1
|
Mitra A. Combatting biofilm-mediated infections in clinical settings by targeting quorum sensing. Cell Surf 2024; 12:100133. [PMID: 39634722 PMCID: PMC11615143 DOI: 10.1016/j.tcsw.2024.100133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Biofilm-associated infections constitute a significant challenge in managing infectious diseases due to their high resistance to antibiotics and host immune responses. Biofilms are responsible for various infections, including urinary tract infections, cystic fibrosis, dental plaque, bone infections, and chronic wounds. Quorum sensing (QS) is a process of cell-to-cell communication that bacteria use to coordinate gene expression in response to cell density, which is crucial for biofilm formation and maintenance.. Its disruption has been proposed as a potential strategy to prevent or treat biofilm-associated infections leading to improved treatment outcomes for infectious diseases. This review article aims to provide a comprehensive overview of the literature on QS-mediated disruption of biofilms for treating infectious diseases. It will discuss the mechanisms of QS disruption and the various approaches that have been developed to disrupt QS in reference to multiple clinical pathogens. In particular, numerous studies have demonstrated the efficacy of QS disruption in reducing biofilm formation in various pathogens, including Pseudomonas aeruginosa and Staphylococcus aureus. Finally, the review will discuss the challenges and future directions for developing QS disruption as a clinical therapy for biofilm-associated infections. This includes the development of effective delivery systems and the identification of suitable targets for QS disruption. Overall, the literature suggests that QS disruption is a promising alternative to traditional antibiotic treatment for biofilm-associated infections and warrants further investigation.
Collapse
Affiliation(s)
- Arindam Mitra
- Department of Biological Sciences, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, India
| |
Collapse
|
2
|
Bahrami M, Serati Shirazi P, Moradi F, Hadi N, Sabbaghi N, Eslaminezhad S. How nanomaterials act against bacterial structures? a narrative review focusing on nanoparticle molecular mechanisms. Microb Pathog 2024; 196:107002. [PMID: 39393474 DOI: 10.1016/j.micpath.2024.107002] [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: 05/22/2024] [Revised: 09/01/2024] [Accepted: 10/07/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVE In recent years, significant progress has been made in the field of nanotechnology for the treatment and prevention of biofilm formation and Multidrug-resistant bacteria (MDR). MDR bacteria challenges is hazardous when microorganisms induce the formation of biofilms, which amplify resistance to antibiotics and promote the development of multidrug-resistant conditions. The unique physicochemical properties of certain nanomaterials make nanotechnology a promising option for combating MDR infections. Several studies have introduced nanomaterials with different antibacterial mechanisms that can effectively destroy MDR bacteria and their biofilms. This study reviews the research results, focusing on the various nanoparticle mechanisms that target bacterial structures. METHOD To accomplish this study, we conducted investigations to gather articles and relevant studies from validated medical databases such as Scopus, PubMed, Google Scholar, and Web of Science. The selected publications from 2007 to 2023. In this review, we provide a brief overview of nanoparticles, their mechanisms, and how they function against the structure of bacteria. Furthermore, we discuss the recent advancements in using certain nanoparticles to combat infection-induced biofilms and complications caused by multidrug resistance. FINDING Our findings demonstrate that various nanoparticles have the potential to effectively overcome bacterial infectious diseases by targeting biofilms and antibiotic-resistant strains. Additionally, the development of a new drug delivery approach based on nanosystems shows promise in overcoming antibiotic resistance and biofilms.
Collapse
Affiliation(s)
- Maryam Bahrami
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Serati Shirazi
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farhad Moradi
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Nahal Hadi
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Sabbaghi
- Department of Parasitology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sahba Eslaminezhad
- Department of Chemical Engineering, Shiraz Branch, Islamic Azad University, Shiraz, Iran; Pars Biotech Research & Development Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Saikia S, Chetia P. Antibiotics: From Mechanism of Action to Resistance and Beyond. Indian J Microbiol 2024; 64:821-845. [PMID: 39282166 PMCID: PMC11399512 DOI: 10.1007/s12088-024-01285-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/15/2024] [Indexed: 09/18/2024] Open
Abstract
Antibiotics are the super drugs that have revolutionized modern medicine by curing many infectious diseases caused by various microbes. They efficiently inhibit the growth and multiplication of the pathogenic microbes without causing adverse effects on the host. However, prescribing suboptimal antibiotic and overuse in agriculture and animal husbandry have led to the emergence of antimicrobial resistance, one of the most serious threats to global health at present. The efficacy of a new antibiotic is high when introduced; however, a small bacterial population attains resistance gradually and eventually survives. Understanding the mode of action of these miracle drugs, as well as their interaction with targets is very complex. However, it is necessary to fulfill the constant need for novel therapeutic alternatives to address the inevitable development of resistance. Therefore, considering the need of the hour, this article has been prepared to discuss the mode of action and recent advancements in the field of antibiotics. Efforts has also been made to highlight the current scenario of antimicrobial resistance and drug repurposing as a fast-track solution to combat the issue.
Collapse
Affiliation(s)
- Shyamalima Saikia
- Molecular Plant Taxonomy and Bioinformatics Research Laboratory, Department of Life Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| | - Pankaj Chetia
- Department of Life Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| |
Collapse
|
4
|
Liu X, Liu C, Lin Q, Shi T, Liu G. Exosome-loaded hydrogels for craniofacial bone tissue regeneration. Biomed Mater 2024; 19:052002. [PMID: 38815606 DOI: 10.1088/1748-605x/ad525c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/30/2024] [Indexed: 06/01/2024]
Abstract
It is common for maladies and trauma to cause significant bone deterioration in the craniofacial bone, which can cause patients to experience complications with their appearance and their ability to function. Regarding grafting procedures' complications and disadvantages, the newly emerging field of tissue regeneration has shown promise. Tissue -engineered technologies and their applications in the craniofacial region are increasingly gaining prominence with limited postoperative risk and cost. MSCs-derived exosomes are widely applied in bone tissue engineering to provide cell-free therapies since they not only do not cause immunological rejection in the same way that cells do, but they can also perform a cell-like role. Additionally, the hydrogel system is a family of multipurpose platforms made of cross-linked polymers with considerable water content, outstanding biocompatibility, and tunable physiochemical properties for the efficient delivery of commodities. Therefore, the promising exosome-loaded hydrogels can be designed for craniofacial bone regeneration. This review lists the packaging techniques for exosomes and hydrogel and discusses the development of a biocompatible hydrogel system and its potential for exosome continuous delivery for craniofacial bone healing.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Plastic Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Chang Liu
- Department of Plastic Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Qingquan Lin
- Institute of Applied Catalysis, College of Chemistry and Chemical Engineering, Yantai University, Yantai, People's Republic of China
| | - Ting Shi
- Department of Plastic Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Guanying Liu
- Department of Hand and Foot Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China
| |
Collapse
|
5
|
Tan Y, Scornet AL, Yap MNF, Zhang D. Machine learning-based classification reveals distinct clusters of non-coding genomic allelic variations associated with Erm-mediated antibiotic resistance. mSystems 2024; 9:e0043024. [PMID: 38953319 PMCID: PMC11264731 DOI: 10.1128/msystems.00430-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/05/2024] [Indexed: 07/04/2024] Open
Abstract
The erythromycin resistance RNA methyltransferase (erm) confers cross-resistance to all therapeutically important macrolides, lincosamides, and streptogramins (MLS phenotype). The expression of erm is often induced by the macrolide-mediated ribosome stalling in the upstream co-transcribed leader sequence, thereby triggering a conformational switch of the intergenic RNA hairpins to allow the translational initiation of erm. We investigated the evolutionary emergence of the upstream erm regulatory elements and the impact of allelic variation on erm expression and the MLS phenotype. Through systematic profiling of the upstream regulatory sequences across all known erm operons, we observed that specific erm subfamilies, such as ermB and ermC, have independently evolved distinct configurations of small upstream ORFs and palindromic repeats. A population-wide genomic analysis of the upstream ermB regions revealed substantial non-random allelic variation at numerous positions. Utilizing machine learning-based classification coupled with RNA structure modeling, we found that many alleles cooperatively influence the stability of alternative RNA hairpin structures formed by the palindromic repeats, which, in turn, affects the inducibility of ermB expression and MLS phenotypes. Subsequent experimental validation of 11 randomly selected variants demonstrated an impressive 91% accuracy in predicting MLS phenotypes. Furthermore, we uncovered a mixed distribution of MLS-sensitive and MLS-resistant ermB loci within the evolutionary tree, indicating repeated and independent evolution of MLS resistance. Taken together, this study not only elucidates the evolutionary processes driving the emergence and development of MLS resistance but also highlights the potential of using non-coding genomic allele data to predict antibiotic resistance phenotypes. IMPORTANCE Antibiotic resistance (AR) poses a global health threat as the efficacy of available antibiotics has rapidly eroded due to the widespread transmission of AR genes. Using Erm-dependent MLS resistance as a model, this study highlights the significance of non-coding genomic allelic variations. Through a comprehensive analysis of upstream regulatory elements within the erm family, we elucidated the evolutionary emergence and development of AR mechanisms. Leveraging population-wide machine learning (ML)-based genomic analysis, we transformed substantial non-random allelic variations into discernible clusters of elements, enabling precise prediction of MLS phenotypes from non-coding regions. These findings offer deeper insight into AR evolution and demonstrate the potential of harnessing non-coding genomic allele data for accurately predicting AR phenotypes.
Collapse
Affiliation(s)
- Yongjun Tan
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, Missouri, USA
| | - Alexandre Le Scornet
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mee-Ngan Frances Yap
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Dapeng Zhang
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, Missouri, USA
- Program of Bioinformatics and Computational Biology, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
6
|
Wang Y, Yang K, Li L, Yang L, Zhang S, Yu F, Hua L. Change characteristics, bacteria host, and spread risks of bioaerosol ARGs/MGEs from different stages in sewage and sludge treatment process. JOURNAL OF HAZARDOUS MATERIALS 2024; 469:134011. [PMID: 38492400 DOI: 10.1016/j.jhazmat.2024.134011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/21/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
The spread of antibiotic resistance genes (ARGs) in the atmospheric environment has seriously threatened human health. Wastewater treatment plants (WWTPs) are an important source of aerosol ARGs. A large WWTP, including sewage treatment process (SWP) and sludge treatment process (SDP), was selected in North China for sampling in this study. The content of ARGs, mobile genetic elements (MGEs), and bacterial genera in sewage/sludge and aerosols from different process stages was detected. The possible correlation between ARGs/ MGEs and bacteria was analyzed. The risk of antibiotic-resistant bacteria was evaluated and the diffusion of ARGs/MGEs was simulated. The results showed that the concentration of ARGs/MGEs varied as the process progressed, and which in the aeration tank was relatively high. The ARGs/MGEs content in SWP aerosol (8.35-163.27 copies/m3) was higher than that in SDP (5.52-16.36 copies/m3). The main ARGs/MGEs detected in SWP aerosol were tnpA-05, tnpA-04, and ermF, while the main ARGs/MGEs detected in SDP aerosol were sul1, ermF, and blaPAO. ARGs were positively correlated with most bacteria and Escherichia coli with ARGs carries higher cytotoxicity. ARGs/MGEs mainly diffused towards the southeast, which may cause harm to urban residents with the diffusion of aerosols. This study provides clues and theoretical basis for preventing the hazards of ARGs from WWTP sources.
