1
|
Zhou W, Li Q, Liu M, Gu X, He X, Xie C, Fan Q. Biodegradable semiconducting polymer nanoparticles for phototheranostics. J Mater Chem B 2025; 13:2242-2253. [PMID: 39815890 DOI: 10.1039/d4tb02437k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Semiconducting polymer nanoparticles (SPNs) have been widely applied for phototheranostics. However, the disadvantage of in vivo long-term metabolism greatly suppresses the clinical application of SPNs. To improve the metabolic rate and minimize the long-term toxicity of SPNs, biodegradable semiconducting polymers (BSPs), whose backbones may be degraded under certain conditions, have been designed. This review summarizes recent advances in BSP-constructed nanoparticles (BSPNs) for phototheranostics. BSPs are divided into two categories: conjugated backbone degradable BSPs (CBD-BSPs) and non-conjugated backbone degradable BSPs (NCBD-BSPs), based on the feature of chemical structure. The biological applications, including cancer imaging and combination therapy, of these BSPNs are described. Finally, the conclusion and future perspectives of this field are discussed.
Collapse
Affiliation(s)
- Wen Zhou
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Qiang Li
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Mingming Liu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Xuxuan Gu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Xiaowen He
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Chen Xie
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Quli Fan
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| |
Collapse
|
2
|
Du M, Chi C, Xiong L, Rong J, Yi M, Zhao Q, Chi X. Convergence of Hypervirulence and Multidrug-Resistance in Burkholderia cepacia Complex Isolates from Patients with COVID-19. Infect Drug Resist 2024; 17:5855-5866. [PMID: 39741887 PMCID: PMC11687121 DOI: 10.2147/idr.s495676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
Purpose Burkholderia is a conditioned pathogen in the medical setting and mainly affects patients with cystic fibrosis. We found co-infection with Burkholderia cepacia complex (Bcc) in many patients with respiratory tract infections, including H7N9 and COVID-19. However, previous studies have not focused on co-infections with BCC and respiratory viruses. Therefore, this study attempted to clarify the evolution of COVID-19-Bcc and H7N9-Bcc in terms of genetic background, antibiotic resistance, and virulence phenotypes. Methods This study retrospectively collected 49 Bcc isolated from patients with H7N9 and COVID-19 in a tertiary hospital of Zhejiang Province, of which 42 isolates were isolated from patients with H7N9, seven isolates were isolated from patients with COVID-19. The collected isolates were tested for antibiotic susceptibility, Galleria mellonella infection model, and whole-genome COVID-19-Bcc Characterization. Results The test results of 49 strains of Bcc showed that the strains isolated from COVID-19 patients accounted for 57.1% of multidrug-resistance resistant strains. Statistical analysis of the median lethal time of G. mellonella showed that the median fatal time for COVID-19-Bcc was shorter and more virulent than that of H7N9-Bcc (P<0.05). The results of phylogenetic analysis indicated that COVID-19-Bcc may have evolved from H7N9-Bcc. Conclusion In this study, co-infection with BCC in many patients with respiratory tract infections, including H7N9 and COVID-19, was first identified and clarified that COVID-19-Bcc may have evolved from H7N9-Bcc and has the characteristics of hypervirulence and multidrug resistance.
Collapse
Affiliation(s)
- Mengjiao Du
- Department of Medical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People’s Republic of China
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Cheng Chi
- Department of Medical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People’s Republic of China
| | - LuYing Xiong
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Jincheng Rong
- Department of Medical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People’s Republic of China
| | - Maoli Yi
- Department of Medical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People’s Republic of China
| | - Qi Zhao
- Department of Medical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People’s Republic of China
| | - Xiaohui Chi
- Department of Medical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People’s Republic of China
| |
Collapse
|
3
|
Zhang Q. Bacteria carrying mobile colistin resistance genes and their control measures, an updated review. Arch Microbiol 2024; 206:462. [PMID: 39516398 DOI: 10.1007/s00203-024-04188-w] [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: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The plasmid encoded mobile colistin resistance (MCRs) enzyme poses a significant challenge to the clinical efficacy of colistin, which is frequently employed as a last resort antibiotic for treating infections caused by multidrug resistant bacteria. This transferase catalyzes the addition of positively charged phosphoethanolamine to lipid A of the outer membrane of gram-negative bacteria, thereby facilitating the acquired colistin resistance. This review aims to summarize and critically discuss recent advancements in the distribution and pathogenesis of mcr-positive bacteria, as well as the various control measures available for treating these infections. In addition, the ecology of mcr genes, colistin-resistance mechanism, co-existence with other antibiotic resistant genes, and their impact on clinical treatment are also analyzed to address the colistin resistance crisis. These insights provide a comprehensive perspective on MCRs and serve as a valuable reference for future therapeutic approaches to effectively combat mcr-positive bacterial infections.
Collapse
Affiliation(s)
- Qi Zhang
- Centre for Eye and Vision Research, Hong Kong Science Park, Hong Kong, China.
| |
Collapse
|
4
|
Lourenço M, Duarte N, Ribeiro IAC. Exploring Biosurfactants as Antimicrobial Approaches. Pharmaceuticals (Basel) 2024; 17:1239. [PMID: 39338401 PMCID: PMC11434949 DOI: 10.3390/ph17091239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Antibacterial resistance is one of the most important global threats to human health. Several studies have been performed to overcome this problem and infection-preventive approaches appear as promising solutions. Novel antimicrobial preventive molecules are needed and microbial biosurfactants have been explored in that scope. Considering their structure, these biomolecules can be divided into different classes, glycolipids and lipopeptides being the most studied. Besides their antimicrobial activity, biosurfactants have the advantage of being biocompatible, biodegradable, and non-toxic, which favor their application in several areas, including the health sector. Often, the most difficult infections to fight are associated with biofilm formation, particularly in medical devices. Strategies to overcome micro-organism attachment are thus emergent, and it is possible to take advantage of the antimicrobial/antibiofilm properties of biosurfactants to produce surfaces that are more resistant to the deposition/attachment of bacteria. Approaches such as the covalent bond of biosurfactants to the medical device surface leading to repulsive physical-chemical interactions or contact killing can be selected. Simpler strategies such as the absorption of biosurfactants on surfaces are also possible, eliminating micro-organisms in the vicinity. This review will focus on the physical and chemical characteristics of biosurfactants, their antimicrobial activity, antimicrobial/antibiofilm approaches, and finally on their structure-activity relationship.
Collapse
Affiliation(s)
| | - Noélia Duarte
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal;
| | - Isabel A. C. Ribeiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal;
| |
Collapse
|
5
|
Stachura DL, Kalyvas JT, Abell AD. New Potent Sulfonamide-Based Inhibitors of S. aureus Biotin Protein Ligase. ACS Med Chem Lett 2024; 15:1467-1473. [PMID: 39291019 PMCID: PMC11403734 DOI: 10.1021/acsmedchemlett.4c00325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024] Open
Abstract
The key regulatory metabolic enzyme, biotin protein ligase (BPL), is an attractive target for the development of novel antibiotics against multi-drug-resistant bacteria, such as Staphylococcus aureus. Here we report the synthesis and assay of a new series of inhibitors (6-9) against S. aureus BPL (SaBPL), where a component sulfonamide linker was used to mimic the acyl-phosphate group of the natural intermediate biotinyl-5'-AMP (1). A pivotal correlation between the acidity of the central NH of the sulfonamide linker of 6-9 and in vitro inhibitory activity against SaBPL was observed. Specifically, sulfonylcarbamate 8, with its highly acidic sulfonyl central NH, as evaluated by 1H NMR spectroscopy, showed exceptional potency (K i = 10.3 ± 3.8 nM). Furthermore, three inhibitors demonstrated minimum inhibitory concentrations of 16-32 μg/mL against clinical methicillin-resistant S. aureus (MRSA) strains.
Collapse
Affiliation(s)
- Damian L Stachura
- Centre for Nanoscale BioPhotonics (CNBP) and Institute of Photonics and Advanced Sensing (IPAS), Department of Chemistry, School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - John T Kalyvas
- Centre for Nanoscale BioPhotonics (CNBP) and Institute of Photonics and Advanced Sensing (IPAS), Department of Chemistry, School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew D Abell
- Centre for Nanoscale BioPhotonics (CNBP) and Institute of Photonics and Advanced Sensing (IPAS), Department of Chemistry, School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
6
|
Thompson TP, Gilmore BF. Exploring halophilic environments as a source of new antibiotics. Crit Rev Microbiol 2024; 50:341-370. [PMID: 37079280 DOI: 10.1080/1040841x.2023.2197491] [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: 09/16/2022] [Accepted: 03/25/2023] [Indexed: 04/21/2023]
Abstract
Microbial natural products from microbes in extreme environments, including haloarchaea, and halophilic bacteria, possess a huge capacity to produce novel antibiotics. Additionally, enhanced isolation techniques and improved tools for genomic mining have expanded the efficiencies in the antibiotic discovery process. This review article provides a detailed overview of known antimicrobial compounds produced by halophiles from all three domains of life. We summarize that while halophilic bacteria, in particular actinomycetes, contribute the vast majority of these compounds the importance of understudied halophiles from other domains of life requires additional consideration. Finally, we conclude by discussing upcoming technologies- enhanced isolation and metagenomic screening, as tools that will be required to overcome the barriers to antimicrobial drug discovery. This review highlights the potential of these microbes from extreme environments, and their importance to the wider scientific community, with the hope of provoking discussion and collaborations within halophile biodiscovery. Importantly, we emphasize the importance of bioprospecting from communities of lesser-studied halophilic and halotolerant microorganisms as sources of novel therapeutically relevant chemical diversity to combat the high rediscovery rates. The complexity of halophiles will necessitate a multitude of scientific disciplines to unravel their potential and therefore this review reflects these research communities.
Collapse
Affiliation(s)
- Thomas P Thompson
- Biofilm Research Group, School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - Brendan F Gilmore
- Biofilm Research Group, School of Pharmacy, Queen's University Belfast, Belfast, UK
| |
Collapse
|
7
|
Yan YF, Liu Y, Liang H, Cai L, Yang XY, Yin TP. The erythromycin polyketide compound TMC-154 stimulates ROS generation to exert antibacterial effects against Streptococcus pyogenes. J Proteomics 2024; 292:105057. [PMID: 38043864 DOI: 10.1016/j.jprot.2023.105057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
The erythromycin polyketide compound TMC-154 is a secondary metabolite that is isolated from the rhizospheric fungus Clonostachys rogersoniana associated with Panax notoginseng, which possesses antibacterial activity. However, its antibacterial mechanism has not been investigated thus far. In this study, proteomics coupled with bioinformatics approaches was used to explore the antibacterial mechanism of TMC-154. KEGG pathway enrichment analysis indicated that eight signaling pathways were associated with TMC-154, including oxidative phosphorylation, cationic antimicrobial peptide (CAMP) resistance, benzoate degradation, heme acquisition systems, glycine/serine and threonine metabolism, beta-lactam resistance, ascorbate and aldarate metabolism, and phosphotransferase system (PTS). Cell biology experiments confirmed that TMC-154 could induce reactive oxygen species (ROS) generation in Streptococcus pyogenes; moreover, TMC-154-induced antibacterial effects could be blocked by the inhibition of ROS generation with the antioxidant N-acetyl L-cysteine. In addition, TMC-154 combined with ciprofloxacin or chloramphenicol had synergistic antibacterial effects. These findings indicate the potential of TMC-154 as a promising drug to treat S. pyogenes infections. SIGNIFICANCE: Streptococcus pyogenes is a nearly ubiquitous human pathogen that causes a variety of diseases ranging from mild pharyngitis and skin infection to fatal sepsis and toxic heat shock syndrome. With the increasing incidence of known antibiotic resistance, there is an urgent need to find novel drugs with good antibacterial activity against S. pyogenes. In this study, we found that TMC-154, a secondary metabolite from the fungus Clonostachys rogersoniana, inhibited the growth of various bacteria, including Staphylococcus aureus, S. pyogenes, Streptococcus mutans, Pseudomonas aeruginosa and Vibrio parahemolyticus. Proteomic analysis combined with cell biology experiments revealed that TMC-154 stimulated ROS generation to exert antibacterial effects against S. pyogenes. This study provides potential options for the treatment of S. pyogenes infections in the future.
