichen Rd. No. 5, Beijing, 100101, China
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, 1 West Beichen Rd. No. 5, Beijing, 100101, China.
| |
Collapse
|
34
|
Yao Q, Zhang J, Pan G, Chen B. Mussel-Inspired Clickable Antibacterial Peptide Coating on Ureteral Stents for Encrustation Prevention. ACS APPLIED MATERIALS & INTERFACES 2022; 14:36473-36486. [PMID: 35917447 DOI: 10.1021/acsami.2c09448] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Long-term indwelling catheters or stents often cause complications like infection, encrustation, hematuria, pain, and so on. The source of these problems is bacteria, which can form biofilms on the stents to reduce antibiotic sensitivity and produce urease to form encrustation by increasing the urine pH. Urinary tract infection (UTI) can aggravate the body damage and even seriously endanger lives, and the encrustation will block the stents, which can cause hydronephrosis and renal function damage. Therefore, the prevention of UTI and encrustation represents a great challenge in clinical ureteral stent uses. In this work, a clickable mussel-inspired peptide and antimicrobial peptide (AMP) were used to functionalize the commercial stents' surfaces to inhibit long-term infection and encrustation caused by bacteria. Copper (Cu) ions were used to coordinate the mussel-inspired peptide to improve the stability. The AMP with an azido group was clicked to the mussel-inspired Cu-coordinated peptide coating through click chemistry. The bio-inspired antibacterial coating was constructed with excellent stability, bactericidal properties, and improved biological compatibility. In in vitro and in vivo experiments, it was further found that the coating showed bactericidal and encrustation reduction abilities. This study thus developed an effective, safe, and stable AMP coating on urinary stents/catheters capable of long-term antibacterial and encrustation inhibition.
Collapse
Affiliation(s)
- Qin Yao
- Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, Jiangsu 212001, P. R. China
| | - Jinyi Zhang
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 304 Xuefu Road, Zhenjiang, Jiangsu 212013, P. R. China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 304 Xuefu Road, Zhenjiang, Jiangsu 212013, P. R. China
| | - Binghai Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, Jiangsu 212001, P. R. China
| |
Collapse
|
35
|
Balcão VM, Moreli FC, Silva EC, Belline BG, Martins LF, Rossi FPN, Pereira C, Vila MMDC, da Silva AM. Isolation and Molecular Characterization of a Novel Lytic Bacteriophage That Inactivates MDR Klebsiella pneumoniae Strains. Pharmaceutics 2022; 14:pharmaceutics14071421. [PMID: 35890314 PMCID: PMC9324672 DOI: 10.3390/pharmaceutics14071421] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023] Open
Abstract
The worldwide increase in serious infections caused by multidrug-resistant (MDR) K. pneumoniae emphasizes the urgent need of new therapeutic strategies for the control of this pathogen. There is growing interest in the use of bacteriophages (or phages) to treat K. pneumoniae infections, and newly isolated phages are needed. Here, we report the isolation and physical/biological/molecular characterization of a novel lytic phage and its efficacy in the control of MDR K. pneumoniae. The phage vB_KpnS_Uniso31, referred to hereafter as phage Kpn31, was isolated from hospital wastewater using K. pneumoniae CCCD-K001 as the host. Phage Kpn31 presents a siphovirus-like morphotype and was classified as Demerecviridae; Sugarlandvirus based on its complete genome sequence. The 113,444 bp Kpn31 genome does not encode known toxins or antimicrobial resistance genes, nor does it encode depolymerases related sequences. Phage Kpn31 showed an eclipse time of 15 min and a burst size of 9.12 PFU/host cell, allowing us to conclude it replicates well in K. pneumoniae CCCD-K001 with a latency period of 30 min. Phage Kpn31 was shown to be effective against at least six MDR K. pneumoniae clinical isolates in in vitro antibacterial activity assays. Based on its features, phage Kpn31 has potential for controlling infections caused by MDR K. pneumoniae.