Collapse
Affiliation(s)
- Yanjie Wang
- School of Public Health, Zhengzhou University, Zhengzhou 450001, PR China; Lancaster Environment Centre, Lancaster University, United Kingdom, UK; State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China.
| | - Kai Yang
- School of Public Health, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Lin Li
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China.
| | - Liying Yang
- School of Public Health, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Song Zhang
- School of Public Health, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Fangfang Yu
- School of Public Health, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Linlin Hua
- School of Public Health, Zhengzhou University, Zhengzhou 450001, PR China; Advanced Medical Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, PR China.
| |
Collapse
|
7
|
Araújo D, Silva AR, Fernandes R, Serra P, Barros MM, Campos AM, Oliveira R, Silva S, Almeida C, Castro J. Emerging Approaches for Mitigating Biofilm-Formation-Associated Infections in Farm, Wild, and Companion Animals. Pathogens 2024; 13:320. [PMID: 38668275 PMCID: PMC11054384 DOI: 10.3390/pathogens13040320] [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: 03/15/2024] [Revised: 04/05/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024] Open
Abstract
The importance of addressing the problem of biofilms in farm, wild, and companion animals lies in their pervasive impact on animal health and welfare. Biofilms, as resilient communities of microorganisms, pose a persistent challenge in causing infections and complicating treatment strategies. Recognizing and understanding the importance of mitigating biofilm formation is critical to ensuring the welfare of animals in a variety of settings, from farms to the wild and companion animals. Effectively addressing this issue not only improves the overall health of individual animals, but also contributes to the broader goals of sustainable agriculture, wildlife conservation, and responsible pet ownership. This review examines the current understanding of biofilm formation in animal diseases and elucidates the complex processes involved. Recognizing the limitations of traditional antibiotic treatments, mechanisms of resistance associated with biofilms are explored. The focus is on alternative therapeutic strategies to control biofilm, with illuminating case studies providing valuable context and practical insights. In conclusion, the review highlights the importance of exploring emerging approaches to mitigate biofilm formation in animals. It consolidates existing knowledge, highlights gaps in understanding, and encourages further research to address this critical facet of animal health. The comprehensive perspective provided by this review serves as a foundation for future investigations and interventions to improve the management of biofilm-associated infections in diverse animal populations.
Collapse
Affiliation(s)
- Daniela Araújo
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
| | - Ana Rita Silva
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Rúben Fernandes
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Patrícia Serra
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Maria Margarida Barros
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CECAV—Veterinary and Animal Research Centre, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Ana Maria Campos
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Ricardo Oliveira
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sónia Silva
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
| | - Carina Almeida
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Joana Castro
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
8
|
Perez-Bou L, Gonzalez-Martinez A, Gonzalez-Lopez J, Correa-Galeote D. Promising bioprocesses for the efficient removal of antibiotics and antibiotic-resistance genes from urban and hospital wastewaters: Potentialities of aerobic granular systems. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 342:123115. [PMID: 38086508 DOI: 10.1016/j.envpol.2023.123115] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 11/07/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023]
Abstract
The use, overuse, and improper use of antibiotics have resulted in higher levels of antibiotic-resistant bacteria (ARB) and antibiotic-resistance genes (ARGs), which have profoundly disturbed the equilibrium of the environment. Furthermore, once antibiotic agents are excreted in urine and feces, these substances often can reach wastewater treatment plants (WWTPs), in which improper treatments have been highlighted as the main reason for stronger dissemination of antibiotics, ARB, and ARGs to the receiving bodies. Hence, achieving better antibiotic removal capacities in WWTPs is proposed as an adequate approach to limit the spread of antibiotics, ARB, and ARGs into the environment. In this review, we highlight hospital wastewater (WW) as a critical hotspot for the dissemination of antibiotic resistance due to its high level of antibiotics and pathogens. Hence, monitoring the composition and structure of the bacterial communities related to hospital WW is a key factor in controlling the spread of ARGs. In addition, we discuss the advantages and drawbacks of the current biological WW treatments regarding the antibiotic-resistance phenomenon. Widely used conventional activated sludge technology has proved to be ineffective in mitigating the dissemination of ARB and ARGs to the environment. However, aerobic granular sludge (AGS) technology is a promising technology-with broad adaptability and excellent performance-that could successfully reduce antibiotics, ARB, and ARGs in the generated effluents. We also outline the main operational parameters involved in mitigating antibiotics, ARB, and ARGs in WWTPs. In this regard, WW operation under long hydraulic and solid retention times allows better removal of antibiotics, ARB, and ARGs independently of the WW technology employed. Finally, we address the current knowledge of the adsorption and degradation of antibiotics and their importance in removing ARB and ARGs. Notably, AGS can enhance the removal of antibiotics, ARB, and ARGs due to the complex microbial metabolism within the granular biomass.
Collapse
Affiliation(s)
- Lizandra Perez-Bou
- Microbiology Department, Faculty of Pharmacy, University of Granada, Granada, Andalucía, Spain; Microbiology and Environmental Technology Section, Institute of Water Research, University of Granada, Granada, Andalucía, Spain; Microbial Biotechnology Group, Microbiology and Virology Department, Faculty of Biology, University of Havana, Cuba
| | - Alejandro Gonzalez-Martinez
- Microbiology Department, Faculty of Pharmacy, University of Granada, Granada, Andalucía, Spain; Microbiology and Environmental Technology Section, Institute of Water Research, University of Granada, Granada, Andalucía, Spain
| | - Jesus Gonzalez-Lopez
- Microbiology Department, Faculty of Pharmacy, University of Granada, Granada, Andalucía, Spain; Microbiology and Environmental Technology Section, Institute of Water Research, University of Granada, Granada, Andalucía, Spain
| | - David Correa-Galeote
- Microbiology Department, Faculty of Pharmacy, University of Granada, Granada, Andalucía, Spain; Microbiology and Environmental Technology Section, Institute of Water Research, University of Granada, Granada, Andalucía, Spain.
| |
Collapse
|
9
|
Hwang J, Barman S, Gao R, Yang X, O'Malley A, Nagarkatti P, Nagarkatti M, Chruszcz M, Tang C. Membrane-Active Metallopolymers: Repurposing and Rehabilitating Antibiotics to Gram-Negative Superbugs. Adv Healthc Mater 2023; 12:e2301764. [PMID: 37565371 PMCID: PMC10842942 DOI: 10.1002/adhm.202301764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/03/2023] [Indexed: 08/12/2023]
Abstract
Among multiple approaches to combating antimicrobial resistance, a combination therapy of existing antibiotics with bacterial membrane-perturbing agents is promising. A viable platform of metallopolymers as adjuvants in combination with traditional antibiotics is reported in this work to combat both planktonic and stationary cells of Gram-negative superbugs and their biofilms. Antibacterial efficacy, toxicity, antibiofilm activity, bacterial resistance propensity, and mechanisms of action of metallopolymer-antibiotic combinations are investigated. These metallopolymers exhibit 4-16-fold potentiation of antibiotics against Gram-negative bacteria with negligible toxicity toward mammalian cells. More importantly, the lead combinations (polymer-ceftazidime and polymer-rifampicin) eradicate preformed biofilms of MDR E. coli and P. aeruginosa, respectively. Further, β-lactamase inhibition, outer membrane permeabilization, and membrane depolarization demonstrate synergy of these adjuvants with different antibiotics. Moreover, the membrane-active metallopolymers enable the antibiotics to circumvent bacterial resistance development. Altogether, the results indicate that such non-antibiotic adjuvants bear the promise to revitalize the efficacy of existing antibiotics to tackle Gram-negative bacterial infections.
Collapse
Affiliation(s)
- JiHyeon Hwang
- Department of Chemistry and BiochemistryUniversity of South CarolinaColumbiaSC29208USA
| | - Swagatam Barman
- Department of Chemistry and BiochemistryUniversity of South CarolinaColumbiaSC29208USA
| | - Ruixuan Gao
- Department of ChemistryUniversity of South FloridaTampaFL33620USA
| | - Xiaoming Yang
- Department of PathologyMicrobiology and ImmunologyUniversity of South CarolinaSchool of MedicineColumbiaSC29209USA
| | - Andrea O'Malley
- Department of Chemistry and BiochemistryUniversity of South CarolinaColumbiaSC29208USA
- Department of Biochemistry and Molecular BiologyMichigan State UniversityEast LansingMI48824USA
| | - Prakash Nagarkatti
- Department of PathologyMicrobiology and ImmunologyUniversity of South CarolinaSchool of MedicineColumbiaSC29209USA
| | - Mitzi Nagarkatti
- Department of PathologyMicrobiology and ImmunologyUniversity of South CarolinaSchool of MedicineColumbiaSC29209USA
| | - Maksymilian Chruszcz
- Department of Chemistry and BiochemistryUniversity of South CarolinaColumbiaSC29208USA
- Department of Biochemistry and Molecular BiologyMichigan State UniversityEast LansingMI48824USA
| | - Chuanbing Tang
- Department of Chemistry and BiochemistryUniversity of South CarolinaColumbiaSC29208USA
| |
Collapse
|
10
|
Petit M, Tessier J, Sahli C, Schmitzer AR. Confronting the Threat: Designing Highly Effective bis-Benzimidazolium Agents to Overcome Biofilm Persistence and Antimicrobial Resistance. ACS Infect Dis 2023; 9:2202-2214. [PMID: 37882623 DOI: 10.1021/acsinfecdis.3c00289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
The objective of this study is to take the initial steps toward developing novel antibiotics to counteract the escalating problem of antimicrobial and bacterial persistence, particularly in relation to biofilms. Our approach involves emulating the structural characteristics of cationic antimicrobial peptides. To circumvent resistance development, we have designed a library of bis-benzimidazolium salts that selectively target the microbial membranes in a nonspecific manner. To explore their structure-activity relationship, we conducted experiments using these compounds on various pathogens known for their resistance to conventional antibiotics, including Gram-positive methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus faecium (VRE), and Gram-negative Escherichia coli (E. coli). Notably, two bis-benzimidazolium salts exhibited robust antimicrobial activity while maintaining a high level of selectivity compared with mammalian cells. Our investigations revealed significant antibiofilm activity, as these compounds rapidly acted against established biofilms. In addition, bis-benzimidazolium compounds exhibited consistent results in resistance development and cross-resistance studies. Consequently, amphiphilic bis-benzimidazolium salts hold promise as potential candidates to combat resistance-associated infections.