Collapse
Affiliation(s)
- Yuan-Feng Yan
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong 519041, China
| | - Ying Liu
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong 519041, China
| | - Hangeri Liang
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong 519041, China
| | - Le Cai
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Functional Molecules Analysis and Biotransformation Key Laboratory of Universities in Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, China
| | - Xiao-Yan Yang
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong 519041, China.
| | - Tian-Peng Yin
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong 519041, China.
| |
Collapse
|
8
|
Nazli A, Qiu J, Tang Z, He Y. Recent Advances and Techniques for Identifying Novel Antibacterial Targets. Curr Med Chem 2024; 31:464-501. [PMID: 36734893 DOI: 10.2174/0929867330666230123143458] [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: 05/24/2022] [Revised: 10/30/2022] [Accepted: 11/11/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND With the emergence of drug-resistant bacteria, the development of new antibiotics is urgently required. Target-based drug discovery is the most frequently employed approach for the drug development process. However, traditional drug target identification techniques are costly and time-consuming. As research continues, innovative approaches for antibacterial target identification have been developed which enabled us to discover drug targets more easily and quickly. METHODS In this review, methods for finding drug targets from omics databases have been discussed in detail including principles, procedures, advantages, and potential limitations. The role of phage-driven and bacterial cytological profiling approaches is also discussed. Moreover, current article demonstrates the advancements being made in the establishment of computational tools, machine learning algorithms, and databases for antibacterial target identification. RESULTS Bacterial drug targets successfully identified by employing these aforementioned techniques are described as well. CONCLUSION The goal of this review is to attract the interest of synthetic chemists, biologists, and computational researchers to discuss and improve these methods for easier and quicker development of new drugs.
Collapse
Affiliation(s)
- Adila Nazli
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China
| | - Jingyi Qiu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Avenue, Chongqing, 400714, P. R. China
| | - Ziyi Tang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Avenue, Chongqing, 400714, P. R. China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China
| |
Collapse
|
9
|
Worku S, Abebe T, Seyoum B, Alemu A, Shimelash Y, Yimer M, Abdissa A, Beyene GT, Swedberg G, Mihret A. Molecular Epidemiology of Methicillin-Resistant Staphylococcus aureus among Patients Diagnosed with Surgical Site Infection at Four Hospitals in Ethiopia. Antibiotics (Basel) 2023; 12:1681. [PMID: 38136715 PMCID: PMC10741212 DOI: 10.3390/antibiotics12121681] [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: 10/31/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a common cause of severe surgical site infections (SSI). The molecular epidemiology of MRSA is poorly documented in Ethiopia. This study is designed to determine the prevalence of MRSA and associated factors among patients diagnosed with SSI. A multicenter study was conducted at four hospitals in Ethiopia. A wound culture was performed among 752 SSI patients. This study isolated S. aureus and identified MRSA using standard bacteriology, Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry (MALDI-TOF MS), and cefoxitin disk diffusion test. The genes mecA, femA, vanA, and vanB were detected through PCR tests. S. aureus was identified in 21.6% of participants, with 24.5% of these being methicillin-resistant Staphylococci and 0.6% showing vancomycin resistance. Using MALDI-TOF MS for the 40 methicillin-resistant Staphylococci, we confirmed that 31 (77.5%) were S. aureus, 6 (15%) were Mammaliicoccus sciuri, and the other 3 (2.5%) were Staphylococcus warneri, Staphylococcus epidermidis, and Staphylococcus haemolyticus. The gene mecA was detected from 27.5% (11/40) of Staphylococci through PCR. Only 36.4% (4/11) were detected in S. aureus, and no vanA or vanB genes were identified. Out of 11 mecA-gene-positive Staphylococci, 8 (72.7%) were detected in Debre Tabor Comprehensive Specialized Hospital. Methicillin-resistant staphylococcal infections were associated with the following risk factors: age ≥ 61 years, prolonged duration of hospital stay, and history of previous antibiotic use, p-values < 0.05. Hospitals should strengthen infection prevention and control strategies and start antimicrobial stewardship programs.
Collapse
Affiliation(s)
- Seble Worku
- Department of Microbiology, Immunology and Parasitology, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa 1165, Ethiopia; (T.A.); (A.M.)
- Department of Medical Laboratory Science, College of Health Sciences, Debre Tabor University, Debre Tabor P.O. Box 272, Ethiopia
- Bacterial and Viral Diseases Research Directorate, Armauer Hansen Research Institute, Addis Ababa 1165, Ethiopia; (B.S.); (A.A.); (M.Y.); (A.A.); (G.T.B.)
| | - Tamrat Abebe
- Department of Microbiology, Immunology and Parasitology, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa 1165, Ethiopia; (T.A.); (A.M.)
| | - Berhanu Seyoum
- Bacterial and Viral Diseases Research Directorate, Armauer Hansen Research Institute, Addis Ababa 1165, Ethiopia; (B.S.); (A.A.); (M.Y.); (A.A.); (G.T.B.)
| | - Ashenafi Alemu
- Bacterial and Viral Diseases Research Directorate, Armauer Hansen Research Institute, Addis Ababa 1165, Ethiopia; (B.S.); (A.A.); (M.Y.); (A.A.); (G.T.B.)
| | - Yidenek Shimelash
- Debre Tabor Comprehensive Specialized Hospital, Debre Tabor P.O. Box 272, Ethiopia;
| | - Marechign Yimer
- Bacterial and Viral Diseases Research Directorate, Armauer Hansen Research Institute, Addis Ababa 1165, Ethiopia; (B.S.); (A.A.); (M.Y.); (A.A.); (G.T.B.)
| | - Alemseged Abdissa
- Bacterial and Viral Diseases Research Directorate, Armauer Hansen Research Institute, Addis Ababa 1165, Ethiopia; (B.S.); (A.A.); (M.Y.); (A.A.); (G.T.B.)
| | - Getachew Tesfaye Beyene
- Bacterial and Viral Diseases Research Directorate, Armauer Hansen Research Institute, Addis Ababa 1165, Ethiopia; (B.S.); (A.A.); (M.Y.); (A.A.); (G.T.B.)
| | - Göte Swedberg
- Department of Medical Biochemistry and Microbiology, Uppsala University, 750 08 Uppsala, Sweden;
| | - Adane Mihret
- Department of Microbiology, Immunology and Parasitology, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa 1165, Ethiopia; (T.A.); (A.M.)
- Bacterial and Viral Diseases Research Directorate, Armauer Hansen Research Institute, Addis Ababa 1165, Ethiopia; (B.S.); (A.A.); (M.Y.); (A.A.); (G.T.B.)
| |
Collapse
|
10
|
Aboelnga MM, Gauld JW. Screening a library of potential competitive inhibitors against bacterial threonyl-tRNA synthetase: DFT calculations. J Biomol Struct Dyn 2023; 42:13555-13563. [PMID: 37909495 DOI: 10.1080/07391102.2023.2276878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
Due to the growing interest in directing aminoacyl-tRNA synthetases for antimicrobial therapies, evaluating the binding proficiency of potential inhibitors against this target holds significant importance. In this work, we proposed potential ligands that could properly bind to the crucial Zn(II) cofactor located in the active site of Threonyl-tRNA synthetases (ThrRS), potentially functioning as competitive inhibitors. Initially, detailed DFT quantum chemical study was conducted to examine the binding ability of threonine against unnatural amino acids to cofactor Zn(II). Then, the binding energy value for each suggested ligand has been determined and compared to the value determined for the native substrate, threonine. Our screening investigation showed that the native threonine should coordinate in a bidentate fashion to this Zn(II) which lead to the highest (binding energy) BE Thereby, the synthetic site of ThrRS rejects unnatural amino acids that cannot afford this type of coordination to Zn(II) ion which has been supported by our calculations. Moreover, based on their binding to the Zn(II) and the obtained BE values compared to the cognate threonine, many potent ligands have been suggested. Importantly, ligands with deprotonated warheads showed the highest binding ability amongst a list of potential hits. Further investigation on the selected ligands using molecular docking and QM/MM calculations confirmed our findings of the suggested ligands being able to bind efficiently in the active site of ThrRS. The suggested hits from this study should be valuable in paving routs for developing candidates as competitive inhibitors against the bacterial ThrRS.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mohamed M Aboelnga
- Department of Chemistry, Faculty of Science, Damietta University, New Damietta, Egypt
| | - James W Gauld
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Canada
| |
Collapse
|
11
|
Jaroni D, Litt PK, Bule P, Rumbaugh K. Effectiveness of Bacteriophages against Biofilm-Forming Shiga-Toxigenic Escherichia coli In Vitro and on Food-Contact Surfaces. Foods 2023; 12:2787. [PMID: 37509879 PMCID: PMC10378794 DOI: 10.3390/foods12142787] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Background: Formation of biofilms on food-contact surfaces by Shiga-toxigenic Escherichia coli (STEC) can pose a significant challenge to the food industry, making conventional control methods insufficient. Targeted use of bacteriophages to disrupt these biofilms could reduce this problem. Previously isolated and characterized bacteriophages (n = 52) were evaluated against STEC biofilms in vitro and on food-contact surfaces. (2) Methods: Phage treatments (9 logs PFU/mL) in phosphate-buffered saline were used individually or as cocktails. Biofilms of STEC (O157, O26, O45, O103, O111, O121, and O145) were formed in 96-well micro-titer plates (7 logs CFU/mL; 24 h) or on stainless steel (SS) and high-density polyethylene (HDPE) coupons (9 logs CFU/cm2; 7 h), followed by phage treatment. Biofilm disruption was measured in vitro at 0, 3, and 6 h as a change in optical density (A595). Coupons were treated with STEC serotype-specific phage-cocktails or a 21-phage cocktail (3 phages/serotype) for 0, 3, 6, and 16 h, and surviving STEC populations were enumerated. (3) Results: Of the 52 phages, 77% showed STEC biofilm disruption in vitro. Serotype-specific phage treatments reduced pathogen population within the biofilms by 1.9-4.1 and 2.3-5.6 logs CFU/cm2, while the 21-phage cocktail reduced it by 4.0 and 4.8 logs CFU/cm2 on SS and HDPE, respectively. (4) Conclusions: Bacteriophages can be used to reduce STEC and their biofilms.
Collapse
Affiliation(s)
- Divya Jaroni
- Department of Animal and Food Sciences, and Food and Agricultural Products Center, Oklahoma State University, N. Monroe Street, Stillwater, OK 74078, USA
| | - Pushpinder Kaur Litt
- Department of Animal and Food Sciences, and Food and Agricultural Products Center, Oklahoma State University, N. Monroe Street, Stillwater, OK 74078, USA
| | - Punya Bule
- Department of Animal and Food Sciences, and Food and Agricultural Products Center, Oklahoma State University, N. Monroe Street, Stillwater, OK 74078, USA
| | - Kaylee Rumbaugh
- Department of Animal and Food Sciences, and Food and Agricultural Products Center, Oklahoma State University, N. Monroe Street, Stillwater, OK 74078, USA
| |
Collapse
|
12
|
Ayon NJ. High-Throughput Screening of Natural Product and Synthetic Molecule Libraries for Antibacterial Drug Discovery. Metabolites 2023; 13:625. [PMID: 37233666 PMCID: PMC10220967 DOI: 10.3390/metabo13050625] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/27/2023] Open
Abstract
Due to the continued emergence of resistance and a lack of new and promising antibiotics, bacterial infection has become a major public threat. High-throughput screening (HTS) allows rapid screening of a large collection of molecules for bioactivity testing and holds promise in antibacterial drug discovery. More than 50% of the antibiotics that are currently available on the market are derived from natural products. However, with the easily discoverable antibiotics being found, finding new antibiotics from natural sources has seen limited success. Finding new natural sources for antibacterial activity testing has also proven to be challenging. In addition to exploring new sources of natural products and synthetic biology, omics technology helped to study the biosynthetic machinery of existing natural sources enabling the construction of unnatural synthesizers of bioactive molecules and the identification of molecular targets of antibacterial agents. On the other hand, newer and smarter strategies have been continuously pursued to screen synthetic molecule libraries for new antibiotics and new druggable targets. Biomimetic conditions are explored to mimic the real infection model to better study the ligand-target interaction to enable the designing of more effective antibacterial drugs. This narrative review describes various traditional and contemporaneous approaches of high-throughput screening of natural products and synthetic molecule libraries for antibacterial drug discovery. It further discusses critical factors for HTS assay design, makes a general recommendation, and discusses possible alternatives to traditional HTS of natural products and synthetic molecule libraries for antibacterial drug discovery.