Collapse
Affiliation(s)
- Victor M Balcão
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
- Department of Biology and CESAM, Campus Universitário de Santiago, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Fernanda C Moreli
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| | - Erica C Silva
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| | - Bianca G Belline
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| | - Layla F Martins
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Fernando P N Rossi
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Carla Pereira
- Department of Biology and CESAM, Campus Universitário de Santiago, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Marta M D C Vila
- PhageLab, Laboratory of Biofilms and Bacteriophages, University of Sorocaba, Sorocaba 18023-000, Brazil
| | - Aline M da Silva
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
36
|
Blair JMA, Zeth K, Bavro VN, Sancho-Vaello E. The role of bacterial transport systems in the removal of host antimicrobial peptides in Gram-negative bacteria. FEMS Microbiol Rev 2022; 46:6617596. [PMID: 35749576 PMCID: PMC9629497 DOI: 10.1093/femsre/fuac032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/23/2022] [Accepted: 06/22/2022] [Indexed: 01/09/2023] Open
Abstract
Antibiotic resistance is a global issue that threatens our progress in healthcare and life expectancy. In recent years, antimicrobial peptides (AMPs) have been considered as promising alternatives to the classic antibiotics. AMPs are potentially superior due to their lower rate of resistance development, since they primarily target the bacterial membrane ('Achilles' heel' of the bacteria). However, bacteria have developed mechanisms of AMP resistance, including the removal of AMPs to the extracellular space by efflux pumps such as the MtrCDE or AcrAB-TolC systems, and the internalization of AMPs to the cytoplasm by the Sap transporter, followed by proteolytic digestion. In this review, we focus on AMP transport as a resistance mechanism compiling all the experimental evidence for the involvement of efflux in AMP resistance in Gram-negative bacteria and combine this information with the analysis of the structures of the efflux systems involved. Finally, we expose some open questions with the aim of arousing the interest of the scientific community towards the AMPs-efflux pumps interactions. All the collected information broadens our understanding of AMP removal by efflux pumps and gives some clues to assist the rational design of AMP-derivatives as inhibitors of the efflux pumps.
Collapse
Affiliation(s)
- Jessica M A Blair
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Kornelius Zeth
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark
| | - Vassiliy N Bavro
- School of Life Sciences, University of Essex, Colchester, CO4 3SQ, United Kingdom
| | - Enea Sancho-Vaello
- Corresponding author. College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom. E-mail:
| |
Collapse
|
37
|
Rahman MRT, Fliss I, Biron E. Insights in the Development and Uses of Alternatives to Antibiotic Growth Promoters in Poultry and Swine Production. Antibiotics (Basel) 2022; 11:766. [PMID: 35740172 PMCID: PMC9219610 DOI: 10.3390/antibiotics11060766] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
The overuse and misuse of antibiotics has contributed to the rise and spread of multidrug-resistant bacteria. To address this global public health threat, many countries have restricted the use of antibiotics as growth promoters and promoted the development of alternatives to antibiotics in human and veterinary medicine and animal farming. In food-animal production, acidifiers, bacteriophages, enzymes, phytochemicals, probiotics, prebiotics, and antimicrobial peptides have shown hallmarks as alternatives to antibiotics. This review reports the current state of these alternatives as growth-promoting factors for poultry and swine production and describes their mode of action. Recent findings on their usefulness and the factors that presently hinder their broader use in animal food production are identified by SWOT (strength, weakness, opportunity, and threat) analysis. The potential for resistance development as well as co- and cross-resistance with currently used antibiotics is also discussed. Using predetermined keywords, we searched specialized databases including Scopus, Web of Science, and Google Scholar. Antibiotic resistance cannot be stopped, but its spreading can certainly be hindered or delayed with the development of more alternatives with innovative modes of action and a wise and careful use of antimicrobials in a One Health approach.