Collapse
Affiliation(s)
- Maude Petit
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
| | - Jérémie Tessier
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
- Collège Bois-de-Boulogne, 10555 Ave. de Bois-de-Boulogne, Montréal H4N 1L4, Canada
| | - Célia Sahli
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
- CNRS-UMR 7086, Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), Université Paris Cité, Paris 75013 , France
| | - Andreea R Schmitzer
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
| |
Collapse
|
11
|
Moradi F, Ghaedi A, Fooladfar Z, Bazrgar A. Recent advance on nanoparticles or nanomaterials with anti-multidrug resistant bacteria and anti-bacterial biofilm properties: A systematic review. Heliyon 2023; 9:e22105. [PMID: 38034786 PMCID: PMC10685370 DOI: 10.1016/j.heliyon.2023.e22105] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/05/2023] [Accepted: 11/04/2023] [Indexed: 12/02/2023] Open
Abstract
Objective With the wide spread of Multidrug-resistant bacteria (MDR) due to the transfer and acquisition of antibiotic resistance genes and the formation of microbial biofilm, various researchers around the world are looking for a solution to overcome these resistances. One potential strategy and the best candidate to overcome these infections is using an effective nanomaterial with antibacterial properties against them. Methods and analysis: In this study, we overview nanomaterials with anti-MDR bacteria and anti-biofilm properties. Hence, we systematically explored biomedical databases (Web of Sciences, Google Scholar, PubMed, and Scopus) to categorize related studies about nanomaterial with anti-MDR bacteria and anti-biofilm activities from 2007 to December 2022. Results In total, forty-one studies were investigated to find antibacterial and anti-biofilm information about the nanomaterial during 2007-2022. According to the collected documents, nineteen types of nanomaterial showed putative antibacterial effects such as Cu, Ag, Au, Au/Pt, TiO2, Al2O3, ZnO, Se, CuO, Cu/Ni, Cu/Zn, Fe3O4, Au/Fe3O4, Au/Ag, Au/Pt, Graphene O, and CuS. In addition, seven types of them considered as anti-biofilm agents such as Ag, ZnO, Au/Ag, Graphene O, Cu, Fe3O4, and Au/Ag. Conclusion According to the studies, each of nanomaterial has been designed with different methods and their effects against standard strains, clinical strains, MDR strains, and bacterial biofilms have been investigated in-vitro and in-vivo conditions. In addition, nanomaterials have different destructive mechanism on bacterial structures. Various nanoparticles (NP) introduced as the best candidate to designing new drug and medical equipment preventing infectious disease outbreaks by overcome antibiotic resistance and bacterial biofilm.
Collapse
Affiliation(s)
- Farhad Moradi
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arshin Ghaedi
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Fooladfar
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Bazrgar
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Kumar G, Engle K. Natural products acting against S. aureus through membrane and cell wall disruption. Nat Prod Rep 2023; 40:1608-1646. [PMID: 37326041 DOI: 10.1039/d2np00084a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Covering: 2015 to 2022Staphylococcus aureus (S. aureus) is responsible for several community and hospital-acquired infections with life-threatening complications such as bacteraemia, endocarditis, meningitis, liver abscess, and spinal cord epidural abscess. In recent decades, the abuse and misuse of antibiotics in humans, animals, plants, and fungi and the treatment of nonmicrobial diseases have led to the rapid emergence of multidrug-resistant pathogens. The bacterial wall is a complex structure consisting of the cell membrane, peptidoglycan cell wall, and various associated polymers. The enzymes involved in bacterial cell wall synthesis are established antibiotic targets and continue to be a central focus for antibiotic development. Natural products play a vital role in drug discovery and development. Importantly, natural products provide a starting point for active/lead compounds that sometimes need modification based on structural and biological properties to meet the drug criteria. Notably, microorganisms and plant metabolites have contributed as antibiotics for noninfectious diseases. In this study, we have summarized the recent advances in understanding the activity of the drugs or agents of natural origin that directly inhibit the bacterial membrane, membrane components, and membrane biosynthetic enzymes by targeting membrane-embedded proteins. We also discussed the unique aspects of the active mechanisms of established antibiotics or new agents.
Collapse
Affiliation(s)
- Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India.
| | - Kritika Engle
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, 500037, India.
| |
Collapse
|
13
|
Sivadas N, Kaul G, Akhir A, Shukla M, Govind MG, Dan M, Radhakrishnan KV, Chopra S. Naturally Derived Malabaricone B as a Promising Bactericidal Candidate Targeting Multidrug-Resistant Staphylococcus aureus also Possess Synergistic Interactions with Clinical Antibiotics. Antibiotics (Basel) 2023; 12:1483. [PMID: 37887184 PMCID: PMC10604362 DOI: 10.3390/antibiotics12101483] [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: 08/22/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/28/2023] Open
Abstract
The emergence of multidrug-resistant (MDR) superbugs underlines the urgent need for innovative treatment options to tackle resistant bacterial infections. The clinical efficacy of natural products directed our efforts towards developing new antibacterial leads from naturally abundant known chemical structures. The present study aimed to explore an unusual class of phenylacylphenols (malabaricones) from Myristicamalabarica as antibacterial agents. In vitro antibacterial activity was determined via broth microdilution, cell viability, time-kill kinetics, biofilm eradication, intracellular killing, and checkerboard assays. The efficacy was evaluated in vivo in murine neutropenic thigh and skin infection models. Confocal and SEM analyses were used for mechanistic studies. Among the tested isolates, malabaricone B (NS-7) demonstrated the best activity against S. aureus with a favorable selectivity index and concentration-dependent, rapid bactericidal killing kinetics. It displayed equal efficacy against MDR clinical isolates of S. aureus and Enterococci, efficiently clearing S. aureus in intracellular and biofilm tests, with no detectable resistance. In addition, NS-7 synergized with daptomycin and gentamicin. In vivo, NS-7 exhibited significant efficacy against S. aureus infection. Mechanistically, NS-7 damaged S. aureus membrane integrity, resulting in the release of extracellular ATP. The results indicated that NS-7 can act as a naturally derived bactericidal drug lead for anti-staphylococcal therapy.
Collapse
Affiliation(s)
- Neethu Sivadas
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019, India;
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
| | - Grace Kaul
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
- Division of Microbiology, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; (A.A.)
| | - Abdul Akhir
- Division of Microbiology, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; (A.A.)
| | - Manjulika Shukla
- Division of Microbiology, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; (A.A.)
| | - Murugan Govindakurup Govind
- Department of Plant Genetics Resource, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode, Thiruvananthapuram 695562, India
| | - Mathew Dan
- Department of Plant Genetics Resource, Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode, Thiruvananthapuram 695562, India
| | - Kokkuvayil Vasu Radhakrishnan
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019, India;
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
| | - Sidharth Chopra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India;
- Division of Microbiology, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; (A.A.)
| |
Collapse
|
14
|
Guliy OI, Zaitsev BD, Borodina IA. Electroacoustic Biosensor Systems for Evaluating Antibiotic Action on Microbial Cells. SENSORS (BASEL, SWITZERLAND) 2023; 23:6292. [PMID: 37514587 PMCID: PMC10383298 DOI: 10.3390/s23146292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023]
Abstract
Antibiotics are widely used to treat infectious diseases. This leads to the presence of antibiotics and their metabolic products in the ecosystem, especially in aquatic environments. In many countries, the growth of pathogen resistance to antibiotics is considered a threat to national security. Therefore, methods for determining the sensitivity/resistance of bacteria to antimicrobial drugs are important. This review discusses the mechanisms of the formation of antibacterial resistance and the various methods and sensor systems available for analyzing antibiotic effects on bacteria. Particular attention is paid to acoustic biosensors with active immobilized layers and to sensors that analyze antibiotics directly in liquids. It is shown that sensors of the second type allow analysis to be done within a short period, which is important for timely treatment.
Collapse
Affiliation(s)
- Olga I Guliy
- Institute of Biochemistry and Physiology of Plants and Microorganisms-Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Saratov 410049, Russia
| | - Boris D Zaitsev
- Kotelnikov Institute of Radio Engineering and Electronics, Russian Academy of Sciences, Saratov Branch, Saratov 410019, Russia
| | - Irina A Borodina
- Kotelnikov Institute of Radio Engineering and Electronics, Russian Academy of Sciences, Saratov Branch, Saratov 410019, Russia
| |
Collapse
|
15
|
Petersson M, Thrane SW, Gram L, Muyldermans S, Laustsen AH. Orally delivered single-domain antibodies against gastrointestinal pathogens. Trends Biotechnol 2023; 41:875-886. [PMID: 36774206 DOI: 10.1016/j.tibtech.2023.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 02/11/2023]
Abstract
Single-domain antibodies (sdAbs) are exceptionally stable fragments derived from the antigen-binding domains of immunoglobulins. They can withstand extreme pH, high temperature, and proteolysis, making them suitable for controlling gastrointestinal (GI) infections in humans and animals. sdAbs may function in their native soluble form, although different derived protein formats and the use of delivery vehicles can be useful for improved oral delivery. We discuss selected examples of the use of orally delivered sdAbs for protecting humans and animals against GI infections caused by pathogenic bacteria, viruses, and parasites. We finally provide perspectives on how sdAbs may be applied industrially and what challenges should be overcome for orally delivered sdAbs to reach the market.
Collapse
Affiliation(s)
- Marcus Petersson
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark; Bactolife A/S, Copenhagen East, Denmark
| | | | - Lone Gram
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Serge Muyldermans
- Department of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark; Bactolife A/S, Copenhagen East, Denmark.
| |
Collapse
|
16
|
Whelan AO, Cooper I, Ooi N, Orr D, Blades K, Kirkham J, Lyons A, Barnes KB, Richards MI, Salisbury AM, Craighead M, Harding SV. In Vitro Activity of Novel Topoisomerase Inhibitors against Francisella tularensis and Burkholderia pseudomallei. Antibiotics (Basel) 2023; 12:983. [PMID: 37370302 DOI: 10.3390/antibiotics12060983] [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: 04/04/2023] [Revised: 05/03/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Antimicrobial resistance is a global issue, and the investigation of alternative therapies that are not traditional antibiotics are warranted. Novel bacterial type II topoisomerase inhibitors (NBTIs) have recently emerged as a novel class of antibiotics with reduced potential for cross-resistance to fluoroquinolones due to their novel mechanism of action. This study investigated the in vitro activity of a series of cyclohexyl-oxazolidinone bacterial topoisomerase inhibitors against type strains of Francisella tularensis and Burkholderia pseudomallei. Broth microdilution, time-kill, and cell infection assays were performed to determine activity against these biothreat pathogens. Two candidates were identified that demonstrated in vitro activity in multiple assays that in some instances was equivalent to ciprofloxacin and doxycycline. These data warrant the further evaluation of these novel NBTIs and future iterations in vitro and in vivo.
Collapse
Affiliation(s)
- Adam O Whelan
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK
| | - Ian Cooper
- Infex Therapeutics Ltd., Mereside, Alderley Park, Macclesfield SK10 4TG, UK
| | - Nicola Ooi
- Infex Therapeutics Ltd., Mereside, Alderley Park, Macclesfield SK10 4TG, UK
| | - David Orr
- Infex Therapeutics Ltd., Mereside, Alderley Park, Macclesfield SK10 4TG, UK
| | - Kevin Blades
- Infex Therapeutics Ltd., Mereside, Alderley Park, Macclesfield SK10 4TG, UK
| | - James Kirkham
- Infex Therapeutics Ltd., Mereside, Alderley Park, Macclesfield SK10 4TG, UK
| | - Amanda Lyons
- Redx Anti-Infectives Ltd., Alderley Park, Macclesfield SK10 4TG, UK
| | - Kay B Barnes
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK
| | - Mark I Richards
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK
| | | | - Mark Craighead
- Redx Anti-Infectives Ltd., Alderley Park, Macclesfield SK10 4TG, UK
| | - Sarah V Harding
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK
- School of Respiratory Sciences, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
17
|
Kumawat M, Nabi B, Daswani M, Viquar I, Pal N, Sharma P, Tiwari S, Sarma DK, Shubham S, Kumar M. Role of bacterial efflux pump proteins in antibiotic resistance across microbial species. Microb Pathog 2023:106182. [PMID: 37263448 DOI: 10.1016/j.micpath.2023.106182] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/03/2023]
Abstract
Efflux proteins are transporter molecules that actively pump out a variety of substrates, including antibiotics, from cells to the environment. They are found in both Gram-positive and Gram-negative bacteria and eukaryotic cells. Based on their protein sequence homology, energy source, and overall structure, efflux proteins can be divided into seven groups. Multidrug efflux pumps are transmembrane proteins produced by microbes to enhance their survival in harsh environments and contribute to antibiotic resistance. These pumps are present in all bacterial genomes studied, indicating their ancestral origins. Many bacterial genes encoding efflux pumps are involved in transport, a significant contributor to antibiotic resistance in microbes. Efflux pumps are widely implicated in the extrusion of clinically relevant antibiotics from cells to the extracellular environment and, as such, represent a significant challenge to antimicrobial therapy. This review aims to provide an overview of the structures and mechanisms of action, substrate profiles, regulation, and possible inhibition of clinically relevant efflux pumps. Additionally, recent advances in research and the pharmacological exploitation of efflux pump inhibitors as a promising intervention for combating drug resistance will be discussed.
