1
|
Barman S, Kurnaz LB, Leighton R, Hossain MW, Decho AW, Tang C. Intrinsic antimicrobial resistance: Molecular biomaterials to combat microbial biofilms and bacterial persisters. Biomaterials 2024; 311:122690. [PMID: 38976935 PMCID: PMC11298303 DOI: 10.1016/j.biomaterials.2024.122690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 05/13/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
The escalating rise in antimicrobial resistance (AMR) coupled with a declining arsenal of new antibiotics is imposing serious threats to global public health. A pervasive aspect of many acquired AMR infections is that the pathogenic microorganisms exist as biofilms, which are equipped with superior survival strategies. In addition, persistent and recalcitrant infections are seeded with bacterial persister cells at infection sites. Together, conventional antibiotic therapeutics often fail in the complete treatment of infections associated with bacterial persisters and biofilms. Novel therapeutics have been attempted to tackle AMR, biofilms, and persister-associated complex infections. This review focuses on the progress in designing molecular biomaterials and therapeutics to address acquired and intrinsic AMR, and the fundamental microbiology behind biofilms and persisters. Starting with a brief introduction of AMR basics and approaches to tackling acquired AMR, the emphasis is placed on various biomaterial approaches to combating intrinsic AMR, including (1) semi-synthetic antibiotics; (2) macromolecular or polymeric biomaterials mimicking antimicrobial peptides; (3) adjuvant effects in synergy; (4) nano-therapeutics; (5) nitric oxide-releasing antimicrobials; (6) antimicrobial hydrogels; (7) antimicrobial coatings. Particularly, the structure-activity relationship is elucidated in each category of these biomaterials. Finally, illuminating perspectives are provided for the future design of molecular biomaterials to bypass AMR and cure chronic multi-drug resistant (MDR) infections.
Collapse
Affiliation(s)
- Swagatam Barman
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States; Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States
| | - Leman Buzoglu Kurnaz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States
| | - Ryan Leighton
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States
| | - Md Waliullah Hossain
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States
| | - Alan W Decho
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States.
| | - Chuanbing Tang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States.
| |
Collapse
|
2
|
Tang DM, Wang ZJ, Zu WB, Jiang YM, Zhu YY, Wei MZ, Luo XD. Activity of the Caged Xanthone Morellic Acid against Vancomycin-Resistant Enterococcus Infection by Targeting the Bacterial Membrane. JOURNAL OF NATURAL PRODUCTS 2024; 87:2366-2375. [PMID: 39388644 DOI: 10.1021/acs.jnatprod.4c00425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Vancomycin-resistant Enterococcus (VRE) is an important nosocomial opportunistic pathogen that is associated with multidrug resistance. Here, we demonstrate that morellic acid inhibits VRE by restoring its sensitivity to vancomycin and ampicillin with low drug resistance and efficient biofilm clearance effects. Morellic acid binds to inner membrane phospholipids, such as phosphatidylethanolamine (PE), phosphatidylglycerol (PG), and cardiolipin (CL) of VRE, such that the fluidity and proton-motive force (PMF) interfere with the damaged inner membrane, causing intracellular reactive oxygen species (ROS) accumulation and bacterial death. Transcriptional analyses supported this effect on inner membrane-related pathways such as fatty acid biosynthesis and glycerophospholipid metabolism. Moreover, morellic acid significantly eliminated residual bacteria in the spleen, liver, kidneys, and abdominal effusion in mice. Our findings indicate the potential applications of morellic acid as an antibacterial agent or adjuvant for treating VRE infections.