Collapse
Affiliation(s)
- Md Ramim Tanver Rahman
- Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada;
- Laboratory of Medicinal Chemistry, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC G1V 0A6, Canada;
| | - Ismail Fliss
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC G1V 0A6, Canada;
- Food Science Department, Faculty of Agriculture and Food Sciences, Université Laval, Québec, QC G1V 0A6, Canada
| | - Eric Biron
- Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada;
- Laboratory of Medicinal Chemistry, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC G1V 0A6, Canada;
| |
Collapse
|
38
|
Longitudinal Evaluation of Gut Bacteriomes and Viromes after Fecal Microbiota Transplantation for Eradication of Carbapenem-Resistant Enterobacteriaceae. mSystems 2022; 7:e0151021. [PMID: 35642928 PMCID: PMC9239097 DOI: 10.1128/msystems.01510-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Understanding the role of fecal microbiota transplantation (FMT) in the decolonization of multidrug-resistant organisms (MDRO) is critical. Specifically, little is known about virome changes in MDRO-infected subjects treated with FMT. Using shotgun metagenomic sequencing, we characterized longitudinal dynamics of the gut virome and bacteriome in three recipients who successfully decolonized carbapenem-resistant Enterobacteriaceae (CRE), including Klebsiella spp. and Escherichia coli, after FMT. We observed large shifts of the fecal bacterial microbiota resembling a donor-like community after transfer of a fecal microbiota dominated by the genus Ruminococcus. We found a substantial expansion of Klebsiella phages after FMT with a concordant decrease of Klebsiella spp. and striking increase of Escherichia phages in CRE E. coli carriers after FMT. We also observed the CRE elimination and similar evolution of Klebsiella phage in mice, which may play a role in the collapse of the Klebsiella population after FMT. In summary, our pilot study documented bacteriome and virome alterations after FMT which mediate many of the effects of FMT on the gut microbiome community. IMPORTANCE Fecal microbiota transplantation (FMT) is an effective treatment for multidrug-resistant organisms; however, introducing a complex mixture of microbes also has unknown consequences for landscape features of gut microbiome. We sought to understand bacteriome and virome alterations in patients undergoing FMT to treat infection with carbapenem-resistant Enterobacteriaceae. This finding indicates that transkingdom interactions between the virome and bacteriome communities may have evolved in part to support efficient FMT for treating CRE.
Collapse
|
39
|
Huang Y, Yang N, Teng D, Mao R, Hao Y, Ma X, Wei L, Wang J. Antibacterial peptide NZ2114-loaded hydrogel accelerates Staphylococcus aureus-infected wound healing. Appl Microbiol Biotechnol 2022; 106:3639-3656. [PMID: 35524777 DOI: 10.1007/s00253-022-11943-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/07/2022] [Accepted: 04/23/2022] [Indexed: 12/19/2022]
Abstract
Wound infection caused by Staphylococcus aureus (S. aureus) is a great challenge which has caused significant burden and economic loss to the medical system. NZ2114, a plectasin-derived peptide, is an antibacterial agent for preventing and treating S. aureus infection, especially for methicillin-resistant S. aureus (MRSA) infection. Here, three-dimensional reticulated antimicrobial peptide (AMP) NZ2114 hydrogels were developed based on hydroxypropyl cellulose (HPC) and sodium alginate (SA); they displayed sustained and stable release properties (97.88 ± 1.79% and 91.1 ± 10.52% release rate in 72 h, respectively) and good short-term cytocompatibility and hemocompatibility. But the HPC-NZ2114 hydrogel had a smaller pore size (diameter 0.832 ± 0.420 μm vs. 3.912 ± 2.881 μm) and better mechanical properties than that of the SA-NZ2114 hydrogel. HPC/SA-NZ2114 hydrogels possess efficient antimicrobial activity in vitro and in vivo. In a full-thickness skin defect model, the wound closure of the 1.024 mg/g HPC-NZ2114 hydrogel group was superior to those of the SA-NZ2114 hydrogel and antibiotic groups on day 7. The HPC-NZ2114 hydrogel accelerated wound healing by reducing inflammation and promoting the production of vascular endothelial growth factor (VEGF), endothelial growth factor (EGF) and angiogenesis (CD31) through histological and immunohistochemistry evaluation. These data indicated that the HPC-NZ2114 hydrogel is an excellent candidate for S. aureus infection wound dressing. KEY POINTS: •NZ2114 hydrogels showed potential in vitro bactericidal activity against S. aureus •NZ2114 hydrogels could release continuously for 72 h and had good biocompatibility •NZ2114 hydrogels could effectively promote S. aureus-infected wound healing.
Collapse
Affiliation(s)
- Yan Huang
- School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology - WIT, Wuhan, People's Republic of China.,Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, and Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Haidian District, 12 Zhongguancun Nandajie St, Beijing, 100081, People's Republic of China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Na Yang
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, and Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Haidian District, 12 Zhongguancun Nandajie St, Beijing, 100081, People's Republic of China. .,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China.
| | - Da Teng
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, and Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Haidian District, 12 Zhongguancun Nandajie St, Beijing, 100081, People's Republic of China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Ruoyu Mao
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, and Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Haidian District, 12 Zhongguancun Nandajie St, Beijing, 100081, People's Republic of China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Ya Hao
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, and Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Haidian District, 12 Zhongguancun Nandajie St, Beijing, 100081, People's Republic of China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Xuanxuan Ma
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, and Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Haidian District, 12 Zhongguancun Nandajie St, Beijing, 100081, People's Republic of China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Lingyun Wei
- School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology - WIT, Wuhan, People's Republic of China.