Collapse
Affiliation(s)
- Manoj Kumawat
- Department of Microbiology, ICMR- National Institute for Research in Environmental Health, Bhopal, 462030, India
| | - Bilkees Nabi
- Department of Biochemistry & Biochemical Engineering, SHUATS, Allahabad, 211007, India
| | - Muskan Daswani
- Department of Biotechnology, SantHirdaram Girls College, Bhopal, 462030, India
| | - Iqra Viquar
- Department of Biotechnology, SantHirdaram Girls College, Bhopal, 462030, India
| | - Namrata Pal
- Department of Microbiology, ICMR- National Institute for Research in Environmental Health, Bhopal, 462030, India
| | - Poonam Sharma
- Department of Microbiology, ICMR- National Institute for Research in Environmental Health, Bhopal, 462030, India
| | - Shikha Tiwari
- Department of Microbiology, ICMR- National Institute for Research in Environmental Health, Bhopal, 462030, India
| | - Devojit Kumar Sarma
- Department of Microbiology, ICMR- National Institute for Research in Environmental Health, Bhopal, 462030, India
| | - Swasti Shubham
- Department of Microbiology, ICMR- National Institute for Research in Environmental Health, Bhopal, 462030, India
| | - Manoj Kumar
- Department of Microbiology, ICMR- National Institute for Research in Environmental Health, Bhopal, 462030, India.
| |
Collapse
|
18
|
Varela MF, Stephen J, Bharti D, Lekshmi M, Kumar S. Inhibition of Multidrug Efflux Pumps Belonging to the Major Facilitator Superfamily in Bacterial Pathogens. Biomedicines 2023; 11:1448. [PMID: 37239119 PMCID: PMC10216197 DOI: 10.3390/biomedicines11051448] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Bacterial pathogens resistant to multiple structurally distinct antimicrobial agents are causative agents of infectious disease, and they thus constitute a serious concern for public health. Of the various bacterial mechanisms for antimicrobial resistance, active efflux is a well-known system that extrudes clinically relevant antimicrobial agents, rendering specific pathogens recalcitrant to the growth-inhibitory effects of multiple drugs. In particular, multidrug efflux pump members of the major facilitator superfamily constitute central resistance systems in bacterial pathogens. This review article addresses the recent efforts to modulate these antimicrobial efflux transporters from a molecular perspective. Such investigations can potentially restore the clinical efficacy of infectious disease chemotherapy.
Collapse
Affiliation(s)
- Manuel F. Varela
- Department of Biology, Eastern New Mexico University, Station 33, Portales, NM 88130, USA
| | - Jerusha Stephen
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India; (J.S.); (D.B.); (M.L.); (S.K.)
| | - Deeksha Bharti
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India; (J.S.); (D.B.); (M.L.); (S.K.)
| | - Manjusha Lekshmi
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India; (J.S.); (D.B.); (M.L.); (S.K.)
| | - Sanath Kumar
- ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India; (J.S.); (D.B.); (M.L.); (S.K.)
| |
Collapse
|
19
|
Wang L, Li J, Zhao J, Li H, Feng J, Zhang P, Pan B. Photodegradation of clindamycin by the dissolved black carbon is simultaneously regulated by ROS generation and the binding effect. WATER RESEARCH 2023; 233:119784. [PMID: 36863283 DOI: 10.1016/j.watres.2023.119784] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/26/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
As an essential source of the natural dissolved organic matter (DOM), dissolved black carbon (DBC) plays a vital role in the photodegradation of organics; however, there is rare information about the DBC-induced photodegradation mechanism of clindamycin (CLM), one of the widely used antibiotics. Herein, we discovered DBC-generated reactive oxygen species (ROS) stimulated CLM photodegradation. Hydroxy radical (•OH) could directly attack CLM by OH-addition reaction, the singlet oxygen (1O2) and superoxide (O2•-) contributed to the CLM degradation by transforming to •OH. In addition, the binding between CLM and DBCs inhibited the photodegradation of CLM by decreasing the concentration of freely dissolved CLM. Binding process inhibited CLM photodegradation by 0.25-1.98% at pH 7.0 and 6.1-41.77% at pH 8.5. These findings suggest that the photodegradation of CLM by DBC is simultaneously regulated by the ROS production and binding effect between CLM and DBC, benefiting the exact evaluation of the environmental impact of DBCs.
Collapse
Affiliation(s)
- Lin Wang
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, 650500, China
| | - Jing Li
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, 650500, China
| | - Jing Zhao
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, 650500, China
| | - Hao Li
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, 650500, China
| | - Jing Feng
- Faculty of Material Science and Engineering, Kunming University of Science and Technology, Kunming,650500, China.
| | - Peng Zhang
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, 650500, China.
| | - Bo Pan
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, 650500, China
| |
Collapse
|
20
|
Dhiman S, Ramirez D, Li Y, Kumar A, Arthur G, Schweizer F. Chimeric Tobramycin-Based Adjuvant TOB-TOB-CIP Potentiates Fluoroquinolone and β-Lactam Antibiotics against Multidrug-Resistant Pseudomonas aeruginosa. ACS Infect Dis 2023; 9:864-885. [PMID: 36917096 DOI: 10.1021/acsinfecdis.2c00549] [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] [Indexed: 03/15/2023]
Abstract
According to the World Health Organization, antibiotic resistance is a global health threat. Of particular importance are infections caused by multidrug-resistant Gram-negative bacteria including Escherichia coli, Acinetobacter baumannii, Klebsiella pneumoniae, and Pseudomonas aeruginosa for which limited treatment options exist. Multiple and simultaneously occurring resistance mechanisms including outer membrane impermeability, overexpression of efflux pumps, antibiotic-modifying enzymes, and modification of genes and antibiotic targets have made antibiotic drug development more difficult against these pathogens. One strategy to cope with these challenges is the use of outer membrane permeabilizers that increase the intracellular concentration of antibiotics when used in combination. In some circumstances, this approach can rescue antibiotics from resistance or repurpose currently marketed antibiotics. Tobramycin-based hybrid antibiotic adjuvants that combine two outer membrane-active components have been previously shown to potentiate antibiotics by facilitating transit through the outer membrane, resulting in increased antibiotic accumulation within the cell. Herein, we extended the concept of tobramycin-based hybrid antibiotic adjuvants to tobramycin-based chimeras by engineering up to three different membrane-active antibiotic warheads such as tobramycin, 1-(1-naphthylmethyl)-piperazine, ciprofloxacin, and cyclam into a central 1,3,5-triazine scaffold. Chimera 4 (TOB-TOB-CIP) consistently synergized with ciprofloxacin, levofloxacin, and moxifloxacin against wild-type and fluoroquinolone-resistant P. aeruginosa. Moreover, the susceptibility breakpoints of ceftazidime, aztreonam, and imipenem were reached using the triple combination of chimera 4 with ceftazidime/avibactam, aztreonam/avibactam, and imipenem/relebactam, respectively, against β-lactamase-harboring P. aeruginosa. Our findings demonstrate that tobramycin-based chimeras form a novel class of antibiotic potentiators capable of restoring the activity of antibiotics against P. aeruginosa.
Collapse
Affiliation(s)
- Shiv Dhiman
- Department of Chemistry, University of Manitoba, Winnipeg R3T 2N2, Manitoba, Canada
| | - Danyel Ramirez
- Department of Chemistry, University of Manitoba, Winnipeg R3T 2N2, Manitoba, Canada
| | - Yanqi Li
- Department of Microbiology, University of Manitoba, Winnipeg R3T 2N2, Manitoba, Canada
| | - Ayush Kumar
- Department of Microbiology, University of Manitoba, Winnipeg R3T 2N2, Manitoba, Canada
| | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg R3E 0J9, Manitoba, Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba, Winnipeg R3T 2N2, Manitoba, Canada
| |
Collapse
|
21
|
Engin AB, Engin ED, Engin A. Effects of co-selection of antibiotic-resistance and metal-resistance genes on antibiotic-resistance potency of environmental bacteria and related ecological risk factors. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 98:104081. [PMID: 36805463 DOI: 10.1016/j.etap.2023.104081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/23/2023] [Accepted: 02/12/2023] [Indexed: 06/18/2023]
Abstract
The inadequate elimination of micropollutants in wastewater treatment plants (WWTP), cause to increase in the incidence of antibiotic resistant bacterial strains. Growth of microbial pathogens in WWTP is one of the serious public health problems. The widespread and simultaneous emergence of antibiotic resistance genes (ARGs) and heavy metal resistance genes (HMRGs) in the environment with heavy metals create persistent and selective pressure for co-selection of both genes on environmental microorganisms. Co-localization of ARGs and HMRGs on the same horizontal mobile genetic elements (MGEs) allows the spreading of numerous antibiotic-resistant strains of bacteria in aquatic and terrestrial environment. The biofilm formation and colonization potential of environmental bacteria leads to the co-selection of multi-antibiotic resistance and multi-metal tolerance. Horizontal gene transfer (HGT), co-localization of both ARGs and HMRGs on the same MGEs, and the shared resistomes are important bacteria-associated ecological risks factors, which reduce the effectiveness of antibiotics against bacterial infections.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, Ankara, Turkey.
| | - Evren Doruk Engin
- Ankara University, Biotechnology Institute, Gumusdere Campus, Kecioren, Ankara, Turkey
| | - Atilla Engin
- Gazi University, Faculty of Medicine, Department of General Surgery, Ankara, Turkey
| |
Collapse
|
22
|
Highly sensitive and selective detection of enrofloxacin residues in chicken based on solution-gated graphene field-effect transistors. J Electroanal Chem (Lausanne) 2023. [DOI: 10.1016/j.jelechem.2023.117325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
|
23
|
Nanomaterials and Coatings for Managing Antibiotic-Resistant Biofilms. Antibiotics (Basel) 2023; 12:antibiotics12020310. [PMID: 36830221 PMCID: PMC9952333 DOI: 10.3390/antibiotics12020310] [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: 01/15/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Biofilms are a global health concern responsible for 65 to 80% of the total number of acute and persistent nosocomial infections, which lead to prolonged hospitalization and a huge economic burden to the healthcare systems. Biofilms are organized assemblages of surface-bound cells, which are enclosed in a self-produced extracellular polymer matrix (EPM) of polysaccharides, nucleic acids, lipids, and proteins. The EPM holds the pathogens together and provides a functional environment, enabling adhesion to living and non-living surfaces, mechanical stability, next to enhanced tolerance to host immune responses and conventional antibiotics compared to free-floating cells. Furthermore, the close proximity of cells in biofilms facilitates the horizontal transfer of genes, which is responsible for the development of antibiotic resistance. Given the growing number and impact of resistant bacteria, there is an urgent need to design novel strategies in order to outsmart bacterial evolutionary mechanisms. Antibiotic-free approaches that attenuate virulence through interruption of quorum sensing, prevent adhesion via EPM degradation, or kill pathogens by novel mechanisms that are less likely to cause resistance have gained considerable attention in the war against biofilm infections. Thereby, nanoformulation offers significant advantages due to the enhanced antibacterial efficacy and better penetration into the biofilm compared to bulk therapeutics of the same composition. This review highlights the latest developments in the field of nanoformulated quorum-quenching actives, antiadhesives, and bactericides, and their use as colloid suspensions and coatings on medical devices to reduce the incidence of biofilm-related infections.