Collapse
Affiliation(s)
- Dong-Mei Tang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Wen-Biao Zu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Yue-Ming Jiang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Mei-Zhen Wei
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, People's Republic of China
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, People's Republic of China
| |
Collapse
|
3
|
Sánchez-Quezada V, Velázquez-Guadarrama N, Mendoza-Elizalde S, Hernández-Iturriaga M, Landaverde PV, Loarca-Piña G. Bioaccessibility of bioactive compounds present in Persea americana Mill. seed ingredient during oral-gastric digestion with antibacterial capacity against Helicobacter pylori. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118259. [PMID: 38685366 DOI: 10.1016/j.jep.2024.118259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In ancient Mexican cultures, the Persea americana Mill seed has been used against gastrointestinal diseases, due to high concentrations of bioactive compounds. According to Traditional Mexican Medicine, P. americana seed aqueous infusion is used against roundworms, intestinal worms, parasites, and gastrointestinal problems, in a dose taken over three or four days. In addition, Mexican Society of Natural History indicates the traditional use of P. americana seed powder as an antiparasitic, and antibacterial. On the other hand, Helicobacter pylori infection is a factor associated with the development of gastric disease, peptic ulcers as well as some types of gastric lymphomas and gastric cancer in humans; in this way is necessary scientific evidence about P. americana seed effect in gastrointestinal disease. AIM OF THE STUDY The work aimed to evaluate bioactive compounds bioaccessibility and antimicrobial potential against Helicobacter pylori during oral-gastric digestion in vitro of food ingredient from Persea americana Mill. seed and elucidate the possible action mechanism using in silico tools. MATERIALS AND METHODS Initially, P. americana seed oil and aqueous extract of P. americana seed were obtained using ultrasound and maceration respectively, and the food ingredient from P. americana seed was obtained. The samples underwent oral-gastric digestions by the INFOGEST method, to continue identifying and quantifying the bioactive compounds by HPLC-DAD and GC-MS. The anti-Helicobacter pylori activity determination were used fourteen Helicobacter pylori clinical strains and reference strains by Susceptibility testing by Minimal Inhibition Concentration, Kinetics of Growth Inhibition of H. pylori, Urease Inhibitory Kinetic. Finally, to elucidate a possible action mechanism used in silico tools (Software AutoDock 4.2.6 and BioVia Discovery v.19.1.0.1.18287). RESULTS The lipophilic fraction of P. americana seed detected oleic acid, linoleic acid, and avocadenofuran compounds, and the phenolic fraction showed the presence of catechin, rutin, ellagic, and chlorogenic acid, among others. Phenolic compounds conformational changes during oral-gastric digestion due to mechanical and acid hydrolysis, while lipophilic compounds showed a 20% increase in the gastric phase. Persea americana Mill. seed ingredient (3.08 μg/mL) showed total in vitro inhibition of clinical and reference strains of H. pylori, likewise, the lipophilic fraction had a lower inhibition concentration (2.59 μg/mL) regardless of the strains. Among the mechanisms found in silico, inhibition of target proteins such as CagA, BabA, and MUC5 were observed, as virulence factors involving adherence and bacterial pathogenicity. CONCLUSIONS This research provides evidence that food ingredient from P. americana seed has antimicrobial in vitro potential against H. pylori clinical strains, through phenolic and mainly lipophilic compounds, opening new scientific evidence that supports the P. americana seed's traditional use.
Collapse
Affiliation(s)
- V Sánchez-Quezada
- Programa de Posgrado en Alimentos del Centro de la República (PROPAC), Research and Graduate Studies in Food Science, School of Chemistry, Universidad Autónoma de Querétaro, Querétaro, Mexico.
| | - N Velázquez-Guadarrama
- Unidad de Investigación en Enfermedades Infecciosas, Área de Genética Bacteriana, Hospital Infantil de México Federico Gómez, Mexico.
| | - S Mendoza-Elizalde
- Unidad de Investigación en Enfermedades Infecciosas, Área de Genética Bacteriana, Hospital Infantil de México Federico Gómez, Mexico.
| | - M Hernández-Iturriaga
- Programa de Posgrado en Alimentos del Centro de la República (PROPAC), Research and Graduate Studies in Food Science, School of Chemistry, Universidad Autónoma de Querétaro, Querétaro, Mexico.
| | - P Vázquez Landaverde
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA), Unidad Querétaro CICATA-IPN Querétaro, Mexico.
| | - G Loarca-Piña
- Programa de Posgrado en Alimentos del Centro de la República (PROPAC), Research and Graduate Studies in Food Science, School of Chemistry, Universidad Autónoma de Querétaro, Querétaro, Mexico.