| | - Jianhua Wang
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, and Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Haidian District, 12 Zhongguancun Nandajie St, Beijing, 100081, People's Republic of China. .,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China.
| |
Collapse
|
40
|
Samir S, El-Far A, Okasha H, Mahdy R, Samir F, Nasr S. Isolation and characterization of lytic bacteriophages from sewage at an egyptian tertiary care hospital against methicillin-resistant Staphylococcus aureus clinical isolates. Saudi J Biol Sci 2022; 29:3097-3106. [PMID: 35360502 PMCID: PMC8961222 DOI: 10.1016/j.sjbs.2022.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 01/25/2022] [Accepted: 03/15/2022] [Indexed: 12/03/2022] Open
Abstract
Background Methicillin resistant Staphylococcus aureus (MRSA) is a pathogen to humans causing life-threatening infections. MRSA have the capability to grow resistance to many antibiotics, and phage therapy is one treatment option for this infection. Objectives The aim of the present study was to isolate and characterize the lytic bacteriophages specific to MRSA from domestic sewage water at a tertiary care hospital in Egypt. Methods Thirty MRSA strains were isolated from different clinical samples admitted to the microbiology lab at Theodor Bilharz Research institute (TBRI) hospital, Giza, Egypt. They were confirmed to be MRSA through phenotypic detection and conventional PCR for mecA gene. They were used for the isolation of phages from sewage water of TBRI hospital. Plaque assay was applied to purify and quantify the titer of the isolated phages. The host range of the isolated phages was detected using the spot test assay. The morphology of phages was confirmed using transmission electron microscope (TEM). Digestion of DNA extracted from phages with endonuclease enzymes including EcoRI and SmaI was performed. SDS-PAGE was performed to analyze MRSA specific phage proteins. As a positive control prophages were isolated from a mitomycin C (MitC) treated culture of S. aureus strain ATCC25923. Further characterization using conventional polymerase chain reaction (PCR) was used to select three known Staphylophages by detecting the endolysin gene of phage K, the polymerase gene of phage 44AHJD, and the minor tail gene of phage P68. Results Isolated phages in this research displayed a wide host range against MRSA using the spot test, out of thirty tested MRSA isolates 24 were sensitive and got lysed (80%). The titer of the phages was estimated to be 1.04 × 106 pfu/ml using plaque test. Identification of head and tail morphology of the phages was achieved using TEM and they were designated to tailed phages of order Caudovirales, they composed an icosahedral capsid. Prophages were isolated through MitC induction. DNA of phages was digested by endonuclease enzymes. Conventional PCR yielded 341 bp of phage K endolysin gene and phage P68 minor tail protein gene 501 bp. Protein analysis using SDS-PAGE showed 4 proteins of sizes between 42 kDa and 140 kDa. Conclusion Phages isolated here are alike to others mentioned in previous studies. The high broad host range of the isolated phages is promising to control MRSA and can be in the future commercially suitable for treatment as lysate preparations. Animal models of phage-bacterial interaction will be our next step that may help in resolving the multidrug resistant crisis of MRSA in Egypt.
Collapse
Key Words
- AMR, antimicrobial resistance
- CLSI, clinical and laboratory standards institute
- Caudovirales
- ESKAPE, (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species)
- FOX, Cefoxitin
- ITS, Internal transcribed spacer
- LB, Luria-Bertani
- Lytic bacteriophages
- MDR, Multidrug-resistant
- MRSA
- MRSA, Methicillin Resistant Staphylococcus aureus
- MitC, mitomycin C
- Mitomycin C
- NGS, double-stranded, ds, next generation sequencing
- OX, Oxacillin
- PCR, Polymerase chain reaction
- PFU, Plaque forming unit
- PTA, phosphotungstic acid
- Polymerase chain reaction
- Restriction digestion
- S. aureus, Staphylococcus aureus
- SDS-polyacrylamide gel electrophoresis
- Sewage
- TBRI, Theodor Bilharz research Institute
- TEM, Transmission electron microscopy
- Transmission electron microscopy
Collapse
Affiliation(s)
- Safia Samir
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| | - Amira El-Far
- Microbiology Department, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| | - Hend Okasha
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| | - Rania Mahdy
- Microbiology Department, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| | - Fatima Samir
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| | - Sami Nasr
- Biochemistry and Molecular Biology Department, Theodor Bilharz Research Institute (TBRI), Giza, Egypt
| |
Collapse
|
41
|
Xiong J, Yang ZR, Lv N, Du K, Suo H, Du S, Tao J, Jiang H, Zhu J. Self-adhesive Hyaluronic Acid/Antimicrobial Peptide Composite Hydrogel with Antioxidant Capability and Photothermal Activity for Infected Wound Healing. Macromol Rapid Commun 2022; 43:e2200176. [PMID: 35451187 DOI: 10.1002/marc.202200176] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/13/2022] [Indexed: 11/06/2022]
Abstract