Collapse
|
24
|
Xie G, Wang X, Mo M, Zhang L, Zhu J. Photothermal Hydrogels for Promoting Infected Wound Healing. Macromol Biosci 2023; 23:e2200378. [PMID: 36337010 DOI: 10.1002/mabi.202200378] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/28/2022] [Indexed: 11/09/2022]
Abstract
Photothermal therapies (PTT), with spatiotemporally controllable antibacterial capabilities without inducing resistance, have shown encouraging prospects in the field of infected wound treatments. As an important platform for PTT, photothermal hydrogels exhibit attractive advantages in the field of infected wound treatment due to their excellent biochemical properties and have been intensively explored in recent years. This review summarizes the progress of the photothermal hydrogels for promoting infected wound healing. Three major elements of photothermal hydrogels, i.e., photothermal materials, hydrogel matrix, and construction methods, are introduced. Furthermore, different strategies of photothermal hydrogels in the treatment of infected wounds are summarized. Finally, the challenges and prospects in the clinical treatment of photothermal hydrogels are discussed.
Collapse
Affiliation(s)
- Ge Xie
- 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
| | - Xiao Wang
- 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
| | - Min Mo
- 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
| | - Lianbin Zhang
- 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
|
25
|
Liu S, Liu J, Fu N, Kornmatitsuk B, Yan Z, Luo J. Phenylalanine-arginine β-naphthylamide could enhance neomycin-sensitivity on Riemerella anatipestifer in vitro and in vivo. Front Microbiol 2023; 13:985789. [PMID: 36713163 PMCID: PMC9873997 DOI: 10.3389/fmicb.2022.985789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
Riemerella anatipestifer is an important duck pathogen responsible for septicemia and infectious serositis, which has caused great economic losses to the duck industry. Phenylalanine-arginine β-naphthylamide (PAβN) is an efflux pump inhibitor, which mainly reduces the efflux effect by competing with antibiotics for efflux pump channels. Here, we found that R. anatipestifer strain GD2019 showed resistances to gentamicin, amikacin, kanamycin, and neomycin. Notably, PAβN could significantly reduce the Minimal inhibitory concentrations (MICs) of neomycin on the GD2019 strain. Moreover, PAβN combined with neomycin significantly decreased bacterial loads, relieved pathological injury and increase survival rate (p < 0.05) for the ducks lethally challenged by the GD2019 strain. Therefore, our results suggested, in vitro and in vivo, PAβN could reduce neomycin-resistant of R. anatipestifer. Importantly, finding of this study provide a new approach for treating antibiotic-resistant R. anatipestifer infection.
Collapse
Affiliation(s)
- Shiqi Liu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China,Jinzhai County Agriculture and Rural Bureau, Jinzhai, Anhui, China
| | - Junfa Liu
- Wen's Group Academy, Xinxing, Guangdong, China
| | - Ning Fu
- Chifeng Institute of Agricultural Sciences, Chifeng, China
| | - Bunlue Kornmatitsuk
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | | | - Junrong Luo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China,*Correspondence: Junrong Luo, ✉
| |
Collapse
|
26
|
Gribble GW. Naturally Occurring Organohalogen Compounds-A Comprehensive Review. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 2023; 121:1-546. [PMID: 37488466 DOI: 10.1007/978-3-031-26629-4_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
The present volume is the third in a trilogy that documents naturally occurring organohalogen compounds, bringing the total number-from fewer than 25 in 1968-to approximately 8000 compounds to date. Nearly all of these natural products contain chlorine or bromine, with a few containing iodine and, fewer still, fluorine. Produced by ubiquitous marine (algae, sponges, corals, bryozoa, nudibranchs, fungi, bacteria) and terrestrial organisms (plants, fungi, bacteria, insects, higher animals) and universal abiotic processes (volcanos, forest fires, geothermal events), organohalogens pervade the global ecosystem. Newly identified extraterrestrial sources are also documented. In addition to chemical structures, biological activity, biohalogenation, biodegradation, natural function, and future outlook are presented.
Collapse
Affiliation(s)
- Gordon W Gribble
- Department of Chemistry, Dartmouth College, Hanover, NH, 03755, USA.
| |
Collapse
|
27
|
Guo N, Liu M, Yang Z, Wu D, Chen F, Wang J, Zhu Z, Wang L. The synergistic mechanism of β-lactam antibiotic removal between ammonia-oxidizing microorganisms and heterotrophs. ENVIRONMENTAL RESEARCH 2023; 216:114419. [PMID: 36174754 DOI: 10.1016/j.envres.2022.114419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
Nitrifying system is an effective strategy to remove numerous antibiotics, however, the contribution of ammonia-oxidizing bacteria (AOB), ammonia-oxidizing archaea (AOA) and heterotrophs for antibiotic removal are still unclear. In this study, the mechanism of β-lactam antibiotic (cefalexin, CFX) removal was studied in a nitrifying sludge system. Results showed that CFX was synergistically removed by AOB (Nitrosomonas, played a major role) and AOA (Candidatus_Nitrososphaera) through ammonia monooxygenase-mediated co-metabolism, and by heterotrophs (Pseudofulvimonas, Hydrogenophaga, RB41, Thauera, UTCFX1, Plasticicumulans, Phaeodactylibacter) through antibiotic resistance genes (ARGs)-encoded β-lactamases-mediated hydrolysis. Regardless of increased archaeal and heterotrophic CFX removal with the upregulation of amoA in AOA and ARGs, the system exhibited poorer CFX removal performance at 10 mg/L, mainly due to the inhibition of AOB. This study provides new reference for the important roles of heterotrophs and ARGs, opening the possibilities for the application of ARGs in antibiotic biodegradation.
Collapse
Affiliation(s)
- Ning Guo
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, 250101, China; Resources and Environment Innovation Institute, Shandong Jianzhu University, Jinan, 250101, China
| | - Mengmeng Liu
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, 250101, China
| | - Zhuhui Yang
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, 250101, China
| | - Daoji Wu
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, 250101, China; Resources and Environment Innovation Institute, Shandong Jianzhu University, Jinan, 250101, China
| | - Feiyong Chen
- Resources and Environment Innovation Institute, Shandong Jianzhu University, Jinan, 250101, China
| | - Jinhe Wang
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, 250101, China; Resources and Environment Innovation Institute, Shandong Jianzhu University, Jinan, 250101, China
| | - Zhaoliang Zhu
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, 250101, China.
| | - Lin Wang
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan, 250101, China; Resources and Environment Innovation Institute, Shandong Jianzhu University, Jinan, 250101, China.
| |
Collapse
|
28
|
Raichur A, Sinha N. Synthesis of multi-layered nanoswabs for simultaneous and expeditious removal of antibiotic-resistant bacteria, dyes, and antibiotics from wastewater. Sep Purif Technol 2022. [DOI: 10.1016/j.seppur.2022.122830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
29
|
Obeng E, Feng J, Wang D, Zheng D, Xiang B, Shen J. Multifunctional phototheranostic agent ZnO@Ag for anti-infection through photothermal/photodynamic therapy. Front Chem 2022; 10:1054739. [PMID: 36438866 PMCID: PMC9682125 DOI: 10.3389/fchem.2022.1054739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/06/2022] [Indexed: 08/22/2023] Open
Abstract
To overcome the limitations of traditional therapeutics, nanotechnology offers a synergistic therapeutic approach for the treatment of bacterial infection and biofilms that has attracted attention. Herein, we report on a ZnO@Ag nanocomposite with good biocompatibility synthesized by doping ZnO NPs with silver nanoparticles (Ag NPs). ZnO@Ag nanocomposites were synthesized with varying ratios of Ag NPs (0.5%, 2%, 8%). Under the same experimental conditions, ZnO@8%Ag exhibited outstanding properties compared to the other nanocomposites and the pristine ZnO NPs. ZnO@8%Ag demonstrated excellent photothermal and photodynamic properties. Also, ZnO@8%Ag demonstrated over 99% inhibition of Staphylococcus aureus (S. aureus) under photothermal therapy (PTT) or photodynamics therapy (PDT) as a result of the excessive generation of reactive oxygen species (ROS) by the Ag+ released, while the pristine ZnO showed an insignificant inhibition rate compared to the PBS group (control). Furthermore, ZnO@8%Ag completely disrupted S. aureus biofilm under a combined PTT/PDT treatment, a synergetic trimodal therapy, although the molecular mechanism of biofilm inhibition remains unclear. Hence, the excellent photothermal, photodynamic, biocompatibility, and bactericidal properties of ZnO@8%Ag present it as an appropriate platform for bacterial and biofilm treatment or other biomedically related applications.
Collapse
Affiliation(s)
- Enoch Obeng
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiayao Feng
- Ningbo Eye Hospital, Ningbo, Zhejiang, China
| | - Danyan Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Dongyang Zheng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Bailin Xiang
- College of Chemistry and Materials Engineering, Huaihua University, Huaihua, China
| | - Jianliang Shen
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| |
Collapse
|
30
|
Li X, Wang Y, Jiang X, Zeng Y, Zhao X, Washio J, Takahashi N, Zhang L. Investigation of drug resistance of caries-related streptococci to antimicrobial peptide GH12. Front Cell Infect Microbiol 2022; 12:991938. [PMID: 36159653 PMCID: PMC9492880 DOI: 10.3389/fcimb.2022.991938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Dental caries is associated with caries-related streptococci and antimicrobial agents have been widely used for caries control, but troubled by antibiotic resistance. This study aimed to investigate the intrinsic and acquired resistance of caries-related streptococci to antimicrobial peptide GH12, which was proven promising for caries control, and preliminarily explore the phenotypic changes and whole genome of stable acquired resistant strains. In this study, susceptibility assays and resistance assays were performed, followed by stability assays of resistance, to evaluate the intrinsic resistance and the potential resistance of caries-related streptococci. Then, the phenotypic changes of the stable acquired resistant strain were explored. The whole genome of the resistant strain was sequenced and analyzed by second-generation and third-generation high-throughput sequencing technologies. Streptococcus gordonii and Streptococcus sanguinis were intrinsically resistant to GH12 compared to cariogenic Streptococcus mutans. Acquired GH12 resistance in one S. sanguinis and four S. mutans clinical strains was transient but stable in one S. mutans strain (COCC33-14). However, acquired resistance to daptomycin (DAP) and chlorhexidine in all strains was stable. Furthermore, the COCC33-14 showed cross-resistance to DAP and delayed growth rates and a lower population. However, no drug-resistant gene mutation was detected in this strain, but 6 new and 5 missing genes were found. Among them, annotation of one new gene (gene 1782|COCC33-14R) is related to the integral component of the membrane, and one missing gene rpsN is associated with the metabolism and growth of bacteria. The results indicate that stable resistant mutants of caries-related streptococci could hardly be selected by exposure to consecutive sublethal GH12, but the risk still existed. Resistance in COCC33-14R is mainly related to changes in the cell envelope.