Collapse
Affiliation(s)
- Navid J Ayon
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
13
|
Gong WT, Zhao XJ, Wang GM, Ma XL, Huang JA. Efficacy of Zidovudine-Amikacin Combination Therapy In Vitro and in a Rat Tissue Cage Infection Model against Amikacin-Resistant, Multidrug-Resistant Enterobacteriales. Microbiol Spectr 2023; 11:e0484322. [PMID: 36946744 PMCID: PMC10101109 DOI: 10.1128/spectrum.04843-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/02/2023] [Indexed: 03/23/2023] Open
Abstract
Multidrug-resistant (MDR) Enterobacteriales infections have become an urgent global threat to public health. The aim of this study was to evaluate the efficacy of zidovudine-amikacin combination therapy in vitro and in vivo. Molecular characteristics and antibiotic resistance profiles of 53 amikacin-resistant MDR, extensively drug-resistant (XDR), or pan-drug-resistant (PDR) clinical isolates were examined via PCR and susceptibility testing. Checkerboard assays were performed for these 53 isolates to assess in vitro synergistic effects of the zidovudine-amikacin combination, and static time-kill experiments were performed for four XDR or PDR Enterobacteriales isolates. A Galleria mellonella model and a rat tissue cage infection model were established to assess in vivo synergistic effects. The aac(6')-Ib gene was detected in 25 (47.2%) isolates, followed by armA in 5 (9.4%) isolates, rmtB in 27 (50.9%) isolates, and rmtC in 3 (5.8%) isolates. Checkerboard assays showed the synergy of this combination against 38 (71.7%) isolates. The time-kill assays further confirmed that zidovudine strongly synergized with amikacin against four XDR or PDR Enterobacteriales isolates. The Galleria mellonella model study showed that the survival benefit of zidovudine-amikacin combination therapy was significantly better than that of monotherapy for those four Enterobacteriales isolates. Furthermore, the rat tissue cage infection model study showed that zidovudine-amikacin combination therapy displayed more potent bactericidal activity than monotherapy after 3 and 7 days of treatment for the above four isolates. Our data support the idea that the zidovudine-amikacin combination could be a plausible alternative therapy against infections with amikacin-resistant MDR Enterobacteriales, especially with XDR and PDR Enterobacteriales. IMPORTANCE Our study revealed for the first time that the zidovudine-amikacin combination shows a significant bactericidal effect against amikacin-resistant MDR, XDR, and PDR Enterobacteriales. Second, using in vitro and in vivo approaches, our study showed that zidovudine strongly synergized with amikacin against amikacin-resistant MDR Enterobacteriales isolates. Most importantly, with regard to survival benefit, pharmacokinetics, and bactericidal effects, our in vivo experiment demonstrated the effectiveness of zidovudine-amikacin.
Collapse
Affiliation(s)
- Wei-Tao Gong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Pulmonary and Critical Care Medicine, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiao-Jie Zhao
- Department of Laboratory Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Gao-Ming Wang
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiao-Lin Ma
- Department of Neurology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jian-An Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
14
|
Antimicrobial Activity of Some Steroidal Hydrazones. Molecules 2023; 28:molecules28031167. [PMID: 36770834 PMCID: PMC9920613 DOI: 10.3390/molecules28031167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Twelve steroid based hydrazones were in silico evaluated using computer program PASS as antimicrobial agents. The experimental evaluation revealed that all compounds have low to moderate antibacterial activity against all bacteria tested, except for B. cereus with MIC at a range of 0.37-3.00 mg/mL and MBC at 0.75-6.00 mg/mL. The most potent appeared to be compound 11 with MIC/MBC of 0.75/1.5 mg/mL, respectively. The evaluation of antibacterial activity against three resistant strains MRSA, E. coli and P. aeruginosa demonstrated superior activity of compounds against MRSA compared with ampicillin, which did not show bacteriostatic or bactericidal activities. All compounds exhibited good antifungal activity with MIC of 0.37-1.50 mg/mL and MFC of 1.50-3.00 mg/mL, but with different sensitivity against fungi tested. According to docking studies, 14-alpha demethylase inhibition may be responsible for antifungal activity. Two compounds were evaluated for their antibiofilm activity. Finally, drug-likeness and docking prediction were performed.
Collapse
|
15
|
Alkatheri AH, Yap PSX, Abushelaibi A, Lai KS, Cheng WH, Erin Lim SH. Microbial Genomics: Innovative Targets and Mechanisms. Antibiotics (Basel) 2023; 12:190. [PMID: 36830101 PMCID: PMC9951906 DOI: 10.3390/antibiotics12020190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Multidrug resistance (MDR) has become an increasing threat to global health because bacteria can develop resistance to antibiotics over time. Scientists worldwide are searching for new approaches that go beyond traditional antibiotic discovery and development pipelines. Advances in genomics, however, opened up an unexplored therapeutic opportunity for the discovery of new antibacterial agents. Genomic approaches have been used to discover several novel antibiotics that target critical processes for bacterial growth and survival, including histidine kinases (HKs), LpxC, FabI, peptide deformylase (PDF), and aminoacyl-tRNA synthetases (AaRS). In this review, we will discuss the use of microbial genomics in the search for innovative and promising drug targets as well as the mechanisms of action for novel antimicrobial agents. We will also discuss future directions on how the utilization of the microbial genomics approach could improve the odds of antibiotic development having a more successful outcome.
Collapse
Affiliation(s)
- Asma Hussain Alkatheri
- Health Sciences Division, Abu Dhabi Women’s College, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Polly Soo-Xi Yap
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor 47500, Malaysia
| | - Aisha Abushelaibi
- Office of Campus Director, Abu Dhabi Colleges, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Kok-Song Lai
- Health Sciences Division, Abu Dhabi Women’s College, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Wan-Hee Cheng
- Faculty of Health and Life Sciences, INTI International University, Persiaran Perdana BBN, Nilai 71800, Malaysia
| | - Swee-Hua Erin Lim
- Health Sciences Division, Abu Dhabi Women’s College, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| |
Collapse
|
16
|
Belakhov VV. Polyfunctional Drugs: Search, Development, Use in Medical Practice, and Environmental Aspects of Preparation and Application (A Review). RUSS J GEN CHEM+ 2022. [DOI: 10.1134/s1070363222130047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
17
|
Volynkina IA, Zakalyukina YV, Alferova VA, Belik AR, Yagoda DK, Nikandrova AA, Buyuklyan YA, Udalov AV, Golovin EV, Kryakvin MA, Lukianov DA, Biryukov MV, Sergiev PV, Dontsova OA, Osterman IA. Mechanism-Based Approach to New Antibiotic Producers Screening among Actinomycetes in the Course of the Citizen Science Project. Antibiotics (Basel) 2022; 11:antibiotics11091198. [PMID: 36139977 PMCID: PMC9495171 DOI: 10.3390/antibiotics11091198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/22/2022] Open
Abstract
Since the discovery of streptomycin, actinomycetes have been a useful source for new antibiotics, but there have been diminishing rates of new finds since the 1960s. The decreasing probability of identifying new active agents led to reduced interest in soil bacteria as a source for new antibiotics. At the same time, actinomycetes remain a promising reservoir for new active molecules. In this work, we present several reporter plasmids encoding visible fluorescent protein genes. These plasmids provide primary information about the action mechanism of antimicrobial agents at an early stage of screening. The reporters and the pipeline described have been optimized and designed to employ citizen scientists without specialized skills or equipment with the aim of essentially crowdsourcing the search for new antibiotic producers in the vast natural reservoir of soil bacteria. The combination of mechanism-based approaches and citizen science has proved its effectiveness in practice, revealing a significant increase in the screening rate. As a proof of concept, two new strains, Streptomyces sp. KB-1 and BV113, were found to produce the antibiotics pikromycin and chartreusin, respectively, demonstrating the efficiency of the pipeline.
Collapse
Affiliation(s)
- Inna A. Volynkina
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, 121205 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
- Correspondence: (I.A.V.); (I.A.O.)
| | - Yuliya V. Zakalyukina
- Center for Translational Medicine, Sirius University of Science and Technology, Olympic Avenue 1, 354340 Sochi, Russia
- Department of Soil Science, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
| | - Vera A. Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia
| | - Albina R. Belik
- Center for Translational Medicine, Sirius University of Science and Technology, Olympic Avenue 1, 354340 Sochi, Russia
| | - Daria K. Yagoda
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
| | - Arina A. Nikandrova
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, 121205 Moscow, Russia
| | - Yuliya A. Buyuklyan
- Center for Translational Medicine, Sirius University of Science and Technology, Olympic Avenue 1, 354340 Sochi, Russia
| | - Andrei V. Udalov
- Center for Translational Medicine, Sirius University of Science and Technology, Olympic Avenue 1, 354340 Sochi, Russia
| | - Evgenii V. Golovin
- Center for Translational Medicine, Sirius University of Science and Technology, Olympic Avenue 1, 354340 Sochi, Russia
| | - Maxim A. Kryakvin
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
| | - Dmitrii A. Lukianov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, 121205 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
| | - Mikhail V. Biryukov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, 121205 Moscow, Russia
- Center for Translational Medicine, Sirius University of Science and Technology, Olympic Avenue 1, 354340 Sochi, Russia
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
| | - Petr V. Sergiev
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, 121205 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
| | - Olga A. Dontsova
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, 121205 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Ilya A. Osterman
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, 121205 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
- Center for Translational Medicine, Sirius University of Science and Technology, Olympic Avenue 1, 354340 Sochi, Russia
- Correspondence: (I.A.V.); (I.A.O.)
| |
Collapse
|
18
|
Xiang YL, Huang SH, Hu QH, Wang QY, Zhao MQ, Jiang YC, Chen X, Lin J, Zhou QH. Zwitterionic meso-silica/polypeptide hybrid nanoparticles for efficient azithromycin delivery and photodynamic therapy for synergistic treatment of drug-resistant bacterial infection. Int J Biol Macromol 2022; 219:597-610. [PMID: 35952811 DOI: 10.1016/j.ijbiomac.2022.08.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/22/2022] [Accepted: 08/05/2022] [Indexed: 12/21/2022]
Abstract
The treatment of drug-resistant bacterial infections attributed to the overuse of antibiotics still remains a serious challenge globally. Herein, zwitterionic charge switchable meso-silica/polypeptide hybrid nanoparticles (MSPNs) were prepared for the synergistic chemo-photodynamic therapy in the treatment of drug-resistant bacterial infections. Subsequently, azithromycin (AZT) and methylene blue (MB) were loaded in the MSPNs to form the combined chemo-photodynamic therapeutic nanoparticles (MSPNs-AZT/MB) for the treatment of methicillin-resistant Staphylococcus aureus (MRSA). Remarkably, the as-prepared MSPNs-AZT/MB exhibited a negative surface charge of -5.2 mV at physiological pH while switching into positive surface charge of 24.7 mv in an acidic environment, leading to enhanced binding with bacterial surface. The lipase-triggered AZT release up to 77.9 % was achieved, and the loaded MB demonstrated efficient singlet oxygen (1O2) generation for photodynamic therapy. The in vitro experimental results displayed an excellent antibacterial effect against MRSA in both planktonic and biofilm phenotypes. Additionally, the as-prepared MSPNs-AZT/MB exhibited synergistic and enhanced antibacterial infection effect up to 94 % comparing to monotherapy in a mice model. Considering the above advantages, the as-prepared combined chemo-photodynamic therapeutic nanoparticles showed promising biocompatibility and clinical potential for the efficient therapy of drug-resistant bacteria.