Bacterial infection can delay wound healing, causing wounds to deteriorate and even threatening the patient's life. Recently, although many composite hydrogels as wound dressing have been developed, it is still highly desired to construct photothermal hydrogels with antimicrobial and antioxidant properties to accelerate the infected wound healing. In this work, we develop a hyaluronic acid (HA)-based composite hydrogel consisting of a dopamine-substituted antimicrobial peptide (DAP) and Iron (III) ions, which exhibits photothermal-assisted promotion and acceleration of healing process of bacteria-infected wounds. DAP, serving as both antimicrobial agent and ROS-scavenger, forms Schiff's base bonds with aldehyde hyaluronic acid (AHA) and iron-catechol coordination bonds to reinforce the composite hydrogel. The presence of Fe3+ can also promote covalent polymerization of dopamine, which endows the hydrogel with photothermal capacity. The in vitro and in vivo experiments prove that the composite hydrogel can effectively accelerate the infected wound healing process, including antibacterial, accelerated collagen deposition and re-epithelization. This study suggests that the multifunctional composite hydrogel possesses remarkable potential for bacteria-infected wound healing by combining inherent antimicrobial activity, antioxidant capability and photothermal effect. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jingyi Xiong
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Zhuo-Ran Yang
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Niannian Lv
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Kehan Du
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Huinan Suo
- Department of Dermatology, Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Shuo Du
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Juan Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Hao Jiang
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Jintao Zhu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| |
Collapse
|
42
|
Al-Zrkani MK, Abdulkareem RA, Al-Fahad D, Al Shouber M, Nasr AMS, Al-Khdhairawi A. Elucidating novel antibacterial compounds from the NPASS database against the FimH lectin domain for the treatment of urinary tract infections: an in-silico study. J Biomol Struct Dyn 2022; 41:3914-3925. [PMID: 35403563 DOI: 10.1080/07391102.2022.2059009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The increase in multidrug-resistant pathogens in urinary tract infections (UTIs) among communities and hospitals threatens our ability to treat these common pathogens. Uropathogenic Escherichia coli (UPEC) strains are the most frequent uropathies linked to the development of UTIs. This work aims to introduce bioactive natural products via virtual screening of small molecules from a public database to prevent biofilm formation by inhibiting FimH, a type 1 fimbriae that plays a crucial role in UPEC pathogenicity. A total of 30926 small molecules from the NPASS database were subjected to screening via molecular docking. Followed by performing in silico ADME studies, seven molecules showed promising docking results ranging from -6.8 to -8.7 kcal/mol. As a result of the docking score findings, 100 ns Molecular dynamics (MD) simulations were performed. Based on MM-PBSA analysis, NPC313334 ligand showed high binding affinity -42 and stability with the binding pocket of FimH protein during molecular dynamic simulations. DFT calculations were also performed on the ligands to calculate the HOMO-LUMO energies of the compounds in order to an idea about their structure and reactivity. This research suggests that NPC313334 may be a possible antibacterial drug candidate that targets FimH to reduce the number of UPEC-related urinary tract infections. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mrtatha K Al-Zrkani
- Institute of Genetic Engineering & Biotechnology Research, Baghdad University, Baghdad, Iraq
| | - Rafid A Abdulkareem
- Institute of Genetic Engineering & Biotechnology Research, Baghdad University, Baghdad, Iraq
| | - Dhurgham Al-Fahad
- Department of Pharmaceutical Science, College of Pharmacy, University of Thi-Qar, Nasiriyah, Iraq
| | - Marwah Al Shouber
- Department of Pharmaceutical, Al Zahra Teaching Hospital of Wasit, Kut, Iraq
| | | | - Ahmad Al-Khdhairawi
- Department of Biological Science and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, UKM Bangi, Selangor, Malaysia
| |
Collapse
|
43
|
Efficacy Assessment of Phage Therapy in Treating Staphylococcus aureus-Induced Mastitis in Mice. Viruses 2022; 14:v14030620. [PMID: 35337027 PMCID: PMC8954217 DOI: 10.3390/v14030620] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 02/06/2023] Open
Abstract
The primary aim of this study was to evaluate the efficacy of phage against mastitis induced by drug-resistant S. aureus in a mouse model. In this study, five S. aureus phages—4086-1, 4086-2, 4086-3, 4086-4, and 4086-6—were isolated from milk samples secreted by mastitis cows. Transmission electron microscopy showed that all the five phages had icosahedral heads and short non-contractile tails, which are typical characteristics of the family Podoviridae. All these phages were species-specific against S. aureus. The one-step growth curve showed a short latency period (10–20 min) and high burst size (up to 400 PFU/infected cell). To evaluate the effectiveness of the phage 4086-1 in the treatment against mastitis, a mouse model of mastitis was challenged with drug-resistant S. aureus. The results showed the proliferation of S. aureus in the mammary glands was significantly inhibited after treating by phage 4086-1. The concentrations of TNF-α and IL-6 decreased significantly, which demonstrated the phages could effectively alleviate the inflammatory responses. Furthermore, the histopathological analysis showed that inflammatory infiltration in the mammary glands was significantly reduced. These results demonstrate that phage may be a promising alternative therapy against mastitis caused by drug-resistant S. aureus.