Collapse
Affiliation(s)
- Xinwei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Stomatology, Chengdu Second People’s Hospital, Chengdu, China
| | - Yufei Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuelian Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuhao Zeng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinran Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jumpei Washio
- Division of Oral Ecology and Biochemistry, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Nobuhiro Takahashi
- Division of Oral Ecology and Biochemistry, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Linglin Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Liu G, Lu D, Zhu S, Hao M, Yang X, Wang X, Zhang Y. A new self-immolative colistin prodrug with dual targeting functionalities and reduced toxicity for the treatment of intracellular bacterial infections. J Biomed Mater Res A 2022; 110:1590-1598. [PMID: 35593460 DOI: 10.1002/jbm.a.37410] [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: 01/07/2022] [Revised: 04/26/2022] [Accepted: 05/05/2022] [Indexed: 11/08/2022]
Abstract
Colistin is a potent antibiotic but its severe side effects including nephrotoxicity and neurotoxicity are the roadblock for their wide use in clinics. To solve this problem, we synthesized a new prodrug, mannose-maltose-colistin conjugate, termed MMCC that can reversibly mask the five amines of colistin that are primarily responsible for the toxicity. The deliberated design of disulfide-based self-immolative linker warranted the reversibly release of the pristine amines of colistin on demand without sacrificing antimicrobial efficacy. Once MMCC was delivered in cells, reducing agents cleaves the disulfide bond and release the pristine amines. The targeting ligands of maltose and mannose were grafted on colistin conjugate for targeting delivery of colistin to bacteria and macrophages, respectively. Taken together, MMCC as a new class of antimicrobial biomaterials, demonstrates its great potential for the treatment of intracellular bacterial infections.
Collapse
Affiliation(s)
- Gengqi Liu
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China
| | - Di Lu
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China
| | - Shiyu Zhu
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, P. R. China
| | - Minchao Hao
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China
| | - Xingyue Yang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China
| | - Xiaojian Wang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, P. R. China
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China
| |
Collapse
|
32
|
Zhang X, Gong Z, Allinson G, Xiao M, Li X, Jia C, Ni Z. Environmental risks caused by livestock and poultry farms to the soils: Comparison of swine, chicken, and cattle farms. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2022; 317:115320. [PMID: 35642811 DOI: 10.1016/j.jenvman.2022.115320] [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] [Received: 11/12/2021] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 06/15/2023]
Abstract
The lack of treatment systems for pollutants in family-livestock and poultry sites results in large amounts of untreated manure and urine being directly discharged to environment. The risks from veterinary antibiotic (VA) and heavy metal (HM) exposure in the rural environment require further research. In this investigation, 221 samples (feed, manure, surface soil, soil profiles, water, and plant) were collected from 41 livestock and poultry farms (swine, chichen, and cattle). Copper (Cu), zinc (Zn), oxytetracycline (OTC), and enrofloxacin (ENR) were frequently detected in the samples. Metals and VAs in sandy loam soils were more inclined to migrate to deep layers than those in loam soils. Copper and Zn in the polluted soils mainly existed in available forms, which facilitated their migration to deep soil layers. In this study, OTC was also observed to migrate more easily to deeper soil layers than ENR due to its relatively high pKa value. Eighteen antibiotic resistance genes (ARGs) and 5 metal resistance genes (MRGs) along with one mobile genetic element (MGE) occurred in the soils at 80 cm depth. Luteimonas, Clostridium_sensu_stricto_1, and Rhodanobacter were dominant genera detected in the soil samples from different sites, which might increase migration of ARGs or degradation of VAs. An ecological risk assessment suggested that VAs posed threats to the growth of Triticum aestivum L, Cucumis sativus L, and Brassiaca chinensis L. Remediation techniques including biochar/modified biochar, anaerobic digestion, and manure composting should be developed urgently for joint HM and VA pollution.
Collapse
Affiliation(s)
- Xiaorong Zhang
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zongqiang Gong
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, PR China.
| | - Graeme Allinson
- School of Science, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Mei Xiao
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xiaojun Li
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, PR China
| | - Chunyun Jia
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, PR China
| | - Zijun Ni
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
33
|
Yang G, Zhang Z, Liu K, Ji X, Fatehi P, Chen J. A cellulose nanofibril-reinforced hydrogel with robust mechanical, self-healing, pH-responsive and antibacterial characteristics for wound dressing applications. J Nanobiotechnology 2022; 20:312. [PMID: 35794620 PMCID: PMC9258071 DOI: 10.1186/s12951-022-01523-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/23/2022] [Indexed: 02/06/2023] Open
Abstract
Background Bacterial infection in wounds has become a major threat to human life and health. With the growth use of synthetic antibiotics and the elevated evolution of drug resistant bacteria in human body cells requires the development of novel wound curing strategies. Herein, a novel pH-responsive hydrogel (RPC/PB) was fabricated using poly(vinyl alcohol)-borax (PB) and natural antibiotic resveratrol grafted cellulose nanofibrils (RPC) for bacterial-infected wound management. Results In this hydrogel matrix, RPC conjugate was interpenetrated in the PB network to form a semi-interpenetrating network that exhibited robust mechanical properties (fracture strength of 149.6 kPa), high self-healing efficiency (> 90%), and excellent adhesion performance (tissue shear stress of 54.2 kPa). Interestingly, the induced RPC/PB hydrogel showed pH-responsive drug release behavior, the cumulative release amount of resveratrol in pH 5.4 was 2.33 times than that of pH 7.4, which was adapted well to the acidic wound microenvironment. Additionally, this RPC/PB hydrogel exhibited excellent biocompatibility and antioxidant effect. Moreover, in vitro and in vivo results revealed that such RPC/PB hydrogel had excellent antibacterial, skin tissue regeneration and wound closure capabilities. Conclusion Therefore, the generated RPC/PB hydrogel could be an excellent wound dressing for bacteria-infected wound healing. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01523-5.
Collapse
|
34
|
Pajares-Chamorro N, Hammer ND, Chatzistavrou X. Materials for restoring lost Activity: Old drugs for new bugs. Adv Drug Deliv Rev 2022; 186:114302. [PMID: 35461913 DOI: 10.1016/j.addr.2022.114302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 11/01/2022]
Abstract
The escalation of bacterial resistance to conventional medical antibiotics is a serious concern worldwide. Improvements to current therapies are urgently needed to address this problem. The synergistic combination of antibiotics with other agents is a strategic solution to combat multi-drug-resistant bacteria. Although these combinations decrease the required high dosages and therefore, reduce the toxicity of both agents without compromising the bactericidal effect, they cannot stop the development of further resistance. Recent studies have shown certain elements restore the ability of antibiotics to destroy bacteria that have acquired resistance to them. Due to these synergistic activities, organic and inorganic molecules have been investigated with the goal of restoring antibiotics in new approaches that mitigate the risk of expanding resistance. Herein, we summarize recent studies that restore antibiotics once thought to be ineffective, but have returned to our armamentarium through innovative, combinatorial efforts. A special focus is placed on the mechanisms that allow the synergistic combinations to combat bacteria. The promising data that demonstrated restoration of antimicrobials, supports the notion to find more combinations that can combat antibiotic-resistant bacteria.
Collapse
|
35
|
Xu Z, Wang T, Liu J. Recent Development of Polydopamine Anti-Bacterial Nanomaterials. Int J Mol Sci 2022; 23:ijms23137278. [PMID: 35806281 PMCID: PMC9266540 DOI: 10.3390/ijms23137278] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/01/2023] Open
Abstract
Polydopamine (PDA), as a mussel-inspired material, exhibits numerous favorable performance characteristics, such as a simple preparation process, prominent photothermal transfer efficiency, excellent biocompatibility, outstanding drug binding ability, and strong adhesive properties, showing great potential in the biomedical field. The rapid development of this field in the past few years has engendered substantial progress in PDA antibacterial materials. This review presents recent advances in PDA-based antimicrobial materials, including the preparation methods and antibacterial mechanisms of free-standing PDA materials and PDA-based composite materials. Furthermore, the urgent challenges and future research opportunities for PDA antibacterial materials are discussed.
Collapse
Affiliation(s)
- Zhengwei Xu
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China;
| | - Tingting Wang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117583, Singapore
- Correspondence: (T.W.); (J.L.)
| | - Junqiu Liu
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China;
- Correspondence: (T.W.); (J.L.)
| |
Collapse
|
36
|
Lyons A, Kirkham J, Blades K, Orr D, Dauncey E, Smith O, Dick E, Walker R, Matthews T, Bunt A, Finlayson J, Morrison I, Savage VJ, Moyo E, Butler HS, Newman R, Ooi N, Smith A, Charrier C, Ratcliffe AJ, Stokes NR, Best S, Salisbury AM, Craighead M, Cooper IR. Discovery and structure-activity relationships of a novel oxazolidinone class of bacterial type II topoisomerase inhibitors. Bioorg Med Chem Lett 2022; 65:128648. [PMID: 35231579 DOI: 10.1016/j.bmcl.2022.128648] [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] [Received: 01/05/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/02/2022]
Abstract
There is an increasingly urgent and unmet medical need for novel antibiotic drugs that tackle infections caused by multidrug-resistant (MDR) pathogens. Novel bacterial type II topoisomerase inhibitors (NBTIs) are of high interest due to limited cross-resistance with fluoroquinolones, however analogues with Gram-negative activity often suffer from hERG channel inhibition. A novel series of bicyclic-oxazolidinone inhibitors of bacterial type II topoisomerase were identified which display potent broad-spectrum anti-bacterial activity, including against MDR strains, along with an encouraging in vitro safety profile. In vivo proof of concept was achieved in a A. baumannii mouse thigh infection model.
Collapse
Affiliation(s)
- Amanda Lyons
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - James Kirkham
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Kevin Blades
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - David Orr
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | | | - Oliver Smith
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Emma Dick
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Rolf Walker
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Teresa Matthews
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Adam Bunt
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | | | - Ian Morrison
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Victoria J Savage
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Emmanuel Moyo
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Hayley S Butler
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Rebecca Newman
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Nicola Ooi
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Andrew Smith
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Cédric Charrier
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | | | - Neil R Stokes
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Stuart Best
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | | | - Mark Craighead
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Ian R Cooper
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK.
| |
Collapse
|
37
|
Fortunato Costa K, Almeida Araújo F, Morais J, Lisboa Frances CR, Ramos RTJ. Text mining for identification of biological entities related to antibiotic resistant organisms. PeerJ 2022; 10:e13351. [PMID: 35539017 PMCID: PMC9080439 DOI: 10.7717/peerj.13351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 04/07/2022] [Indexed: 01/13/2023] Open
Abstract
Antimicrobial resistance is a significant public health problem worldwide. In recent years, the scientific community has been intensifying efforts to combat this problem; many experiments have been developed, and many articles are published in this area. However, the growing volume of biological literature increases the difficulty of the biocuration process due to the cost and time required. Modern text mining tools with the adoption of artificial intelligence technology are helpful to assist in the evolution of research. In this article, we propose a text mining model capable of identifying and ranking prioritizing scientific articles in the context of antimicrobial resistance. We retrieved scientific articles from the PubMed database, adopted machine learning techniques to generate the vector representation of the retrieved scientific articles, and identified their similarity with the context. As a result of this process, we obtained a dataset labeled "Relevant" and "Irrelevant" and used this dataset to implement one supervised learning algorithm to classify new records. The model's overall performance reached 90% accuracy and the f-measure (harmonic mean between the metrics) reached 82% accuracy for positive class and 93% for negative class, showing quality in the identification of scientific articles relevant to the context. The dataset, scripts and models are available at https://github.com/engbiopct/TextMiningAMR.