Collapse
Affiliation(s)
- Ya-Li Xiang
- Key Laboratory of Pollution Control Chemistry and Environmental Functional Materials for Qinghai-Tibet Plateau of the National Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, First Ring Road, 4th Section No.16, Chengdu, Sichuan 610041, China
| | - Shuang-Hui Huang
- Key Laboratory of Pollution Control Chemistry and Environmental Functional Materials for Qinghai-Tibet Plateau of the National Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, First Ring Road, 4th Section No.16, Chengdu, Sichuan 610041, China
| | - Qiu-Hui Hu
- Key Laboratory of Pollution Control Chemistry and Environmental Functional Materials for Qinghai-Tibet Plateau of the National Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, First Ring Road, 4th Section No.16, Chengdu, Sichuan 610041, China
| | - Qiu-Yue Wang
- Key Laboratory of Pollution Control Chemistry and Environmental Functional Materials for Qinghai-Tibet Plateau of the National Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, First Ring Road, 4th Section No.16, Chengdu, Sichuan 610041, China
| | - Mei-Qi Zhao
- Key Laboratory of Pollution Control Chemistry and Environmental Functional Materials for Qinghai-Tibet Plateau of the National Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, First Ring Road, 4th Section No.16, Chengdu, Sichuan 610041, China
| | - Yu-Chen Jiang
- Key Laboratory of Pollution Control Chemistry and Environmental Functional Materials for Qinghai-Tibet Plateau of the National Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, First Ring Road, 4th Section No.16, Chengdu, Sichuan 610041, China
| | - Xiao Chen
- Key Laboratory of Pollution Control Chemistry and Environmental Functional Materials for Qinghai-Tibet Plateau of the National Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, First Ring Road, 4th Section No.16, Chengdu, Sichuan 610041, China
| | - Juan Lin
- School of Biomedical Sciences and Technology, Chengdu Medical College, Xindu Road No.783, Chengdu, Sichuan 610500, China.
| | - Qing-Han Zhou
- Key Laboratory of Pollution Control Chemistry and Environmental Functional Materials for Qinghai-Tibet Plateau of the National Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, First Ring Road, 4th Section No.16, Chengdu, Sichuan 610041, China; Key Laboratory of General Chemistry of the National Ethnic Affairs Commission, School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, China.
| |
Collapse
|
19
|
In silico Methods for Identification of Potential Therapeutic Targets. Interdiscip Sci 2022; 14:285-310. [PMID: 34826045 PMCID: PMC8616973 DOI: 10.1007/s12539-021-00491-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 11/01/2022]
Abstract
AbstractAt the initial stage of drug discovery, identifying novel targets with maximal efficacy and minimal side effects can improve the success rate and portfolio value of drug discovery projects while simultaneously reducing cycle time and cost. However, harnessing the full potential of big data to narrow the range of plausible targets through existing computational methods remains a key issue in this field. This paper reviews two categories of in silico methods—comparative genomics and network-based methods—for finding potential therapeutic targets among cellular functions based on understanding their related biological processes. In addition to describing the principles, databases, software, and applications, we discuss some recent studies and prospects of the methods. While comparative genomics is mostly applied to infectious diseases, network-based methods can be applied to infectious and non-infectious diseases. Nonetheless, the methods often complement each other in their advantages and disadvantages. The information reported here guides toward improving the application of big data-driven computational methods for therapeutic target discovery.
Graphical abstract
Collapse
|
20
|
Guan S, Fu N. Class imbalance learning with Bayesian optimization applied in drug discovery. Sci Rep 2022; 12:2069. [PMID: 35136094 PMCID: PMC8827090 DOI: 10.1038/s41598-022-05717-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/11/2022] [Indexed: 11/12/2022] Open
Abstract
Machine intelligence (MI), including machine learning and deep learning, have been regarded as promising methods to reduce the prohibitively high cost of drug development. However, a dilemma within MI has limited its wide application: machine learning models are easier to interpret but yield worse predictive performance than deep learning models. Therefore, we propose a pipeline called Class Imbalance Learning with Bayesian Optimization (CILBO) to improve the performance of machine learning models in drug discovery. To demonstrate the efficacy of the CILBO pipeline, we developed an example model to predict antibacterial candidates. Comparison of the antibacterial prediction performance between our model and a well-known deep learning model published by Stokes et al. suggests that our model can perform as well as the deep learning model in drug activity prediction. The CILBO pipeline we propose provides a simple, alternative approach to accelerate preliminary screenings and decrease the cost of drug discovery.
Collapse
Affiliation(s)
- Shenmin Guan
- Shanghai GenomSeqCare Biotechnology Co. Ltd., Shanghai, 200052, China.
| | - Ning Fu
- Shanghai GenomSeqCare Biotechnology Co. Ltd., Shanghai, 200052, China
| |
Collapse
|
21
|
The Effect of the Use of Essential Oils in the Feed of Bee Families on Honey Chemical Composition and Antimicrobial Activity. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Honey is a natural food with pharmacological properties. The present study was focused on the use of essential oils in the supplementary feeding of bee families for three weeks (spring). The purpose was to monitor the effect of essential oils (basil, thyme, juniper, cloves, mint, cinnamon, oregano, rosemary) on the chemical components of the resulting honey. The honey sampling period was carried out before the administration of essential oils in the supplementary feed of the bee families, after the administration of essential oils in the bee feed, respectively, after the first harvest (rapeseed). The honey samples were subjected to chemical analysis to determine humidity, impurities, ash, pH, acidity, total phenolic content (TPC) and flavonoid content (FC), reducing sugar content and antioxidant activity. In addition, the antimicrobial activity against nine strains was tested. We found out that all the essential oils used had a positive effect on the chemical composition of honey, especially the essential oil of oregano, mint, thyme, cinnamon. Experimental variants in which juniper and clove essential oil were introduced reduced the acidity of honey by 28.12% and 35.48%, respectively. Ash content varies between 0.23% and 0.46%, impurities content between 4.11% and 9.11%, while the values for pH were between 3.42 and 4.03. As for the TPC, they have increased considerably in all experimental variants to which essential oil has been added, compared to the batch fed only with sugar syrup, the highest value being recorded for the sample treated with cinnamon after the third harvest (163.94 mg/100 g). The FC values vary between 8.41–44.36 mg/100 g, depending of the treatment applied and the period of harvesting. Regarding the antimicrobial activity, the results highlighted that the essential oils present in the diet of bees produced honey with antimicrobial effect increased after two weeks after administration.
Collapse
|
22
|
Ashfaq M, Talreja N, Chauhan D, Viswanathan MR. Synthesis of Cu-doped 2D-WS 2 nanosheet-based nano-antibiotic materials for inhibiting E. Coli and S. aureus bacterial strains. NEW J CHEM 2022. [DOI: 10.1039/d1nj05931a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A new class of nano-antibiotic materials must be developed to treat bacterial infectious diseases. In this context, the synthesizing Cu-metal incorporated WS2 nanosheet (Cu–WS2-NS)-based antibiotic materials might overcome such associated issues.
Collapse
Affiliation(s)
- Mohammad Ashfaq
- Advanced Ceramics and Nanotechnology Laboratory, Department of Materials Engineering, Faculty of Engineering, University of Concepción, Concepción 4070409, Chile
- School of Life Science, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
| | - Neetu Talreja
- Advanced Ceramics and Nanotechnology Laboratory, Department of Materials Engineering, Faculty of Engineering, University of Concepción, Concepción 4070409, Chile
| | - Divya Chauhan
- Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL, USA
| | - Mangalaraja Ramalinga Viswanathan
- Advanced Ceramics and Nanotechnology Laboratory, Department of Materials Engineering, Faculty of Engineering, University of Concepción, Concepción 4070409, Chile
- Technological Development, Unit (UDT) University of Concepcion, Coronel Industrial Park, Coronel, Chile
| |
Collapse
|
23
|
Egbujor MC, Okoro UC, Okafor SN, Egu SA, Amasiatu IS, Egwuatu PI, Umeh OR, Ibo EM. Design, synthesis, and molecular docking of cysteine-based sulphonamide derivatives as antimicrobial agents. Res Pharm Sci 2021; 17:99-110. [PMID: 34909048 PMCID: PMC8621839 DOI: 10.4103/1735-5362.329930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 04/14/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
Background and purpose: The preponderance of microbial infections remains a global challenge. In the present study, synthesis of novel cysteine-based antimicrobial agents and their biological evaluation is reported. Experimental approach: The reaction of p-toluenesulphonyl chloride with cysteine afforded 2-{[(4-methylphenyl)sulphonyl]amino}-3-sulphanylpropanoic acid (3) which was acetylated based on Lumiere-Barbier method using acetic anhydride. The ammonolysis of the acetylated compound (4) gave the carboxamide derivative (5) which reacted with aniline, aminopyridine and diaminopyrimidine via nickel catalyzed Buchwald-Hartwig amidation reaction to afford compounds 6a, 6b, and 6c, respectively. The compounds were characterized using FTIR, 1H-NMR, 13C-NMR, and elemental analysis. The in vitro antimicrobial activities were determined. Their physicochemical properties were generated in silico and the molecular docking studied bacterial and fungal infections. Findings/Results: Compounds 4, 6b, and 6c exhibited excellent in vitro antibacterial activities while compound 4 had the best antifungal activities. From the in silico antimicrobial results, compound 3 had a better binding affinity (-10.95 kcal/mol) than penicillin (-10.89 kcal/mol) while compounds 3 and 4 had binding affinities (-10.07 and -10.62kcal/mol) comparable to ketoconazole (-10.85 kcal/mol). Conclusion and implication: All the synthesized compounds exhibited significant antibacterial and antifungal activities and were confirmed to be potential antimicrobial agents.
Collapse
Affiliation(s)
- Melford C Egbujor
- Department of Industrial Chemistry, Renaissance University, Ugbawka, Enugu State, Nigeria
| | - Uchechukwu C Okoro
- Synthetic Organic Chemistry Division, Department of Pure and Industrial Chemistry, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Sunday N Okafor
- Department of Pharmaceutical and Medicinal Chemistry, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Samuel A Egu
- Department of Pure and Industrial Chemistry, Kogi State University, Anyigba, Kogi State, Nigeria
| | - Ifeanyi S Amasiatu
- Department of Biochemistry, Renaissance University, Ugbawka, Enugu State, Nigeria
| | - Pius I Egwuatu
- Department of Microbiology, Renaissance University, Ugbawka, Enugu State, Nigeria
| | - Odera R Umeh
- Department of Applied Microbiology and Brewing, Nnamdi Azikiwe University, P.M.B 5025 Awka, Anambra State, Nigeria
| | - Eziafakaego M Ibo
- Department of Applied Microbiology and Brewing, Nnamdi Azikiwe University, P.M.B 5025 Awka, Anambra State, Nigeria
| |
Collapse
|
24
|
Tang XD, Ji TT, Dong JR, Feng H, Chen FQ, Chen X, Zhao HY, Chen DK, Ma WT. Pathogenesis and Treatment of Cytokine Storm Induced by Infectious Diseases. Int J Mol Sci 2021; 22:13009. [PMID: 34884813 PMCID: PMC8658039 DOI: 10.3390/ijms222313009] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Cytokine storm is a phenomenon characterized by strong elevated circulating cytokines that most often occur after an overreactive immune system is activated by an acute systemic infection. A variety of cells participate in cytokine storm induction and progression, with profiles of cytokines released during cytokine storm varying from disease to disease. This review focuses on pathophysiological mechanisms underlying cytokine storm induction and progression induced by pathogenic invasive infectious diseases. Strategies for targeted treatment of various types of infection-induced cytokine storms are described from both host and pathogen perspectives. In summary, current studies indicate that cytokine storm-targeted therapies can effectively alleviate tissue damage while promoting the clearance of invading pathogens. Based on this premise, "multi-omics" immune system profiling should facilitate the development of more effective therapeutic strategies to alleviate cytokine storms caused by various diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - De-Kun Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China; (X.-D.T.); (T.-T.J.); (J.-R.D.); (H.F.); (F.-Q.C.); (X.C.); (H.-Y.Z.)
| | - Wen-Tao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China; (X.-D.T.); (T.-T.J.); (J.-R.D.); (H.F.); (F.-Q.C.); (X.C.); (H.-Y.Z.)
| |
Collapse
|
25
|
Mubeen B, Ansar AN, Rasool R, Ullah I, Imam SS, Alshehri S, Ghoneim MM, Alzarea SI, Nadeem MS, Kazmi I. Nanotechnology as a Novel Approach in Combating Microbes Providing an Alternative to Antibiotics. Antibiotics (Basel) 2021; 10:1473. [PMID: 34943685 PMCID: PMC8698349 DOI: 10.3390/antibiotics10121473] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/19/2021] [Accepted: 11/25/2021] [Indexed: 12/15/2022] Open
Abstract
The emergence of infectious diseases promises to be one of the leading mortality factors in the healthcare sector. Although several drugs are available on the market, newly found microorganisms carrying multidrug resistance (MDR) against which existing drugs cannot function effectively, giving rise to escalated antibiotic dosage therapies and the need to develop novel drugs, which require time, money, and manpower. Thus, the exploitation of antimicrobials has led to the production of MDR bacteria, and their prevalence and growth are a major concern. Novel approaches to prevent antimicrobial drug resistance are in practice. Nanotechnology-based innovation provides physicians and patients the opportunity to overcome the crisis of drug resistance. Nanoparticles have promising potential in the healthcare sector. Recently, nanoparticles have been designed to address pathogenic microorganisms. A multitude of processes that can vary with various traits, including size, morphology, electrical charge, and surface coatings, allow researchers to develop novel composite antimicrobial substances for use in different applications performing antimicrobial activities. The antimicrobial activity of inorganic and carbon-based nanoparticles can be applied to various research, medical, and industrial uses in the future and offer a solution to the crisis of antimicrobial resistance to traditional approaches. Metal-based nanoparticles have also been extensively studied for many biomedical applications. In addition to reduced size and selectivity for bacteria, metal-based nanoparticles have proven effective against pathogens listed as a priority, according to the World Health Organization (WHO). Moreover, antimicrobial studies of nanoparticles were carried out not only in vitro but in vivo as well in order to investigate their efficacy. In addition, nanomaterials provide numerous opportunities for infection prevention, diagnosis, treatment, and biofilm control. This study emphasizes the antimicrobial effects of nanoparticles and contrasts nanoparticles' with antibiotics' role in the fight against pathogenic microorganisms. Future prospects revolve around developing new strategies and products to prevent, control, and treat microbial infections in humans and other animals, including viral infections seen in the current pandemic scenarios.