Collapse
|
44
|
Isolation, Characterization, and Genomic Analysis of Three Novel E. coli Bacteriophages That Effectively Infect E. coli O18. Microorganisms 2022; 10:microorganisms10030589. [PMID: 35336164 PMCID: PMC8954371 DOI: 10.3390/microorganisms10030589] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 01/27/2023] Open
Abstract
Escherichia coli (E. coli) is one of the most common pathogenic bacteria worldwide. Avian pathogenic E. coli (APEC) causes severe systemic disease in poultry (Colibacillosis), and accordingly, has an extreme risk to the poultry industry and public health worldwide. Due to the increased rate of multi-drug resistance among these bacteria, it is necessary to find an alternative therapy to antibiotics to treat such infections. Bacteriophages are considered one of the best solutions. This study aimed to isolate, characterize, and evaluate the potential use of isolated bacteriophages to control E. coli infections in poultry. Three novel phages against E. coli O18 were isolated from sewage water and characterized in vitro. The genome size of the three phages was estimated to be 44,776 bp, and the electron microscopic analysis showed that they belonged to the Siphoviridae family, in the order Caudovirales. Phages showed good tolerance to a broad range of pH and temperature. The complete genomes of three phages were sequenced and deposited into the GenBank database. The closely related published genomes of Escherichia phages were identified using BLASTn alignment and phylogenetic trees. The prediction of the open reading frames (ORFs) identified protein-coding genes that are responsible for functions that have been assigned such as cell lysis proteins, DNA packaging proteins, structural proteins, and DNA replication/transcription/repair proteins.
Collapse
|
45
|
Bacteriophages in the Control of Aeromonas sp. in Aquaculture Systems: An Integrative View. Antibiotics (Basel) 2022; 11:antibiotics11020163. [PMID: 35203766 PMCID: PMC8868336 DOI: 10.3390/antibiotics11020163] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 11/17/2022] Open
Abstract
Aeromonas species often cause disease in farmed fish and are responsible for causing significant economic losses worldwide. Although vaccination is the ideal method to prevent infectious diseases, there are still very few vaccines commercially available in the aquaculture field. Currently, aquaculture production relies heavily on antibiotics, contributing to the global issue of the emergence of antimicrobial-resistant bacteria and resistance genes. Therefore, it is essential to develop effective alternatives to antibiotics to reduce their use in aquaculture systems. Bacteriophage (or phage) therapy is a promising approach to control pathogenic bacteria in farmed fish that requires a heavy understanding of certain factors such as the selection of phages, the multiplicity of infection that produces the best bacterial inactivation, bacterial resistance, safety, the host’s immune response, administration route, phage stability and influence. This review focuses on the need to advance phage therapy research in aquaculture, its efficiency as an antimicrobial strategy and the critical aspects to successfully apply this therapy to control Aeromonas infection in fish.