Collapse
Affiliation(s)
- Kelle Fortunato Costa
- Programa de pós-graduação em Engenharia Elétrica, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Fabrício Almeida Araújo
- Biological Science Institute, Universidade Federal do Pará, Belém, Pará, Brazil,Universidade Federal Rural da Amazônia, Belém, Pará, Brazil
| | | | | | - Rommel T. J. Ramos
- Biological Science Institute, Universidade Federal do Para, Belém, Pará, Brazil
| |
Collapse
|
38
|
Li Y, Feng T, Wang Y. The role of bacterial signaling networks in antibiotics response and resistance regulation. MARINE LIFE SCIENCE & TECHNOLOGY 2022; 4:163-178. [PMID: 37073223 PMCID: PMC10077285 DOI: 10.1007/s42995-022-00126-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/07/2022] [Indexed: 05/03/2023]
Abstract
Excessive use of antibiotics poses a threat to public health and the environment. In ecosystems, such as the marine environment, antibiotic contamination has led to an increase in bacterial resistance. Therefore, the study of bacterial response to antibiotics and the regulation of resistance formation have become an important research field. Traditionally, the processes related to antibiotic responses and resistance regulation have mainly included the activation of efflux pumps, mutation of antibiotic targets, production of biofilms, and production of inactivated or passivation enzymes. In recent years, studies have shown that bacterial signaling networks can affect antibiotic responses and resistance regulation. Signaling systems mostly alter resistance by regulating biofilms, efflux pumps, and mobile genetic elements. Here we provide an overview of how bacterial intraspecific and interspecific signaling networks affect the response to environmental antibiotics. In doing so, this review provides theoretical support for inhibiting bacterial antibiotic resistance and alleviating health and ecological problems caused by antibiotic contamination.
Collapse
Affiliation(s)
- Yuying Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
| | - Tao Feng
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
| | - Yan Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- Laboratory for Marine Ecology and Environmental Science, National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266071 China
| |
Collapse
|
39
|
Duah IK, Khaligh A, Koç A, Başaran DDA, Tuncel D. Porphyrin cross‐linked conjugated polymer nanoparticles‐based photosensitizer for antimicrobial and anticancer photodynamic therapies. J Appl Polym Sci 2022. [DOI: 10.1002/app.51777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
| | - Aisan Khaligh
- Department of Chemistry Bilkent University Ankara Turkey
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM) Bilkent University Ankara Turkey
| | - Ahmet Koç
- Department of Chemistry Bilkent University Ankara Turkey
| | - Duygu Deniz Akolpoğlu Başaran
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM) Bilkent University Ankara Turkey
| | - Dönüs Tuncel
- Department of Chemistry Bilkent University Ankara Turkey
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM) Bilkent University Ankara Turkey
| |
Collapse
|
40
|
Synthesis and antimicrobial potential of spirooxindolopyrrolidine tethered oxindole heterocyclic hybrid against multidrug resistant microbial pathogens. Process Biochem 2022. [DOI: 10.1016/j.procbio.2021.12.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
41
|
Dey N, Kamatchi C, Vickram AS, Anbarasu K, Thanigaivel S, Palanivelu J, Pugazhendhi A, Ponnusamy VK. Role of nanomaterials in deactivating multiple drug resistance efflux pumps - A review. ENVIRONMENTAL RESEARCH 2022; 204:111968. [PMID: 34453898 DOI: 10.1016/j.envres.2021.111968] [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] [Received: 05/17/2021] [Revised: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 06/13/2023]
Abstract
The changes in lifestyle and living conditions have affected not only humans but also microorganisms. As man invents new drugs and therapies, pathogens alter themselves to survive and thrive. Multiple drug resistance (MDR) is the talk of the town for decades now. Many generations of medications have been termed useless as MDR rises among the infectious population. The surge in nanotechnology has brought a new hope in reducing this aspect of resistance in pathogens. It has been observed in several laboratory-based studies that the use of nanoparticles had a synergistic effect on the antibiotic being administered to the pathogen; several resistant strains scummed to the stress created by the nanoparticles and became susceptible to the drug. The major cause of resistance to date is the efflux system, which makes the latest generation of antibiotics ineffective without reaching the target site. If species-specific nanomaterials are used to control the activity of efflux pumps, it could revolutionize the field of medicine and make the previous generation resistant medications active once again. Therefore, the current study was devised to assess and review nanoparticles' role on efflux systems and discuss how specialized particles can be designed towards an infectious host's particular drug ejection systems.
Collapse
Affiliation(s)
- Nibedita Dey
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - C Kamatchi
- Department of Biotechnology, The Oxford College of Science, Bengaluru, India
| | - A S Vickram
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - K Anbarasu
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - S Thanigaivel
- Department of Biomedical Engineering, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Jeyanthi Palanivelu
- Department of Biotechnology, Vel Tech Rangarajan Dr. Sagunthala R&D Institute of Science and Technology, Chennai, India
| | | | - Vinoth Kumar Ponnusamy
- Department of Medicinal and Applied Chemistry & Research Center for Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital (KMUH), Kaohsiung City, 807, Taiwan; Program of Aquatic Science and Technology, College of Hydrosphere Science, National Kaohsiung University of Science and Technology (NKUST), Kaohsiung City, Taiwan.
| |
Collapse
|
42
|
Garcia ÍR, de Oliveira Garcia FA, Pereira PS, Coutinho HDM, Siyadatpanah A, Norouzi R, Wilairatana P, de Lourdes Pereira M, Nissapatorn V, Tintino SR, Rodrigues FFG. Microbial resistance: The role of efflux pump superfamilies and their respective substrates. Life Sci 2022; 295:120391. [PMID: 35149116 DOI: 10.1016/j.lfs.2022.120391] [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] [Received: 10/09/2021] [Revised: 02/02/2022] [Accepted: 02/06/2022] [Indexed: 12/24/2022]
Abstract
The microorganism resistance to antibiotics has become one of the most worrying issues for science due to the difficulties related to clinical treatment and the rapid spread of diseases. Efflux pumps are classified into six groups of carrier proteins that are part of the different types of mechanisms that contribute to resistance in microorganisms, allowing their survival. The present study aimed to carry out a bibliographic review on the superfamilies of carriers in order to understand their compositions, expressions, substrates, and role in intrinsic resistance. At first, a search for manuscripts was carried out in the databases Medline, Pubmed, ScienceDirect, and Scielo, using as descriptors: efflux pump, expression, pump inhibitors and efflux superfamily. For article selection, two criteria were taken into account: for inclusion, those published between 2000 and 2020, including textbooks, and for exclusion, duplicates and academic collections. In this research, 139,615 published articles were obtained, with 312 selected articles and 7 book chapters that best met the aim. From the comprehensive analysis, it was possible to consider that the chromosomes and genetic elements can contain genes encoding efflux pumps and are responsible for multidrug resistance. Even though this is a well-explored topic in the scientific community, understanding the behavior of antibiotics as substrates that increase the expression of pump-encoding genes has challenged medicine. This review study succinctly summarizes the most relevant features of these systems, as well as their contribution to multidrug resistance.
Collapse
Affiliation(s)
| | | | | | | | - Abolghasem Siyadatpanah
- Ferdows School of Paramedical and Health, Birjand University of Medical Sciences, Birjand, Iran
| | - Roghayeh Norouzi
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials & Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences and Research Excellence Center for Innovation and Health, Walailak University, Thailand
| | | | | |
Collapse
|
43
|
Wu L, Zhang Q, Deng Z, Yu Y. From solo to duet, intersections of natural product assembly with self-resistance. Nat Prod Rep 2022; 39:919-925. [PMID: 34989738 DOI: 10.1039/d1np00064k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Covering: up to 2021Self-resistance mechanisms adopted by natural product producers have long been recognized and studied as a standalone system separated from the assembly machinery. However, as more examples of self-resistance have been characterized in detail, it has been revealed that self-resistance could associate with the assembly machinery to fulfill the task of biosynthesis. This review summarizes different self-resistance mechanisms showing a common feature: intersection with natural product assembly. Furthermore, their possible evolutionary origin and synthetic biology applications are discussed.
Collapse
Affiliation(s)
- Linrui Wu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Qian Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Zixin Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Yi Yu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
44
|
Seregina TA, Lobanov KV, Shakulov RS, Mironov AS. Enhancement of the Bactericidal Effect of Antibiotics by Inhibition of Enzymes Involved in Production of Hydrogen Sulfide in Bacteria. Mol Biol 2022; 56:638-648. [PMID: 36217334 PMCID: PMC9534473 DOI: 10.1134/s0026893322050120] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 11/23/2022]
Abstract
Counteraction of the origin and distribution of multidrug-resistant pathogens responsible for intra-hospital infections is a worldwide issue in medicine. In this brief review, we discuss the results of our recent investigations, which argue that many antibiotics, along with inactivation of their traditional biochemical targets, can induce oxidative stress (ROS production), thus resulting in increased bactericidal efficiency. As we previously showed, hydrogen sulfide, which is produced in the cells of different pathogens protects them not only against oxidative stress but also against bactericidal antibiotics. Next, we clarified the interplay of oxidative stress, cysteine metabolism, and hydrogen sulfide production. Finally, demonstrated that small molecules, which inhibit a bacterial enzyme involved in hydrogen sulfide production, potentiate bactericidal antibiotics including quinolones, beta-lactams, and aminoglycosides against bacterial pathogens in in vitro and in mouse models of infection. These inhibitors also suppress bacterial tolerance to antibiotics by disrupting the biofilm formation and substantially reducing the number of persister bacteria, which survive the antibiotic treatment. We hypothesise that agents which limit hydrogen sulfide biosynthesis are effective tools to counteract the origin and distribution of multidrug-resistant pathogens.
Collapse
Affiliation(s)
- T. A. Seregina
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - K. V. Lobanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - R. S. Shakulov
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - A. S. Mironov
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| |
Collapse
|
45
|
Saleem H, Ashfaq UA, Nadeem H, Zubair M, Siddique MH, Rasul I. Subtractive genomics and molecular docking approach to identify drug targets against Stenotrophomonas maltophilia. PLoS One 2021; 16:e0261111. [PMID: 34910751 PMCID: PMC8673605 DOI: 10.1371/journal.pone.0261111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/25/2021] [Indexed: 11/18/2022] Open
Abstract
Stenotrophomonas maltophilia is a multidrug resistant pathogen associated with high mortality and morbidity in patients having compromised immunity. The efflux systems of S. maltophilia include SmeABC and SmeDEF proteins, which assist in acquisition of multiple-drug-resistance. In this study, proteome based mapping was utilized to find out the potential drug targets for S. maltophilia strain k279a. Various tools of computational biology were applied to remove the human-specific homologous and pathogen-specific paralogous sequences from the bacterial proteome. The CD-HIT analysis selected 4315 proteins from total proteome count of 4365 proteins. Geptop identified 407 essential proteins, while the BlastP revealed approximately 85 non-homologous proteins in the human genome. Moreover, metabolic pathway and subcellular location analysis were performed for essential bacterial genes, to describe their role in various cellular processes. Only two essential proteins (Acyl-[acyl-carrier-protein]—UDP-N acetyl glucosamine O-acyltransferase and D-alanine-D-alanine ligase) as candidate for potent targets were found in proteome of the pathogen, in order to design new drugs. An online tool, Swiss model was employed to model the 3D structures of both target proteins. A library of 5000 phytochemicals was docked against those proteins through the molecular operating environment (MOE). That resulted in to eight inhibitors for both proteins i.e. enterodiol, aloin, ononin and rhinacanthinF for the Acyl-[acyl-carrier-protein]—UDP-N acetyl glucosamine O-acyltransferase, and rhazin, alkannin beta, aloesin and ancistrocladine for the D-alanine-D-alanine ligase. Finally the ADMET was done through ADMETsar. This study supported the development of natural as well as cost-effective drugs against S. maltophilia. These inhibitors displayed the effective binding interactions and safe drug profiles. However, further in vivo and in vitro validation experiment might be performed to check their drug effectiveness, biocompatibility and their role as effective inhibitors.