| |
Collapse
|
4
|
Wang C, Ji Y, Huo X, Li X, Lu W, Zhang Z, Dong W, Wang X, Chen H, Tan C. Discovery of Salifungin as a Repurposed Antibiotic against Methicillin-Resistant Staphylococcus aureus with Limited Resistance Development. ACS Infect Dis 2024; 10:1576-1589. [PMID: 38581387 DOI: 10.1021/acsinfecdis.3c00611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2024]
Abstract
Exploring novel antimicrobial drugs and strategies has become essential to the fight MRSA-associated infections. Herein, we found that membrane-disrupted repurposed antibiotic salifungin had excellent bactericidal activity against MRSA, with limited development of drug resistance. Furthermore, adding salifungin effectively decreased the minimum inhibitory concentrations of clinical antibiotics against Staphylococcus aureus. Evaluations of the mechanism demonstrated that salifungin disrupted the level of H+ and K+ ions using hydrophilic and lipophilic groups to interact with bacterial membranes, causing the disruption of bacterial proton motive force followed by impacting on bacterial the function of the respiratory chain and adenosine 5'-triphosphate, thereby inhibiting phosphatidic acid biosynthesis. Moreover, salifungin also significantly inhibited the formation of bacterial biofilms and eliminated established bacterial biofilms by interfering with bacterial membrane potential and inhibiting biofilm-associated gene expression, which was even better than clinical antibiotics. Finally, salifungin exhibited efficacy comparable to or even better than that of vancomycin in the MRSA-infected animal models. In conclusion, these results indicate that salifungin can be a potential drug for treating MRSA-associated infections.
Collapse
Affiliation(s)
- Chenchen Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430000, Hubei, China
- Hubei Hongshan Laboratory, Wuhan 430000, Hubei, China
| | - Yueyue Ji
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430000, Hubei, China
- Hubei Hongshan Laboratory, Wuhan 430000, Hubei, China
| | - Xingyu Huo
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430000, Hubei, China
- Hubei Hongshan Laboratory, Wuhan 430000, Hubei, China
| | - Xiaodan Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430000, Hubei, China
- Hubei Hongshan Laboratory, Wuhan 430000, Hubei, China
| | - Wenjia Lu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430000, Hubei, China
- Hubei Hongshan Laboratory, Wuhan 430000, Hubei, China
| | - Zhaoran Zhang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430000, Hubei, China
- Hubei Hongshan Laboratory, Wuhan 430000, Hubei, China
| | - Wenqi Dong
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430000, Hubei, China
- Hubei Hongshan Laboratory, Wuhan 430000, Hubei, China
| | - Xiangru Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430000, Hubei, China
- Hubei Hongshan Laboratory, Wuhan 430000, Hubei, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430000, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan 430000, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430000, Hubei, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430000, Hubei, China
- Hubei Hongshan Laboratory, Wuhan 430000, Hubei, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430000, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan 430000, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430000, Hubei, China
| | - Chen Tan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430000, Hubei, China
- Hubei Hongshan Laboratory, Wuhan 430000, Hubei, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430000, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan 430000, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430000, Hubei, China
| |
Collapse
|
5
|
Zhong Y, Liu H, Chen F, He Q, Zhang X, Lan L, Yang C. Design, synthesis and biological evaluation of thiazolyl-halogenated pyrroles or pyrazoles as novel antibacterial and antibiofilm agents. Eur J Med Chem 2024; 268:116221. [PMID: 38382392 DOI: 10.1016/j.ejmech.2024.116221] [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: 12/20/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/23/2024]
Abstract
The formation of biofilm is one of the important factors for bacteria to develop drug-resistant. A series of halogenated-pyrroles or pyrazoles containing thiazole groups as antibacterial agents were designed and synthesized to target biofilms. Among them, compound 8c showed antibacterial activity against various Gram-positive bacteria, particularly against vancomycin-resistant Enterococcus faecalis (MIC ≤0.125 μg/mL). Additionally, this compound significantly inhibited biofilm formation of Staphylococcus aureus and Pseudomonas aeruginosa at sub-MIC doses. Furthermore, compound 8c exhibited significantly lower mammalian cell toxicity compared to pyrrolomycin C and its hepatic microsomal metabolic stability in various species was also evaluated. Further experiment on the infection model of Galleria mellonella proved that the compound was effective in vivo.