Collapse
|
46
|
Santamarina SC, Heredia DA, Durantini AM, Durantini EN. Antimicrobial Photosensitizing Material Based on Conjugated Zn(II) Porphyrins. Antibiotics (Basel) 2022; 11:91. [PMID: 35052968 PMCID: PMC8773278 DOI: 10.3390/antibiotics11010091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 01/13/2023] Open
Abstract
The widespread use of antibiotics has led to a considerable increase in the resistance of microorganisms to these agents. Consequently, it is imminent to establish new strategies to combat pathogens. An alternative involves the development of photoactive polymers that represent an interesting strategy to kill microbes and maintain aseptic surfaces. In this sense, a conjugated polymer (PZnTEP) based on Zn(II) 5,10,15,20-tetrakis-[4-(ethynyl)phenyl]porphyrin (ZnTEP) was obtained by the homocoupling reaction of terminal alkyne groups. PZnTEP exhibits a microporous structure with high surface areas allowing better interaction with bacteria. The UV-visible absorption spectra show the Soret and Q bands of PZnTEP red-shifted by about 18 nm compared to those of the monomer. Also, the conjugate presents the two red emission bands, characteristic of porphyrins. This polymer was able to produce singlet molecular oxygen and superoxide radical anion in the presence of NADH. Photocytotoxic activity sensitized by PZnTEP was investigated in bacterial suspensions. No viable Staphylococcus aureus cells were detected using 0.5 µM PZnTEP and 15 min irradiation. Under these conditions, complete photoinactivation of Escherichia coli was observed in the presence of 100 mM KI. Likewise, no survival was detected for E. coli incubated with 1.0 µM PZnTEP after 30 min irradiation. Furthermore, polylactic acid surfaces coated with PZnTEP were able to kill efficiently these bacteria. This surface can be reused for at least three photoinactivation cycles. Therefore, this conjugated photodynamic polymer is an interesting antimicrobial photoactive material for designing and developing self-sterilizing surfaces.
Collapse
Affiliation(s)
- Sofía C Santamarina
- IDAS-CONICET, Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta Nacional 36 Km 601, Río Cuarto, Córdoba X5804BYA, Argentina
| | - Daniel A Heredia
- IDAS-CONICET, Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta Nacional 36 Km 601, Río Cuarto, Córdoba X5804BYA, Argentina
| | - Andrés M Durantini
- IDAS-CONICET, Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta Nacional 36 Km 601, Río Cuarto, Córdoba X5804BYA, Argentina
| | - Edgardo N Durantini
- IDAS-CONICET, Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta Nacional 36 Km 601, Río Cuarto, Córdoba X5804BYA, Argentina
| |
Collapse
|
47
|
Harada LK, Silva EC, Rossi FP, Cieza B, Oliveira TJ, Pereira C, Tomazetto G, Silva BB, Squina FM, Vila MM, Setubal JC, Ha T, da Silva AM, Balcão VM. Characterization and in vitro testing of newly isolated lytic bacteriophages for the biocontrol of Pseudomonas aeruginosa. Future Microbiol 2022; 17:111-141. [PMID: 34989245 DOI: 10.2217/fmb-2021-0027] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aim: Two lytic phages were isolated using P. aeruginosa DSM19880 as host and fully characterized. Materials & methods: Phages were characterized physicochemically, biologically and genomically. Results & conclusion: Host range analysis revealed that the phages also infect some multidrug-resistant (MDR) P. aeruginosa clinical isolates. Increasing MOI from 1 to 1000 significantly increased phage efficiency and retarded bacteria regrowth, but phage ph0034 (reduction of 7.5 log CFU/ml) was more effective than phage ph0031 (reduction of 5.1 log CFU/ml) after 24 h. Both phages belong to Myoviridae family. Genome sequencing of phages ph0031 and ph0034 showed that they do not carry toxin, virulence, antibiotic resistance and integrase genes. The results obtained are highly relevant in the actual context of bacterial resistance to antibiotics.