Collapse
Affiliation(s)
- Hira Saleem
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Habibullah Nadeem
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Zubair
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Hussnain Siddique
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Ijaz Rasul
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
- * E-mail:
| |
Collapse
|
46
|
Wang Q, Zhu L, Yu Y, Guan H, Xu Z. Microbial Screening of Marine Natural Product Inhibitors for the 6′-Aminoglycoside Acetyltransferase 2″-Aminoglycoside Phosphotransferase [AAC(6′)-APH(2″)] Bifunctional Enzyme by Ultra-High Performance Liquid Chromatography–Mass Spectrometry (UHPLC-MS). ANAL LETT 2021. [DOI: 10.1080/00032719.2021.1903025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Qian Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Innovation Center for Marine Drugs Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Li Zhu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Innovation Center for Marine Drugs Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yi Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Innovation Center for Marine Drugs Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Huashi Guan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Innovation Center for Marine Drugs Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Zhe Xu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Innovation Center for Marine Drugs Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
47
|
Huang H, Wang X, Wang W, Qu X, Song X, Zhang Y, Zhong L, Yang DP, Dong X, Zhao Y. Injectable hydrogel for postoperative synergistic photothermal-chemodynamic tumor and anti-infection therapy. Biomaterials 2021; 280:121289. [PMID: 34861512 DOI: 10.1016/j.biomaterials.2021.121289] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 02/06/2023]
Abstract
Tumor surgery is usually accompanied by neoplasm residual, tissue defects, and multi-drug resistant bacterial infection, causing high tumor recurrence, low survival rate, and chronic wounds. Herein, a light-activated injectable hydrogel based on bioactive nanocomposite system is developed by incorporating Ag2S nanodots conjugated Fe-doped bioactive glass nanoparticles (BGN-Fe-Ag2S) into biodegradable PEGDA and AIPH solution for inhibiting tumor growth, treating bacterial infection, and promoting wound healing. Under laser irradiation, the photothermal effect mediated by Ag2S nanodots would trigger the decomposition of AIPH, generating alkyl radicals to initiate the gelation of PEGDA. The in-situ gelatinized hydrogel, with outstanding photothermal effect and chemodynamic effect derived from the doped Fe in BGN-Fe-Ag2S, can not only eliminate multidrug-resistant bacteria but also efficiently ablated tumor during treatment. Moreover, the hydrogel significantly accelerated wound healing with more skin appendages in the full-thickness cutaneous wounds model because of the hydrolysis of bioactive glass. These results manifest that this multifunctional hydrogel is a suitable biomaterial to inhibit tumor proliferation and overcome tissue bacterial infection after surgical removal of tumors.
Collapse
Affiliation(s)
- Han Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Xiaorui Wang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Weili Wang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Xinyu Qu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Xuejiao Song
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China.
| | - Yewei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 211166, China
| | - Liping Zhong
- National Center for International Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Theranostics, Guangxi Medical University, Guangxi, 530021, China
| | - Da-Peng Yang
- College of Chemical Engineering and Materials Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China.
| | - Yongxiang Zhao
- National Center for International Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Theranostics, Guangxi Medical University, Guangxi, 530021, China.
| |
Collapse
|
48
|
Synthesis and bioactivities of new N-terminal dipeptide mimetics with aromatic amide moiety: Broad-spectrum antibacterial activity and high antineoplastic activity. Eur J Med Chem 2021; 228:113977. [PMID: 34772526 DOI: 10.1016/j.ejmech.2021.113977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 11/20/2022]
Abstract
The increasingly growing epidemics of multidrug-resistant bacteria are becoming severe public health threat. There is in an urgent need to develop new antibacterial agents with broad-spectrum antibacterial activity and high selectivity. Here, a series of N-terminal dipeptide mimetics with an aromatic amide moiety were synthesized from amino acids. The effects of amino acid type and aromatic moiety on the biological activities of the mimetics were evaluated. The dipeptide mimetics not only showed significant broad-spectrum antibacterial activity against Gram-negative (Escherichia coli and Klebsiella pneumoniae), Gram-positive (Staphylococcus aureus) and drug-resistant bacterium MRSA (methicillin-resistant S. aureus) but also demonstrated high selectivity for S. aureus versus mammalian erythrocytes. The coupling product of L-valine with p-alkynylaniline (dipeptide mimetic 7) exhibited the best antibacterial activities with minimum inhibitory concentration (MIC) ranging from 2.5 to 5 μg/mL. Moreover, the bactericidal kinetics and multi-passage resistance tests indicated that the mimetic 7 both rapidly killed bacteria and had a low probability of emergence of antimalarial resistance. Meanwhile, the mimetic 7 possessed the ability to both inhibit bacterial biofilm formation and eradicate mature biofilm. The depolarization and destruction of the bacterial cell membrane is the main sterilization mechanism, which hinders the propensity to develop bacterial resistance. Furthermore, the mimetic 7 also showed good antineoplastic activity against gastric cancer cell (SGC 7901, IC50 = 70.8 μg/mL), while it had very low toxicity to mammalian cell (L929). The mimetics bear considerable potential to be used as antibacterial and anticancer agents to combat antibiotic resistance.
Collapse
|
49
|
Arumugam N, Almansour AI, Suresh Kumar R. Antimicrobial activities of spirooxindolopyrrolidine tethered dicarbonitrile heterocycles against multidrug resistant nosocomial pathogens. J Infect Public Health 2021; 14:1810-1814. [PMID: 34776342 DOI: 10.1016/j.jiph.2021.10.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/23/2021] [Accepted: 10/27/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Microbial infections together with rising drug resistance pose a threat to immunocompromised individual. In this perspective, compounds with spirooxindolopyrrolidine play a significant role in research on antimicrobial drug delivery research owing to their various pharmaceutical activities. Spiroheterocyclic compounds are present in number of medications as active motif due to their exceptional structural properties which enable for easy interaction with the protein of the biological target. Inspired by this biological precedent encouraged to synthesize a new class of dispirooxindole fused pyrrolidine heterocycles via a three-component cycloaddition strategy. MATERIALS AND METHODS The new class of structurally intriguing spirooxindolopyrrolidines were synthesized through three component cycloaddition process and the structure of products were assigned through spectroscopic analysis. The newly synthesized compounds were assessed for their antimicrobial sensitivity test with standard Kirby Bauer method with common drugs. RESULTS The structurally unexplored hybrid heterocycles fused spirooxindolopyrrolidine exhibited excellent antimicrobial activity against the common nosocomial microbial pathogens. Of four compounds, the compound bearing a chlorine atom on the aryl ring (4a) exhibited significant antimicrobial activity (zone of inhibition: 9.00 ± 1.00-17.00 ± 0.35 mm and MIC: 16.00-256.00 μg/mL) against selected nosocomial infection causing microbial pathogens. Hence, the compound 4a has been considered as an effective drug of interest in therapeutic field for compacting infectious diseases causing pathogens. CONCLUSION With an aim of developing more effective and economically more affordable antimicrobial leads with a unique mechanism of action, we have designed and synthesized structurally diverse spirooxindolopyrrolidine tethered hybrids that has been assayed against multidrug resistant nosocomial pathogens. The regioisomer having chloro substituted on the phenyl ring showed potent activity when compared to standard drug. Future studies are required to explicate the pharmacological properties of new hybrid heterocycles that have been synthesized in our laboratory for the novel therapeutic development.
Collapse
Affiliation(s)
- Natarajan Arumugam
- Department of Chemistry, College of Science, King Saud University, P.O Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdulrahman I Almansour
- Department of Chemistry, College of Science, King Saud University, P.O Box 2455, Riyadh 11451, Saudi Arabia.
| | - Raju Suresh Kumar
- Department of Chemistry, College of Science, King Saud University, P.O Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
50
|
Mataracı-Kara E, Bayrak N, Yıldız M, Yıldırım H, Özbek-Çelik B, Tuyun AF. Discovery and structure-activity relationships of the quinolinequinones: Promising antimicrobial agents and mode of action evaluation. Drug Dev Res 2021; 83:628-636. [PMID: 34668593 DOI: 10.1002/ddr.21893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/11/2021] [Accepted: 10/04/2021] [Indexed: 01/09/2023]
Abstract
In our pursuit of developing the novel, potent, and selective antimicrobial agents, we managed to obtain the quinolinequinone for their antimicrobial profile with minimal inhibitory concentrations (MICs) determined against a panel of seven bacterial strains (three gram-positive and four gram-negative bacteria) and three fungi. The structure-activity relationship (SAR) for the quinolinequinone class of antimicrobials was determined. Interestingly, QQ1, QQ4, QQ6-9, QQ12, and QQ13 displayed equal antibacterial potential against S. aureus (MIC = 1.22 mg/L), respectively, to the standard positive control Cefuroxime-Na. QQ10 had the best inhibitory activity with the MIC value of 1.22 mg/L (fourfold more potent compared to reference standard Clotrimazole) against Candida albicans. On the other hand, while QQ10 is not too effective against gram-positive bacteria as much as the other analogs, QQ10 was the most effective quinolinequinones against fungi. Selected quinolinequinones were further evaluated for the mode of action, using in vitro antibiofilm activity, bactericidal activity by using time-kill curve assay, antibiofilm activity, and potential antimicrobial activity against each of 32 clinically obtained resistant strains of Gram-positive Bacteria. The results also revealed that the QQ14 had specific antifungal activity against fungi in particular C. albicans. Our results clearly showed that quinolinequinones are much more active in the inhibition of the biofilm attachment process than the inhibition of mature biofilm formation. Thus, as treatment options are narrowing for Methicillin-resistant Staphylococcus spp., Vancomycin-resistant Staphylococcus spp. daily, the quinolinequinones reported herein display promise as the lead candidates for further clinical applications against serious infections.
Collapse
Affiliation(s)
- Emel Mataracı-Kara
- Pharmaceutical Microbiology Department, Pharmacy Faculty, Istanbul University, Istanbul, Turkey
| | - Nilüfer Bayrak
- Department of Chemistry, Faculty of Engineering, Istanbul University, Istanbul, Turkey
| | - Mahmut Yıldız
- Chemistry Department, Gebze Technical University, Kocaeli, Turkey
| | - Hatice Yıldırım
- Department of Chemistry, Faculty of Engineering, Istanbul University, Istanbul, Turkey
| | - Berna Özbek-Çelik
- Pharmaceutical Microbiology Department, Pharmacy Faculty, Istanbul University, Istanbul, Turkey
| | - Amaç Fatih Tuyun
- Department of Chemistry, Faculty of Science, Istanbul University, Istanbul, Turkey
| |
Collapse
|