Collapse
Affiliation(s)
- Bismillah Mubeen
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan; (B.M.); (A.N.A.); (R.R.); (I.U.)
| | - Aunza Nayab Ansar
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan; (B.M.); (A.N.A.); (R.R.); (I.U.)
| | - Rabia Rasool
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan; (B.M.); (A.N.A.); (R.R.); (I.U.)
| | - Inam Ullah
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan; (B.M.); (A.N.A.); (R.R.); (I.U.)
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.S.I.); (S.A.)
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.S.I.); (S.A.)
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia;
| | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
26
|
Singh SS, Sharma D, Baindara P, Choksket S, Harshvardhan, Mandal SM, Grover V, Korpole S. Characterization and Antimicrobial Studies of Iturin-Like and Bogorol-Like Lipopeptides From Brevibacillus spp. Strains GI9 and SKDU10. Front Microbiol 2021; 12:729026. [PMID: 34782829 PMCID: PMC8589628 DOI: 10.3389/fmicb.2021.729026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/05/2021] [Indexed: 11/30/2022] Open
Abstract
Accession numbers for whole-genome sequence of Brevibacillus sp. strain GI9 and SKDU10 are CAGD01000001 to CAGD01000061 and LSSO00000000, respectively. Members of the genus Brevibacillus have been demonstrated to produce a variety of bioactive compounds including polyketides, lipopeptides and bacteriocins. Lipopeptides are non-ribosomally synthesized surface-active compounds with antimicrobial, antitumor, and immune-stimulatory activities. They usually exhibit strong antifungal and antibacterial activities and are considered as promising compounds in controlling fungal diseases. In this study, we have characterized two lipopeptides from Brevibacillus sp. strains GI9 and SKDU10. The corresponding lipopeptides were purified by reverse-phase high-performance liquid chromatography. Mass analysis and characterization by MALDI-TOF-MS (Matrix-assisted laser desorption ionization time-of-flight mass spectrometry) analysis revealed production of an iturin-like lipopeptide by strain GI9 and bogorol-like lipopeptide by strain SKDU10. Both lipopeptides exhibited broad spectrum antibacterial activity and inhibited the growth of various fungi. They showed minimum inhibitory concentration (MIC) values between 90 and 300 μg/ml against indicator strains of bacteria and drug-resistant Candida indicator strains. The lipopeptides did not show phytotoxic effect in seed germination experiments but caused hemolysis. Further, both lipopeptides inhibited the growth of fungi on fruits and vegetables in in vitro experiments, thereby exhibited potential use in biotechnological industry as effective biocontrol agents.
Collapse
Affiliation(s)
| | - Deepika Sharma
- CSIR-Institute of Microbial Technology, Chandigarh, India
| | | | | | - Harshvardhan
- CSIR-Institute of Microbial Technology, Chandigarh, India
| | | | - Vishakha Grover
- Dr. Harvansh Singh Judge Institute of Dental Sciences and Hospital, Panjab University, Chandigarh, India
| | - Suresh Korpole
- CSIR-Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
27
|
Abd El-Aleam RH, George RF, Georgey HH, Abdel-Rahman HM. Bacterial virulence factors: a target for heterocyclic compounds to combat bacterial resistance. RSC Adv 2021; 11:36459-36482. [PMID: 35494393 PMCID: PMC9043591 DOI: 10.1039/d1ra06238g] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/01/2021] [Indexed: 12/17/2022] Open
Abstract
Antibiotic resistance is one of the most important challenges of the 21st century. However, the growing understanding of bacterial pathogenesis and cell-to-cell communication has revealed many potential strategies for the discovery of drugs that can be used for the treatment of bacterial infections. Interfering with bacterial virulence and/or quorum sensing could be a particularly interesting approach, because it is believed to exert less selective pressure on the bacterial resistance than with traditional strategies, geared toward killing bacteria or preventing their growth. Here, we discuss the mechanism of bacterial virulence, presenting promising strategies and recently synthesized heterocyclic compounds to combat future bacterial infections.
Collapse
Affiliation(s)
- Rehab H Abd El-Aleam
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information MTI Cairo 11571 Egypt
| | - Riham F George
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University Cairo 11562 Egypt
| | - Hanan H Georgey
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University Cairo 11562 Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University Cairo 11786 Egypt
| | - Hamdy M Abdel-Rahman
- Medicinal Chemistry Department, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Nahda University Beni Suef Egypt
| |
Collapse
|
28
|
Banoub NG, Saleh SE, Helal HS, Aboshanab KM. Antibiotics Combinations and Chitosan Nanoparticles for Combating Multidrug Resistance Acinetobacter baumannii. Infect Drug Resist 2021; 14:3327-3339. [PMID: 34447258 PMCID: PMC8384262 DOI: 10.2147/idr.s328788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022] Open
Abstract
Background Successful treatment of Acinetobacter (A.) baumannii-associated infection is complicated by the emergence of multidrug resistance (MDR), particularly in clinical settings. This urges searching for new alternatives to encounter such health problem. Aim This study aimed to evaluate certain antibiotic combinations and CNPs either alone or in combination of some selected antibiotics for the purpose of combating MDR A. baumannii clinical isolates. Methods A total of 51 A. baumannii clinical isolates were recovered from discharged clinical specimens of the Clinical Microbiology Central Laboratory of AL Kasr Al Aini hospital, Cairo, Egypt. Conventional standard Lab tests were used for identification followed by recA gene testing for confirmation. Antimicrobial susceptibility tests were conducted out according to CLSI guidelines. Genotypic analysis using Enterobacterial Repetitive Intergenic Consensus-polymerase chain reaction (ERIC-PCR) of the respective isolates showed that they were clustered in nine clones. The prepared CNPs were characterized by dynamic light scattering and HR-transmission electron microscope imaging. Antibiotic combinations and co-effect of CNPs with some selected antibiotics (either each alone or in combination of two) were evaluated using the Checkerboard microdilution and minimum inhibitor concentration decrease factor (MDF) methods, respectively. Results The recovered 51 A. baumannii clinical isolates were MDR (100%) of these 92% (47/51) were extensively drug resistance (XDR). Combinations of colistin (CT)+meropenem (MEM) and MEM+tigecycline (TGC) showed synergism in 77.7% and 44.4% and additive effects in 22.3% and 55.6% of the tested MDR A. baumannii isolates (n=51), respectively. However, CT+TGC combination showed antagonism. CNPs exhibited good inhibitory activity (inhibition zones ranged from 24 to 31 mm) against selected nine MDR A. baumannii isolates (one isolate from each clone). The MIC of CNPs at concentrations (ranging from 1 to 5 mg/mL) were from 0.16 to 0.25 mg/mL, indicating good in vitro antimicrobial activities. CNPs (5 mg/mL) when combined with CT, TGC or MEM, CT+MEM and TGC+MEM significantly increased the susceptibilities of the MDR A. baumannii isolates to these antibiotics by 88.8%, 66.6%, 100%, 77.7%, and 44.4%, respectively. No significant effects were observed when CNPs (5 mg/mL) were combined with CT+TGC. Conclusion The current study demonstrated the significant in-vitro activities of CNPs either alone or in combination with CT, TGC or MEM, CT+MEM and TGC+MEM and the successful combinations of MEM either with CT or with TGC against the MDR A. baumannii pathogens. However, further in vivo studies should be conducted to verify such activities and their potential use in human.
Collapse
Affiliation(s)
- Nancy G Banoub
- Department of Microbiology and Immunology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Sarra E Saleh
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Hala S Helal
- Department of Microbiology and Immunology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Khaled M Aboshanab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
29
|
Hu XL, Shang Y, Yan KC, Sedgwick AC, Gan HQ, Chen GR, He XP, James TD, Chen D. Low-dimensional nanomaterials for antibacterial applications. J Mater Chem B 2021; 9:3640-3661. [PMID: 33870985 DOI: 10.1039/d1tb00033k] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The excessive use of antibiotics has led to a rise in drug-resistant bacteria. These "superbugs" are continuously emerging and becoming increasingly harder to treat. As a result, new and effective treatment protocols that have minimal risks of generating drug-resistant bacteria are urgently required. Advanced nanomaterials are particularly promising due to their drug loading/releasing capabilities combined with their potential photodynamic/photothermal therapeutic properties. In this review, 0-dimensional, 1-dimensional, 2-dimensional, and 3-dimensional nanomaterial-based systems are comprehensively discussed for bacterial-based diagnostic and treatment applications. Since the use of these platforms as antibacterials is relatively new, this review will provide appropriate insight into their construction and applications. As such, we hope this review will inspire researchers to explore antibacterial-based nanomaterials with the aim of developing systems for clinical applications.
Collapse
Affiliation(s)
- Xi-Le Hu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Ying Shang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Kai-Cheng Yan
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| | - Adam C Sedgwick
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, USA
| | - Hui-Qi Gan
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Guo-Rong Chen
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Tony D James
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, UK. and School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Daijie Chen
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, China.
| |
Collapse
|
30
|
Gardouh AR, Srag El-Din ASG, Salem MSH, Moustafa Y, Gad S. Starch Nanoparticles for Enhancement of Oral Bioavailability of a Newly Synthesized Thienopyrimidine Derivative with Anti-Proliferative Activity Against Pancreatic Cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3071-3093. [PMID: 34305395 PMCID: PMC8292977 DOI: 10.2147/dddt.s321962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/08/2021] [Indexed: 01/02/2023]
Abstract
Purpose This research aimed to improve water solubility and oral bioavailability of a newly synthesized thienopyrimidine derivative (TPD) with anti-pancreatic cancer activity by loading on starch nanoparticles (SNPs). Methods TPD was synthesized, purified and its ADME behavior was predicted using Swiss ADME software. A UV spectroscopy method was developed and validated to measure TPD concentration at various dosage forms. SNPs loaded with TPD (SNPs-TPD) were prepared, characterized for particle size, polydispersity index, zeta potential, transmission electron microscopy, Fourier transform infrared spectroscopy (FT-IR), differential scanning calorimetry (DSC), entrapment efficiency, in-vitro release, and in-vivo animal study. Results The Swiss ADME results showed that TPD can be administered orally; however, it has low oral bioavailability (0.55) and poor water solubility. The significant regression coefficient of the calibration curve (r2 = 0.9995), the precision (%RSD < 0.5%) and the accuracy (99.46−101.72%) confirmed the efficacy of the developed UV method. SNPs-TPD had a spherical monodispersed (PDI= 0.12) shape, nanoparticle size (22.98 ± 4.23) and good stability (−21 ± 4.72 mV). Moreover, FT-IR and DSC revealed changes in the physicochemical structure of starch resulting in SNPs formation. The entrapment efficiency was 97% ± 0.45%, and the in-vitro release showed that the SNPs enhanced the solubility of the TPD. The in-vivo animal study and histopathology showed that SNPs enhanced the oral bioavailability of TPD against solid Ehrlich carcinoma. Conclusion SNPs-TPD were superior in drug solubility and oral bioavailability than those obtained from TPD suspension.