Collapse
Affiliation(s)
- Liliam K Harada
- PhageLab - Laboratory of Biofilms & Bacteriophages, University of Sorocaba, Sorocaba/SP, Brazil
| | - Erica C Silva
- PhageLab - Laboratory of Biofilms & Bacteriophages, University of Sorocaba, Sorocaba/SP, Brazil
| | - Fernando Pn Rossi
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Basilio Cieza
- Department of Biophysics & Biophysical Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - Thais J Oliveira
- PhageLab - Laboratory of Biofilms & Bacteriophages, University of Sorocaba, Sorocaba/SP, Brazil
| | - Carla Pereira
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Geizecler Tomazetto
- Department of Engineering, Biological & Chemical Engineering Section (BCE), Aarhus University, Aarhus, Denmark
| | - Bianca B Silva
- PhageLab - Laboratory of Biofilms & Bacteriophages, University of Sorocaba, Sorocaba/SP, Brazil
| | - Fabio M Squina
- PhageLab - Laboratory of Biofilms & Bacteriophages, University of Sorocaba, Sorocaba/SP, Brazil
| | - Marta Mdc Vila
- PhageLab - Laboratory of Biofilms & Bacteriophages, University of Sorocaba, Sorocaba/SP, Brazil
| | - João C Setubal
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Taekjip Ha
- Department of Biophysics & Biophysical Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - Aline M da Silva
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Victor M Balcão
- PhageLab - Laboratory of Biofilms & Bacteriophages, University of Sorocaba, Sorocaba/SP, Brazil.,Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| |
Collapse
|
48
|
Yao Q, Chen B, Bai J, He W, Chen X, Geng D, Pan G. Bio-inspired antibacterial coatings on urinary stents for encrustation prevention. J Mater Chem B 2022; 10:2584-2596. [PMID: 34984428 DOI: 10.1039/d1tb02318g] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Urinary tract infection (UTI) represents one of the most common nosocomial infections, which is mainly related to indwelling catheters or stents. In addition to the formation of biofilms to reduce antibiotic sensitivity, the urease-producing bacteria can also increase urine pH, causing Ca2+ and Mg2+ deposition and finally catheter obstruction. The prevention of UTIs and its complication (i.e., encrustation) thus is a great challenge in design of catheters and ureteral stents. In this work, a metal-catechol-assisted mussel chemistry (i.e., dopamine self-polymerization) was employed for surface functionalization of commercially available catheters with antimicrobial peptides (AMP), for the purpose of long-term anti-infection and encrustation prevention. To improve the stability of the polydopamine coating on polymeric stents, we used Cu2+-coordinated dopamine self-polymerization. Then, a cysteine-terminated AMP was introduced on the polydopamine coating through Michael addition. We found that the Cu2+-coordinated polydopamine coating showed improved stability and antibacterial effect. The cytotoxicity test confirmed that the bioinspired antibacterial coating showed good biocompatibility and no obvious toxicity. The results confirmed that the stents with AMP could in situ inhibit bacterial growth and biofilm formation, and finally reduce the deposition of struvite and hydroxyapatite crystals both in vitro and in vivo. We anticipate that this bioinspired strategy would develop a safe, stable and effective antibacterial coating on urinary tract medical devices for long-term bacterial inhibition and encrustation prevention.
Collapse
Affiliation(s)
- Qin Yao
- Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, Jiangsu, 212001, China.
| | - Binghai Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, Jiangsu, 212001, China.
| | - Jiaxiang Bai
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Wenbo He
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China.
| | - Xu Chen
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China.
| | - Dechun Geng
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Medical College, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
49
|
PATO U, RIFTYAN E, AYU DF, JONNAIDI NN, WAHYUNI MS, FERUNI JA, ABDEL-WAHHAB MA. Antibacterial efficacy of lactic acid bacteria and bacteriocin isolated from Dadih’s against Staphylococcus aureus. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.27121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
50
|
Reduced Susceptibility and Increased Resistance of Bacteria against Disinfectants: A Systematic Review. Microorganisms 2021; 9:microorganisms9122550. [PMID: 34946151 PMCID: PMC8706950 DOI: 10.3390/microorganisms9122550] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 01/22/2023] Open
Abstract
Disinfectants are used to reduce the concentration of pathogenic microorganisms to a safe level and help to prevent the transmission of infectious diseases. However, bacteria have a tremendous ability to respond to chemical stress caused by biocides, where overuse and improper use of disinfectants can be reflected in a reduced susceptibility of microorganisms. This review aims to describe whether mutations and thus decreased susceptibility to disinfectants occur in bacteria during disinfectant exposure. A systematic literature review following PRISMA guidelines was conducted with the databases PubMed, Science Direct and Web of Science. For the final analysis, 28 sources that remained of interest were included. Articles describing reduced susceptibility or the resistance of bacteria against seven different disinfectants were identified. The important deviation of the minimum inhibitory concentration was observed in multiple studies for disinfectants based on triclosan and chlorhexidine. A reduced susceptibility to disinfectants and potentially related problems with antibiotic resistance in clinically important bacterial strains are increasing. Since the use of disinfectants in the community is rising, it is clear that reasonable use of available and effective disinfectants is needed. It is necessary to develop and adopt strategies to control disinfectant resistance.
Collapse
|