Collapse
Affiliation(s)
- Yuanchen Zhong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; Department of Chemistry, Shanghai University, Shanghai 200444, China
| | - Huan Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; School of Pharmacy, University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Feifei Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; School of Pharmacy, University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Qian He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Xiaofei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
| | - Lefu Lan
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; School of Pharmacy, University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
| | - Chunhao Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
6
|
Guo W, Yang Z, Wang K, Li W, Zhao Y, Yang Y, Chang W, Gong Z, Liu Z, Chen Y, Li Q. Discovery of Unique Bis-Substituted Aromatic Amide Derivatives as Novel Highly Potent Antibiotics for Combating Methicillin-Resistant Staphylococcus aureus (MRSA). J Med Chem 2024; 67:2129-2151. [PMID: 38289145 DOI: 10.1021/acs.jmedchem.3c02064] [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: 02/09/2024]
Abstract
Due to the increasing antibiotic resistance, developing novel antimicrobials to fight infections caused by resistant bacteria is imperative. Herein, a series of novel bis-substituted aromatic amides were designed and synthesized through modifying the hit compound 1, and their antimicrobial activities were evaluated. Among them, compound 4t, as the most potent lead, exhibited excellent antimicrobial activities against Gram-positive bacteria, including clinical methicillin-resistant Staphylococcus aureus (MRSA) isolates, while keeping weak hemolytic and mammalian cytotoxic activities. Furthermore, compound 4t displayed rapid bactericidal capabilities, low tendency to produce resistance, and favorable capacities to destroy bacterial biofilms. Further explorations indicated that compound 4t induces bacterial death by binding to cardiolipin (CL) on the bacterial membrane, disrupting the cell membrane, and facilitating the accumulation of reactive oxygen species (ROS). Additionally, compound 4t showed remarkable anti-MRSA activity in vivo, demonstrating compound 4t could be developed as a potential candidate to combat MRSA infections.
Collapse
Affiliation(s)
- Weikai Guo
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Zhengfan Yang
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Kexiao Wang
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Wenyu Li
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Yanyang Zhao
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Yuqing Yang
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| | - Wenjing Chang
- Henan Provincial Center for Disease Control and Prevention, Zhengzhou 450003, China
| | - Zhen Gong
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhou Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yihua Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qiming Li
- The Jointed National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng 475004, China
| |
Collapse
|
7
|
Zhou XM, Li QY, Lu X, Bheemanaboina RRY, Fang B, Cai GX, Zhou CH. Identification of unique indolylcyanoethylenyl sulfonylanilines as novel structural scaffolds of potential antibacterial agents. Eur J Med Chem 2023; 260:115773. [PMID: 37669594 DOI: 10.1016/j.ejmech.2023.115773] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 09/07/2023]
Abstract
The increasing incidence of antibiotic resistance has forced the development of unique antimicrobials with novel multitargeting mechanisms to combat infectious diseases caused by multidrug-resistant pathogens. Structurally unique indolylcyanoethylenyl sulfonylanilines (ISs) were exploited as novel promising antibacterial agents to confront stubborn drug resistance. Some prepared ISs possessed favorable bacteriostatic action towards the tested bacteria. Especially, hydroxyethyl IS 14a exerted 8-fold more potent inhibitory efficacy against multidrug-resistant A. baumannii and E. coli 25922 with the low MIC of 0.5 μg/mL than norfloxacin, and showed low cell toxicity and rapid bactericidal property. Moreover, this compound also possessed obvious effect of eradicating bacterial biofilm, which could effectually relieve the development of drug resistance. A preliminary assessment of the antibacterial mechanism indicated that compound 14a could disintegrate membrane integrity leading to the leakage of intracellular protein, inactivation of lactate dehydrogenase and metabolism inhibition. Hydroxyethyl IS 14a mediated the accumulation of excess reactive oxygen species, which further contributed to reducing glutathione, resulting in oxidative damage to bacteria. Furthermore, IS 14a could intercalate into DNA to hinder the biological function of DNA. Quantum chemical study disclosed that IS 14a with the lowest energy gap was conducive to displaying high bioactivity. These findings demonstrated that hydroxyethyl IS 14a as a prospective antimicrobial candidate for combating A. baumannii and E. coli 25922 would be a promising starting point.
Collapse
Affiliation(s)
- Xue-Mei Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Aplied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Qian-Yue Li
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Aplied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Xing Lu
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Aplied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Rammohan R Yadav Bheemanaboina
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Aplied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Bo Fang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, PR China.
| | - Gui-Xin Cai
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Aplied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China.