Collapse
Affiliation(s)
- Ahmed R Gardouh
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.,Department of Pharmaceutical Sciences, Faculty of Pharmacy, Jadara University, Irbid, 21110, Jordan
| | - Ahmed S G Srag El-Din
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science & Technology, Gamasa City, Egypt
| | - Mohamed S H Salem
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.,The Institute of Scientific and Industrial Research (ISIR), Osaka University, Ibaraki-shi, Osaka, 567-0047, Japan
| | - Yasser Moustafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Shadeed Gad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
31
|
Hassett DJ, Kovall RA, Schurr MJ, Kotagiri N, Kumari H, Satish L. The Bactericidal Tandem Drug, AB569: How to Eradicate Antibiotic-Resistant Biofilm Pseudomonas aeruginosa in Multiple Disease Settings Including Cystic Fibrosis, Burns/Wounds and Urinary Tract Infections. Front Microbiol 2021; 12:639362. [PMID: 34220733 PMCID: PMC8245851 DOI: 10.3389/fmicb.2021.639362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
The life-threatening pandemic concerning multi-drug resistant (MDR) bacteria is an evolving problem involving increased hospitalizations, billions of dollars in medical costs and a remarkably high number of deaths. Bacterial pathogens have demonstrated the capacity for spontaneous or acquired antibiotic resistance and there is virtually no pool of organisms that have not evolved such potentially clinically catastrophic properties. Although many diseases are linked to such organisms, three include cystic fibrosis (CF), burn/blast wounds and urinary tract infections (UTIs), respectively. Thus, there is a critical need to develop novel, effective antimicrobials for the prevention and treatment of such problematic infections. One of the most formidable, naturally MDR bacterial pathogens is Pseudomonas aeruginosa (PA) that is particularly susceptible to nitric oxide (NO), a component of our innate immune response. This susceptibility sets the translational stage for the use of NO-based therapeutics during the aforementioned human infections. First, we discuss how such NO therapeutics may be able to target problematic infections in each of the aforementioned infectious scenarios. Second, we describe a recent discovery based on years of foundational information, a novel drug known as AB569. AB569 is capable of forming a "time release" of NO from S-nitrosothiols (RSNO). AB569, a bactericidal tandem consisting of acidified NaNO2 (A-NO2 -) and Na2-EDTA, is capable of killing all pathogens that are associated with the aforementioned disorders. Third, we described each disease state in brief, the known or predicted effects of AB569 on the viability of PA, its potential toxicity and highly remote possibility for resistance to develop. Finally, we conclude that AB569 can be a viable alternative or addition to conventional antibiotic regimens to treat such highly problematic MDR bacterial infections for civilian and military populations, as well as the economical burden that such organisms pose.
Collapse
Affiliation(s)
- Daniel J Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, Cincinnati, OH, United States
| | - Rhett A Kovall
- Department of Molecular Genetics, Biochemistry and Microbiology, Cincinnati, OH, United States
| | - Michael J Schurr
- Department of Immunology and Microbiology, University of Colorado Health Sciences, Denver, CO, United States
| | - Nalinikanth Kotagiri
- Division of Pharmacy, University of Colorado Health Sciences, Denver, CO, United States
| | - Harshita Kumari
- Division of Pharmacy, University of Colorado Health Sciences, Denver, CO, United States
| | - Latha Satish
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Shriners Hospitals for Children-Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
32
|
Assaf R, Xia F, Stevens R. Identifying genomic islands with deep neural networks. BMC Genomics 2021; 22:281. [PMID: 34078279 PMCID: PMC8170982 DOI: 10.1186/s12864-021-07575-5] [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: 03/07/2021] [Accepted: 03/31/2021] [Indexed: 12/05/2022] Open
Abstract
Background Horizontal gene transfer is the main source of adaptability for bacteria, through which genes are obtained from different sources including bacteria, archaea, viruses, and eukaryotes. This process promotes the rapid spread of genetic information across lineages, typically in the form of clusters of genes referred to as genomic islands (GIs). Different types of GIs exist, and are often classified by the content of their cargo genes or their means of integration and mobility. While various computational methods have been devised to detect different types of GIs, no single method is capable of detecting all types. Results We propose a method, which we call Shutter Island, that uses a deep learning model (Inception V3, widely used in computer vision) to detect genomic islands. The intrinsic value of deep learning methods lies in their ability to generalize. Via a technique called transfer learning, the model is pre-trained on a large generic dataset and then re-trained on images that we generate to represent genomic fragments. We demonstrate that this image-based approach generalizes better than the existing tools. Conclusions We used a deep neural network and an image-based approach to detect the most out of the correct GI predictions made by other tools, in addition to making novel GI predictions. The fact that the deep neural network was re-trained on only a limited number of GI datasets and then successfully generalized indicates that this approach could be applied to other problems in the field where data is still lacking or hard to curate.
Collapse
Affiliation(s)
- Rida Assaf
- Department of Computer Science, University of Chicago, S. Ellis Ave., Chicago, 60637, USA.
| | - Fangfang Xia
- Computing Environment and Life Sciences Division, Argonne National Laboratory, S. Cass Ave., Lemont, 60439, USA.,Data Science and Learning Division, Argonne National Laboratory, S. Cass Ave., Lemont, 60439, USA
| | - Rick Stevens
- Computing Environment and Life Sciences Division, Argonne National Laboratory, S. Cass Ave., Lemont, 60439, USA.,The University of Chicago Consortium for Advanced Science and Engineering, University of Chicago, S. Ellis Ave., Chicago, 60637, USA
| |
Collapse
|
33
|
Multisubstituted pyrimidines effectively inhibit bacterial growth and biofilm formation of Staphylococcus aureus. Sci Rep 2021; 11:7931. [PMID: 33846401 PMCID: PMC8041844 DOI: 10.1038/s41598-021-86852-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 03/22/2021] [Indexed: 11/25/2022] Open
Abstract
Biofilms are multicellular communities of microorganisms that generally attach to surfaces in a self-produced matrix. Unlike planktonic cells, biofilms can withstand conventional antibiotics, causing significant challenges in the healthcare system. Currently, new chemical entities are urgently needed to develop novel anti-biofilm agents. In this study, we designed and synthesized a set of 2,4,5,6-tetrasubstituted pyrimidines and assessed their antibacterial activity against planktonic cells and biofilms formed by Staphylococcus aureus. Compounds 9e, 10d, and 10e displayed potent activity for inhibiting the onset of biofilm formation as well as for killing pre-formed biofilms of S. aureus ATCC 25923 and Newman strains, with half-maximal inhibitory concentration (IC50) values ranging from 11.6 to 62.0 µM. These pyrimidines, at 100 µM, not only decreased the number of viable bacteria within the pre-formed biofilm by 2–3 log10 but also reduced the amount of total biomass by 30–50%. Furthermore, these compounds were effective against planktonic cells with minimum inhibitory concentration (MIC) values lower than 60 µM for both staphylococcal strains. Compound 10d inhibited the growth of S. aureus ATCC 25923 in a concentration-dependent manner and displayed a bactericidal anti-staphylococcal activity. Taken together, our study highlights the value of multisubstituted pyrimidines to develop novel anti-biofilm agents.
Collapse
|
34
|
Purification and characterization of bacteriocins-like inhibitory substances from food isolated Enterococcus faecalis OS13 with activity against nosocomial enterococci. Sci Rep 2021; 11:3795. [PMID: 33589735 PMCID: PMC7884432 DOI: 10.1038/s41598-021-83357-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 01/25/2021] [Indexed: 01/02/2023] Open
Abstract
Nosocomial infections caused by enterococci are an ongoing global threat. Thus, finding therapeutic agents for the treatment of such infections are crucial. Some Enterococcus faecalis strains are able to produce antimicrobial peptides called bacteriocins. We analyzed 65 E. faecalis isolates from 43 food samples and 22 clinical samples in Egypt for 17 common bacteriocin-encoding genes of Enterococcus spp. These genes were absent in 11 isolates that showed antimicrobial activity putatively due to bacteriocins (three from food, including isolate OS13, and eight from clinical isolates). The food-isolated E. faecalis OS13 produced bacteriocin-like inhibitory substances (BLIS) named enterocin OS13, which comprised two peptides (enterocin OS13α OS13β) that inhibited the growth of antibiotic-resistant nosocomial E. faecalis and E. faecium isolates. The molecular weights of enterocin OS13α and OS13β were determined as 8079 Da and 7859 Da, respectively, and both were heat-labile. Enterocin OS13α was sensitive to proteinase K, while enterocin OS13β was resistant. Characterization of E. faecalis OS13 isolate revealed that it belonged to sequence type 116. It was non-hemolytic, bile salt hydrolase-negative, gelatinase-positive, and sensitive to ampicillin, penicillin, vancomycin, erythromycin, kanamycin, and gentamicin. In conclusion, BLIS as enterocin OS13α and OS13β represent antimicrobial agents with activities against antibiotic-resistant enterococcal isolates.
Collapse
|
35
|
Anggani HS, Perdana RG, Siregar E, Bachtiar EW. The effect of coating chitosan on Porphyromonas gingivalis biofilm formation in the surface of orthodontic mini-implant. J Adv Pharm Technol Res 2021; 12:84-88. [PMID: 33532361 PMCID: PMC7832189 DOI: 10.4103/japtr.japtr_95_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/03/2020] [Accepted: 10/03/2020] [Indexed: 12/31/2022] Open
Abstract
Infection is the main problem for the failure of orthodontic mini-implant. Modern prevention of infection is now focused on local antibacterial coatings on implant devices. Chitosan is biocompatible and has antibacterial properties. Azithromycin is a synthetic antibiotic with immunomodulatory properties in which it has an advantage over the rest of antibiotics. This study aimed to evaluate the effect coating chitosan on the orthodontic mini-implant in Porphyromonas gingivalis biofilm formation. This is an experimental study using 25 orthodontic mini-implants. Five samples were coated with chitosan, 5 samples were coated with chitosan-azithromycin, 5 samples were coated with azithromycin, 5 samples were uncoated, and 5 samples were uncoated and were not exposed to P. gingivalis. P. gingivalis biofilms on the surface of the orthodontic mini-implant were observed after 24 h of incubation. P. gingivalis biofilm mass inhibition was highest in the azithromycin-treated group, followed by chitosan + azithromycin and chitosan only. The one-way ANOVA statistic test and post hoc Bonferroni statistic test of P. gingivalis biofilm mass show a significant difference between and within groups of experiments (P < 0.05). The Pearson correlation test with a value of R = +0.88, indicated that the bacterial viability count and the biofilm mass have a strong positive correlation. In conclusion, orthodontic mini-implant coated with chitosan, chitosan with azithromycin, or azithromycin only effectively suppressed P. gingivalis biofilm formation.
Collapse
Affiliation(s)
- Haru Setyo Anggani
- Department of Orthodontic, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Renaldo Guruh Perdana
- Department of Orthodontic, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Erwin Siregar
- Department of Orthodontic, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Endang Winiati Bachtiar
- Department of Oral Biology, Oral Science Research Center, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
36
|
Litt PK, Kakani R, Jadeja R, Saha J, Kountoupis T, Jaroni D. Effectiveness of Bacteriophages Against Biofilm-Forming Shiga-Toxigenic Escherichia coli on Leafy Greens and Cucumbers. PHAGE (NEW ROCHELLE, N.Y.) 2020; 1:213-222. [PMID: 36147291 PMCID: PMC9041472 DOI: 10.1089/phage.2020.0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Background: Shiga-toxigenic Escherichia coli (STEC) have caused several produce-associated outbreaks, making it challenging to control these pathogens. Bacteriophages could serve as effective biocontrol. Materials and Methods: Spinach, lettuce, and cucumbers, inoculated with STEC (O157, O26, O45, O103, O111, O121, O145), were treated with lytic bacteriophages and stored at 4°C for 3 days. Surviving STEC were enumerated and observed under scanning electron microscope (SEM), and data analyzed using one-way analysis of variance (ANOVA) (p < 0.05). Results: Bacteriophage treatments significantly reduced STEC populations, compared with the control (p < 0.05). On spinach and romaine, STEC O26, O45, and O103 were reduced to undetectable levels and STEC O157, O111, O121, and O145 by ∼2 logs CFU/cm2. Multiserotype phage cocktail reduced STEC on leafy greens by 1.4 CFU/cm2 and on cucumbers by 1.7 logs CFU/cucumber. Clusters of STEC cells, surrounded by extracellular matrix, were observed under SEM of positive control, whereas phage-treated produce surface showed fewer cells, with cellular damage. Conclusions: Bacteriophages could be utilized as biocontrol against STEC on fresh produce.
Collapse
Affiliation(s)
- Pushpinder K. Litt
- Department of Animal and Food Sciences, Food and Agricultural Products Center, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Radhika Kakani
- Department of Animal and Food Sciences, Food and Agricultural Products Center, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Ravirajsinh Jadeja
- Department of Animal and Food Sciences, Food and Agricultural Products Center, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Joyjit Saha
- Department of Animal and Food Sciences, Food and Agricultural Products Center, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Tony Kountoupis
- Department of Animal and Food Sciences, Food and Agricultural Products Center, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Divya Jaroni
- Department of Animal and Food Sciences, Food and Agricultural Products Center, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
37
|
Dhingra S, Rahman NAA, Peile E, Rahman M, Sartelli M, Hassali MA, Islam T, Islam S, Haque M. Microbial Resistance Movements: An Overview of Global Public Health Threats Posed by Antimicrobial Resistance, and How Best to Counter. Front Public Health 2020; 8:535668. [PMID: 33251170 PMCID: PMC7672122 DOI: 10.3389/fpubh.2020.535668] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022] Open
Abstract
Antibiotics changed medical practice by significantly decreasing the morbidity and mortality associated with bacterial infection. However, infectious diseases remain the leading cause of death in the world. There is global concern about the rise in antimicrobial resistance (AMR), which affects both developed and developing countries. AMR is a public health challenge with extensive health, economic, and societal implications. This paper sets AMR in context, starting with the history of antibiotics, including the discovery of penicillin and the golden era of antibiotics, before exploring the problems and challenges we now face due to AMR. Among the factors discussed is the low level of development of new antimicrobials and the irrational prescribing of antibiotics in developed and developing countries. A fundamental problem is the knowledge, attitude, and practice (KAP) regarding antibiotics among medical practitioners, and we explore this aspect in some depth, including a discussion on the KAP among medical students. We conclude with suggestions on how to address this public health threat, including recommendations on training medical students about antibiotics, and strategies to overcome the problems of irrational antibiotic prescribing and AMR.
Collapse
Affiliation(s)
- Sameer Dhingra
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Nor Azlina A. Rahman
- Department of Physical Rehabilitation Sciences, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Malaysia
| | - Ed Peile
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Motiur Rahman
- Oxford University Clinical Research Unit, Wellcome Trust Asia Programme, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| | - Massimo Sartelli
- Department of General and Emergency Surgery, Macerata Hospital, Macerata, Italy
| | - Mohamed Azmi Hassali
- The Discipline of Social and Administrative Pharmacy, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Malaysia
| | | | - Salequl Islam
- Department of Microbiology, Jahangirnagar University, Dhaka, Bangladesh
| | - Mainul Haque
- The Unit of Pharmacology, Faculty of Medicine and Defence Health, National Defence University of Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Antibacterial Activity of Synthetic Cationic Iron Porphyrins. Antioxidants (Basel) 2020; 9:antiox9100972. [PMID: 33050461 PMCID: PMC7601463 DOI: 10.3390/antiox9100972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/28/2022] Open
Abstract
Widespread antibiotic resistance demands new strategies for fighting infections. Porphyrin-based compounds were long ago introduced as photosensitizers for photodynamic therapy, but light-independent antimicrobial activity of such compounds has not been systematically explored. The results of this study demonstrate that synthetic cationic amphiphilic iron N-alkylpyridylporphyrins exert strong bactericidal action at concentrations as low as 5 μM. Iron porphyrin, FeTnHex-2-PyP, which is well tolerated by laboratory animals, efficiently killed Gram-negative and Gram-positive microorganisms. Its bactericidal activity was oxygen-independent and was controlled by the lipophilicity and accumulation of the compound in bacterial cells. Such behavior is in contrast with the anionic gallium protoporphyrin IX, whose efficacy depends on cellular heme uptake systems. Under aerobic conditions, however, the activity of FeTnHex-2-PyP was limited by its destruction due to redox-cycling. Neither iron released from the Fe-porphyrin nor other decomposition products were the cause of the bactericidal activity. FeTnHex-2-PyP was as efficient against antibiotic-sensitive E. coli and S. aureus as against their antibiotic-resistant counterparts. Our data demonstrate that development of amphiphilic, positively charged metalloporphyrins might be a promising approach in the introduction of new weapons against antibiotic-resistant strains.
Collapse
|
39
|
Antibacterial activity of MPA-capped CdTe and Ag-doped CdTe nanocrystals: Showing different activity against gram-positive and gram-negative bacteria. CHEMICAL PAPERS 2020. [DOI: 10.1007/s11696-020-01170-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
40
|
Chung PY. Novel targets of pentacyclic triterpenoids in Staphylococcus aureus: A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 73:152933. [PMID: 31103429 DOI: 10.1016/j.phymed.2019.152933] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Staphylococcus aureus is an important pathogen both in community-acquired and healthcare-associated infections, and has successfully evolved numerous strategies for resisting the action to practically all antibiotics. Resistance to methicillin is now widely described in the community setting (CMRSA), thus the development of new drugs or alternative therapies is urgently necessary. Plants and their secondary metabolites have been a major alternative source in providing structurally diverse bioactive compounds as potential therapeutic agents for the treatment of bacterial infections. One of the classes of natural secondary metabolites from plants with the most bioactive compounds are the triterpenoids, which comprises structurally diverse organic compounds. In nature, triterpenoids are often found as tetra- or penta-cyclic structures. AIM This review highlights the anti-staphylococcal activities of pentacyclic triterpenoids, particularly α-amyrin (AM), betulinic acid (BA) and betulinaldehyde (BE). These compounds are based on a 30-carbon skeleton comprising five six-membered rings (ursanes and lanostanes) or four six-membered rings and one five-membered ring (lupanes and hopanes). METHODS Electronic databases such as ScienceDirect, PubMed and Scopus were used to search scientific contributions until March 2018, using relevant keywords. Literature focusing on the antimicrobial and antibiofilms of effects of pentacyclic triterpenoids on S. aureus were identified and summarized. RESULTS Pentacyclic triterpenoids can be divided into three representative classes, namely ursane, lupane and oleananes. This class of compounds have been shown to exhibit analgesic, immunomodulatory, anti-inflammatory, anticancer, antioxidant, antifungal and antibacterial activities. In studies of the antimicrobial activities and targets of AM, BA and BE in sensitive and multidrug-resistant S. aureus, these compounds acted synergistically and have different targets from the conventional antibiotics. CONCLUSION The inhibitory mechanisms of S. aureus in novel targets and pathways should stimulate further researches to develop AM, BA and BE as therapeutic agents for infections caused by S. aureus. Continued efforts to identify and exploit synergistic combinations by the three compounds and peptidoglycan inhibitors, are also necessary as alternative treatment options for S. aureus infections.
Collapse
Affiliation(s)
- Pooi Yin Chung
- Department of Pathology, School of Medicine, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
41
|
The multifunctional enzyme S-adenosylhomocysteine/methylthioadenosine nucleosidase is a key metabolic enzyme in the virulence of Salmonella enterica var Typhimurium. Biochem J 2020; 476:3435-3453. [PMID: 31675053 DOI: 10.1042/bcj20190297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 11/17/2022]
Abstract
Key physiological differences between bacterial and mammalian metabolism provide opportunities for the development of novel antimicrobials. We examined the role of the multifunctional enzyme S-adenosylhomocysteine/Methylthioadenosine (SAH/MTA) nucleosidase (Pfs) in the virulence of S. enterica var Typhimurium (S. Typhimurium) in mice, using a defined Pfs deletion mutant (i.e. Δpfs). Pfs was essential for growth of S. Typhimurium in M9 minimal medium, in tissue cultured cells, and in mice. Studies to resolve which of the three known functions of Pfs were key to murine virulence suggested that downstream production of autoinducer-2, spermidine and methylthioribose were non-essential for Salmonella virulence in a highly sensitive murine model. Mass spectrometry revealed the accumulation of SAH in S. Typhimurium Δpfs and complementation of the Pfs mutant with the specific SAH hydrolase from Legionella pneumophila reduced SAH levels, fully restored growth ex vivo and the virulence of S. Typhimurium Δpfs for mice. The data suggest that Pfs may be a legitimate target for antimicrobial development, and that the key role of Pfs in bacterial virulence may be in reducing the toxic accumulation of SAH which, in turn, suppresses an undefined methyltransferase.
Collapse
|
42
|
Motika SE, Ulrich RJ, Geddes EJ, Lee HY, Lau GW, Hergenrother PJ. Gram-Negative Antibiotic Active Through Inhibition of an Essential Riboswitch. J Am Chem Soc 2020; 142:10856-10862. [PMID: 32432858 DOI: 10.1021/jacs.0c04427] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multidrug-resistant Gram-negative (GN) infections for which there are few available treatment options are increasingly common. The development of new antibiotics for these pathogens is challenging because of the inability of most small molecules to accumulate inside GN bacteria. Using recently developed predictive guidelines for compound accumulation in Escherichia coli, we have converted the antibiotic Ribocil C, which targets the flavin mononucleotide (FMN) riboswitch, from a compound lacking whole-cell activity against wild-type GN pathogens into a compound that accumulates to a high level in E. coli, is effective against Gram-negative clinical isolates, and has efficacy in mouse models of GN infections. This compound allows for the first assessment of the translational potential of FMN riboswitch binders against wild-type Gram-negative bacteria.
Collapse
Affiliation(s)
| | | | | | | | - Gee W Lau
- Department of Pathobiology, University of Illinois, Urbana, Illinois 61802, United States
| | | |
Collapse
|
43
|
Photodegradation of Antibiotics by Noncovalent Porphyrin-Functionalized TiO 2 in Water for the Bacterial Antibiotic Resistance Risk Management. Int J Mol Sci 2020; 21:ijms21113775. [PMID: 32471075 PMCID: PMC7312883 DOI: 10.3390/ijms21113775] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022] Open
Abstract
Antibiotics represent essential drugs to contrast the insurgence of bacterial infections in humans and animals. Their extensive use in livestock farming, including aquaculture, has improved production performances and food safety. However, their overuse can implicate a risk of water pollution and related antimicrobial resistance. Consequently, innovative strategies for successfully removing antibiotic contaminants have to be advanced to protect human health. Among them, photodegradation TiO2-driven under solar irradiation appears not only as a promising method, but also a sustainable pathway. Hence, we evaluated several composite TiO2 powders with H2TCPP, CuTCPP, ZnTCPP, and SnT4 porphyrin for this scope in order to explore the effect of porphyrins sensitization on titanium dioxide. The synthesis was realized through a fully non-covalent functionalization in water at room conditions. The efficacy of obtained composite materials was also tested in photodegrading oxolinic acid and oxytetracycline in aqueous solution at micromolar concentrations. Under simulated solar irradiation, TiO2 functionalized with CuTCPP has shown encouraging results in the removal of oxytetracycline from water, by opening the way as new approaches to struggle against antibiotic's pollution and, finally, to represent a new valuable tool of public health.
Collapse
|
44
|
Abouhmad A, Korany AH, Grey C, Dishisha T, Hatti-Kaul R. Exploring the Enzymatic and Antibacterial Activities of Novel Mycobacteriophage Lysin B Enzymes. Int J Mol Sci 2020; 21:ijms21093176. [PMID: 32365915 PMCID: PMC7246905 DOI: 10.3390/ijms21093176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 01/01/2023] Open
Abstract
Mycobacteriophages possess different sets of lytic enzymes for disruption of the complex cell envelope of the mycobacteria host cells and release of the viral progeny. Lysin B (LysB) enzymes are mycolylarabinogalactan esterases that cleave the ester bond between the arabinogalactan and mycolic acids in the mycolylarabinogalactan-peptidoglycan (mAGP) complex in the cell envelope of mycobacteria. In the present study, four LysB enzymes were produced recombinantly and characterized with respect to their enzymatic and antibacterial activities. Examination of the kinetic parameters for the hydrolysis of para-nitrophenyl ester substrates, shows LysB-His6 enzymes to be active against a range of substrates (C4–C16), with a catalytic preference towards p-nitrophenyl laurate (C12). With p-nitrophenyl butyrate as substrate, LysB-His6 enzymes showed highest activity at 37 °C. LysB-His6 enzymes also hydrolyzed different Tween substrates with highest activity against Tween 20 and 80. Metal ions like Ca2+ and Mn2+ enhanced the enzymatic activity of LysB-His6 enzymes, while transition metal ions like Zn2+ and Cu2+ inhibited the enzymatic activity. The mycolylarabinogalactan esterase activity of LysB-His6 enzymes against mAGP complex was confirmed by LC-MS. LysB-His6 enzymes showed marginal antibacterial activity when tested alone against Mycobacterium smegmatis, however a synergetic activity was noticed when combined with outer membrane permealizers. These results confirm that LysB enzymes are lipolytic enzymes with potential application as antimycobacterials.