| | - Cheng-He Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Aplied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
8
|
Dubinin MV, Ilzorkina AI, Salimova EV, Landage MS, Khoroshavina EI, Gudkov SV, Belosludtsev KN, Parfenova LV. Effect of Fusidic Acid and Some Nitrogen-Containing Derivatives on Liposomal and Mitochondrial Membranes. MEMBRANES 2023; 13:835. [PMID: 37888007 PMCID: PMC10608686 DOI: 10.3390/membranes13100835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/05/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
The paper assesses the membranotropic action of the natural antibiotic fusidic acid (FA) and its derivatives. It was found that a FA analogue with ethylenediamine moiety (derivative 2), in contrast to native FA and 3,11-dioxime analogue (derivative 1), is able to increase the mobility of the lipid bilayer in the zone of lipid headgroups, as well as to induce permeabilization of lecithin liposome membranes. A similar effect of derivative 2 is also observed in the case of rat liver mitochondrial membranes. We noted a decrease in the microviscosity of the mitochondrial membrane and nonspecific permeabilization of organelle membranes in the presence of this agent, which was accompanied by a decrease in mitochondrial Δψ and OXPHOS efficiency. This led to a reduction in mitochondrial calcium retention capacity. The derivatives also reduced the production of H2O2 by mitochondria. The paper considers the relationship between the structure of the tested compounds and the observed effects.
Collapse
Affiliation(s)
- Mikhail V. Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
| | - Anna I. Ilzorkina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Elena V. Salimova
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, Prospekt Oktyabrya 141, 450075 Ufa, Russia
| | - Manish S. Landage
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
| | - Ekaterina I. Khoroshavina
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
| | - Sergey V. Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Vavilov St. 38, 119991 Moscow, Russia
| | - Konstantin N. Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Lyudmila V. Parfenova
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, Prospekt Oktyabrya 141, 450075 Ufa, Russia
| |
Collapse
|
9
|
Ganesan N, Mishra B, Felix L, Mylonakis E. Antimicrobial Peptides and Small Molecules Targeting the Cell Membrane of Staphylococcus aureus. Microbiol Mol Biol Rev 2023; 87:e0003722. [PMID: 37129495 PMCID: PMC10304793 DOI: 10.1128/mmbr.00037-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
Clinical management of Staphylococcus aureus infections presents a challenge due to the high incidence, considerable virulence, and emergence of drug resistance mechanisms. The treatment of drug-resistant strains, such as methicillin-resistant S. aureus (MRSA), is further complicated by the development of tolerance and persistence to antimicrobial agents in clinical use. To address these challenges, membrane disruptors, that are not generally considered during drug discovery for agents against S. aureus, should be explored. The cell membrane protects S. aureus from external stresses and antimicrobial agents, but membrane-targeting antimicrobial agents are probably less likely to promote bacterial resistance. Nontypical linear cationic antimicrobial peptides (AMPs), highly modified AMPs such as daptomycin (lipopeptide), bacitracin (cyclic peptide), and gramicidin S (cyclic peptide), are currently in clinical use. Recent studies have demonstrated that AMPs and small molecules can penetrate the cell membrane of S. aureus, inhibit phospholipid biosynthesis, or block the passage of solutes between the periplasm and the exterior of the cell. In addition to their primary mechanism of action (MOA) that targets the bacterial membrane, AMPs and small molecules may also impact bacteria through secondary mechanisms such as targeting the biofilm, and downregulating virulence genes of S. aureus. In this review, we discuss the current state of research into cell membrane-targeting AMPs and small molecules and their potential mechanisms of action against drug-resistant physiological forms of S. aureus, including persister cells and biofilms.
Collapse
Affiliation(s)
- Narchonai Ganesan
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Biswajit Mishra
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Department of Medicine, The Miriam Hospital, Providence, Rhode Island, USA
| | - LewisOscar Felix
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Department of Medicine, Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
10
|
Frolov NA, Seferyan MA, Valeev AB, Saverina EA, Detusheva EV, Vereshchagin AN. The Antimicrobial and Antibiofilm Potential of New Water-Soluble Tris-Quaternary Ammonium Compounds. Int J Mol Sci 2023; 24:10512. [PMID: 37445691 DOI: 10.3390/ijms241310512] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
The invention and innovation of highly effective antimicrobials are always crucial tasks for medical and organic chemistry, especially at the current time, when there is a serious threat of shortages of effective antimicrobials following the pandemic. In the study presented in this article, we established a new approach to synthesizing three novel series of bioactive water-soluble tris-quaternary ammonium compounds using an optimized one-pot method, and we assessed their antimicrobial and antibiofilm potential. Five pathogenic microorganisms of the ESKAPE group, including highly resistant clinical isolates, were used as the test samples. Moreover, we highlighted the dependence of antibacterial activity from the hydrophilic-hydrophobic balance of the QACs and noted the significant performance of the desired products on biofilms with MBEC as low as 16 mg/L against bacteria and 8 mg/L against fungi. Particularly notable was the high activity against Pseudomonas aeruginosa and Acinetobacter baumannii, which are among the most resilient bacteria known. The presented work will provide useful insights for future research on the topic.