Collapse
Affiliation(s)
- Adel Abouhmad
- Division of Biotechnology, Department of Chemistry, Center for Chemistry and Chemical Engineering, Lund University, P.O. Box 124, SE-22100 Lund, Sweden; (A.A.); (C.G.)
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Ahmed H. Korany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt;
| | - Carl Grey
- Division of Biotechnology, Department of Chemistry, Center for Chemistry and Chemical Engineering, Lund University, P.O. Box 124, SE-22100 Lund, Sweden; (A.A.); (C.G.)
| | - Tarek Dishisha
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62511, Egypt;
| | - Rajni Hatti-Kaul
- Division of Biotechnology, Department of Chemistry, Center for Chemistry and Chemical Engineering, Lund University, P.O. Box 124, SE-22100 Lund, Sweden; (A.A.); (C.G.)
- Correspondence: ; Tel.: +46-462-224-840
| |
Collapse
|
45
|
Potential application in amoxicillin removal of Alcaligenes sp. MMA and enzymatic studies through molecular docking. Arch Microbiol 2020; 202:1489-1495. [DOI: 10.1007/s00203-020-01868-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/21/2020] [Accepted: 03/12/2020] [Indexed: 01/17/2023]
|
46
|
Li X, Chen J, Andersen JM, Chu J, Jensen PR. Cofactor Engineering Redirects Secondary Metabolism and Enhances Erythromycin Production in Saccharopolyspora erythraea. ACS Synth Biol 2020; 9:655-670. [PMID: 32078772 DOI: 10.1021/acssynbio.9b00528] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Saccharopolyspora erythraea is used for industrial erythromycin production. To explore the physiological role of intracellular energy state in metabolic regulation by S. erythraea, we initially overexpressed the F1 part of the endogenous F1F0-ATPase in the high yielding erythromycin producing strain E3. The F1-ATPase expression resulted in lower [ATP]/[ADP] ratios, which was accompanied by a strong increase in the production of a reddish pigment and a decreased erythromycin production. Subsequent transcriptional analysis revealed that the lower intracellular [ATP]/[ADP] ratios exerted a pleotropic regulation on the metabolism of S. erythraea. The lower [ATP]/[ADP] ratios induced physiological changes to restore the energy balance, mainly via pathways that tend to produce ATP or regenerate NADH. The F1-ATPase overexpression strain exhibited a state of redox stress, which was correlated to an alteration of electron transport at the branch of the terminal oxidases, and S. erythraea channeled the enhanced glycolytic flux toward a reddish pigment in order to reduce NADH formation. The production of erythromycin was decreased, which is in accordance with the net ATP requirement and the excess NADH formed through this pathway. Partial growth inhibition by apramycin increased the intracellular [ATP]/[ADP] ratios and demonstrated a positive correlation between [ATP]/[ADP] ratios and erythromycin synthesis. Finally, overexpression of the entire F1F0-ATPase complex resulted in 28% enhanced erythromycin production and markedly reduced pigment synthesis in E3. The work illustrates a feasible strategy to optimize the distribution of fluxes in secondary metabolism.
Collapse
Affiliation(s)
- Xiaobo Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People’s Republic of China
- National Food Institute, Technical University of Denmark, Kemitorvet Building 201, DK2800 Kongens Lyngby, Denmark
| | - Jun Chen
- National Food Institute, Technical University of Denmark, Kemitorvet Building 201, DK2800 Kongens Lyngby, Denmark
| | - Joakim M. Andersen
- National Food Institute, Technical University of Denmark, Kemitorvet Building 201, DK2800 Kongens Lyngby, Denmark
| | - Ju Chu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People’s Republic of China
| | - Peter R. Jensen
- National Food Institute, Technical University of Denmark, Kemitorvet Building 201, DK2800 Kongens Lyngby, Denmark
| |
Collapse
|
47
|
Annunziato G. Strategies to Overcome Antimicrobial Resistance (AMR) Making Use of Non-Essential Target Inhibitors: A Review. Int J Mol Sci 2019; 20:E5844. [PMID: 31766441 PMCID: PMC6928725 DOI: 10.3390/ijms20235844] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/23/2022] Open
Abstract
Antibiotics have always been considered as one of the most relevant discoveries of the twentieth century. Unfortunately, the dawn of the antibiotic era has sadly corresponded to the rise of the phenomenon of antimicrobial resistance (AMR), which is a natural process whereby microbes evolve in such a way to withstand the action of drugs. In this context, the identification of new potential antimicrobial targets and/or the identification of new chemical entities as antimicrobial drugs are in great demand. To date, among the many possible approaches used to deal with antibiotic resistance is the use of antibiotic adjuvants that hit bacterial non-essential targets. In this review, the author focuses on the discovery of antibiotic adjuvants and on new tools to study and reduce the prevalence of resistant bacterial infections.
Collapse
Affiliation(s)
- Giannamaria Annunziato
- Probes for Targets Group (P4T group), Department of food and Drug, University of Parma, 43124 Parma, Italy
| |
Collapse
|
48
|
Rello J, Parisella FR, Perez A. Alternatives to antibiotics in an era of difficult-to-treat resistance: new insights. Expert Rev Clin Pharmacol 2019; 12:635-642. [PMID: 31092053 DOI: 10.1080/17512433.2019.1619454] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: The rise of antibiotic resistance, the limited efficacy and the adverse events associated with antibiotics have urged the development of alternative measures to treat bacterial infections. Novel therapies which are pathogen specific and are safer to the healthy microbiome are being developed. Areas covered: This manuscript provides a compact overview of the feasibility and clinical impact of the latest novel therapies, with a focus on monoclonal antibodies (mAbs), vaccines, stem cells, bacteriophages, and liposomes. This is a follow-up of a previous manuscript (doi: 10.1080/17512433.2016.1241141); a database search (PubMed, EMBASE, Cochrane) was used to identify recently published literature (from January 2016) which was not covered in the previous publication. Expert opinion: Among non-traditional agents, monoclonal antibodies have not been as successful as in other therapeutic areas. In particular many are developed to prevent hospital-acquired infections caused by S. aureus or P. aeruginosa and, so far, results have been overall disappointing. Stem cells and bacteriophages still have a long way to go. Vaccines are always desirable to prevent infections but again there is a lack of confirmatory results. Broad spectrum liposomes have shown promising results in treating severely infected patients and could be game changers in patient management.
Collapse
Affiliation(s)
- Jordi Rello
- a Vall d'Hebron Institut of Research (VHIR), Barcelona, Spain & Centro de Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES) , Instituto Salud Carlos III , Barcelona , Spain.,b Centrode Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES) , Instituto Salud Carlos III , Barcelona , Spain
| | - Francesca Romana Parisella
- c UQ Centre for Clinical Research, Faculty of Medicine , The University of Queensland , Brisbane , Australia
| | - Antonio Perez
- d Medical Department , Combioxin SA , Geneve , Switzerland
| |
Collapse
|
49
|
Bai J, Yang E, Chang PS, Ryu S. Preparation and characterization of endolysin-containing liposomes and evaluation of their antimicrobial activities against gram-negative bacteria. Enzyme Microb Technol 2019; 128:40-48. [PMID: 31186109 DOI: 10.1016/j.enzmictec.2019.05.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/25/2019] [Accepted: 05/13/2019] [Indexed: 11/18/2022]
Abstract
The overuse and misuse of antibiotics in treating bacterial infections cause the rapid emergence of drug-resistant bacteria, suggesting that the development of alternative strategies to control antibiotic-resistant bacteria is urgently needed. Endolysins are bacteriophage-encoded enzymes that can degrade peptidoglycan in bacterial cell walls, and they have great potential as alternative antimicrobial agents. However, exogenous application of recombinant endolysin is limited to Gram-positive bacteria because endolysins cannot penetrate the outer membrane of Gram-negative bacteria. Here, a liposome-mediated endolysin encapsulation system was developed, and its ability to penetrate the outer membrane of Gram-negative bacteria was tested. The phage-derived endolysin BSP16Lys was isolated, characterized, and used for encapsulation into a cationic liposome comprised of dipalmitoylphosphatidylcholine (DPPC), cholesterol, and hexadecylamine. The BSP16Lys-encapsulated liposome had a high zeta potential value (over 30 mV) with an average diameter of 303 nm. The encapsulation efficiency of BSP16Lys into the liposome was 35.27%. Salmonella Typhimuriumand Escherichia coli cells treated with BSP16Lys-encapsulated liposomes showed 2.2-log CFU/mL and 1.6-log CFU/mL reductions in the viable cell numbers, respectively, without treatment of a membrane permeabilizer. These results showed potential for liposome-mediated delivery of endolysin for exogenous application against Gram-negative bacteria.
Collapse
Affiliation(s)
- Jaewoo Bai
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eunhye Yang
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Pahn-Shick Chang
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea; Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sangryeol Ryu
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea; Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
50
|
Li Y, Gardner JJ, Fortney KR, Leus IV, Bonifay V, Zgurskaya HI, Pletnev AA, Zhang S, Zhang ZY, Gribble GW, Spinola SM, Duerfeldt AS. First-generation structure-activity relationship studies of 2,3,4,9-tetrahydro-1H-carbazol-1-amines as CpxA phosphatase inhibitors. Bioorg Med Chem Lett 2019; 29:1836-1841. [PMID: 31104993 DOI: 10.1016/j.bmcl.2019.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 12/26/2022]
Abstract
Genetic activation of the bacterial two-component signal transduction system, CpxRA, abolishes the virulence of a number of pathogens in human and murine infection models. Recently, 2,3,4,9-tetrahydro-1H-carbazol-1-amines were shown to activate the CpxRA system by inhibiting the phosphatase activity of CpxA. Herein we report the initial structure-activity relationships of this scaffold by focusing on three approaches 1) A-ring substitution, 2) B-ring deconstruction to provide N-arylated amino acid derivatives, and 3) C-ring elimination to give 2-ethylamino substituted indoles. These studies demonstrate that the A-ring is amenable to functionalization and provides a promising avenue for continued optimization of this chemotype. Further investigations revealed that the C-ring is not necessary for activity, although it likely provides conformational constraint that is beneficial to potency, and that the (R) stereochemistry is required at the primary amine. Simplification of the scaffold through deconstruction of the B-ring led to inactive compounds, highlighting the importance of the indole core. A new lead compound 26 was identified, which manifests a ∼30-fold improvement in CpxA phosphatase inhibition over the initial hit. Comparison of amino and des-amino derivatives in bacterial strains differing in membrane permeability and efflux capabilities demonstrate that the amine is required not only for target engagement but also for permeation and accumulation in Escherichia coli.
Collapse
Affiliation(s)
- Yangxiong Li
- Institute for Natural Products Applications and Research Technologies, Norman, OK 73019-5251, United States; Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Norman, OK 73019-5251, United States
| | - Jessi J Gardner
- Institute for Natural Products Applications and Research Technologies, Norman, OK 73019-5251, United States; Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Norman, OK 73019-5251, United States
| | - Katherine R Fortney
- Departments of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Inga V Leus
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Norman, OK 73019-5251, United States
| | - Vincent Bonifay
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Norman, OK 73019-5251, United States
| | - Helen I Zgurskaya
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Norman, OK 73019-5251, United States
| | - Alexandre A Pletnev
- Department of Chemistry, Dartmouth College, Hanover New Hampshire, 03755, United States
| | - Sheng Zhang
- Biochemistry, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Zhong-Yin Zhang
- Biochemistry, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, United States; The Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, United States
| | - Gordon W Gribble
- Department of Chemistry, Dartmouth College, Hanover New Hampshire, 03755, United States
| | - Stanley M Spinola
- Departments of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Adam S Duerfeldt
- Institute for Natural Products Applications and Research Technologies, Norman, OK 73019-5251, United States; Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Norman, OK 73019-5251, United States.
| |
Collapse
|