Collapse
Affiliation(s)
- Nikita A Frolov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia
| | - Mary A Seferyan
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia
| | - Anvar B Valeev
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia
- Higher Chemical College of the Russian Academy of Sciences, D. Mendeleev University of Chemical Technology of Russia, Miusskaya Square 9, 125047 Moscow, Russia
| | - Evgeniya A Saverina
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia
- Laboratory of Biologically Active Compounds and Biocomposites, Tula State University, Lenin Prospect. 92, 300012 Tula, Russia
| | - Elena V Detusheva
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia
- State Research Center for Applied Microbiology and Biotechnology, 142279 Obolensk, Russia
| | - Anatoly N Vereshchagin
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia
| |
Collapse
|
11
|
Salimova EV, Mozgovoj OS, Efimova SS, Ostroumova OS, Parfenova LV. 3-Amino-Substituted Analogues of Fusidic Acid as Membrane-Active Antibacterial Compounds. MEMBRANES 2023; 13:309. [PMID: 36984696 PMCID: PMC10056636 DOI: 10.3390/membranes13030309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 06/18/2023]
Abstract
Fusidic acid (FA) is an antibiotic with high activity against Staphylococcus aureus; it has been used in clinical practice since the 1960s. However, the narrow antimicrobial spectrum of FA limits its application in the treatment of bacterial infections. In this regard, this work aims both at the study of the antimicrobial effect of a number of FA amines and at the identification of their potential biological targets. In this way, FA analogues containing aliphatic and aromatic amino groups and biogenic polyamine, spermine and spermidine, moieties at the C-3 atom, were synthesized (20 examples). Pyrazinecarboxamide-substituted analogues exhibit a high antibacterial activity against S. aureus (MRSA) with MIC ≤ 0.25 μg/mL. Spermine and spermidine derivatives, along with activity against S. aureus, also inhibit the growth and reproduction of Gram-negative bacteria Escherichia coli, Acinetobacter baumannii, and Pseudomonas aeruginosa, and have a high fungicidal effect against Candida albicans and Cryptococcus neoformans. The study of the membrane activity demonstrated that the spermidine- and spermine-containing compounds are able to immerse into membranes and disorder the lipidsleading to a detergent effect. Moreover, spermine-based compounds are also able to form ion-permeable pores in the lipid bilayers mimicking the bacterial membranes. Using molecular docking, inhibition of the protein synthesis elongation factor EF-G was proposed, and polyamine substituents were shown to make the greatest contribution to the stability of the complexes of fusidic acid derivatives with biological targets. This suggests that the antibacterial effect of the obtained compounds may be associated with both membrane activity and inhibition of the elongation factor EF-G.
Collapse
Affiliation(s)
- Elena V. Salimova
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141 Prospect Oktyabrya, 450075 Ufa, Russia
| | - Oleg S. Mozgovoj
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141 Prospect Oktyabrya, 450075 Ufa, Russia
| | - Svetlana S. Efimova
- Institute of Cytology of Russian Academy of Sciences, 4 Tikhoretsky Prospect, 194064 Saint Petersburg, Russia
| | - Olga S. Ostroumova
- Institute of Cytology of Russian Academy of Sciences, 4 Tikhoretsky Prospect, 194064 Saint Petersburg, Russia
| | - Lyudmila V. Parfenova
- Institute of Petrochemistry and Catalysis, Ufa Federal Research Center, Russian Academy of Sciences, 141 Prospect Oktyabrya, 450075 Ufa, Russia
| |
Collapse
|
12
|
Zhou XM, Hu YY, Fang B, Zhou CH. Benzenesulfonyl thiazoloimines as unique multitargeting antibacterial agents towards Enterococcus faecalis. Eur J Med Chem 2023; 248:115088. [PMID: 36623329 DOI: 10.1016/j.ejmech.2023.115088] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/26/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023]
Abstract
New efficient antimicrobial agents are urgently needed to combat invasive multidrug-resistant pathogens infections. Structurally unique benzenesulfonyl thiazoloimines (BSTIs) were exploited as novel potential antibacterial victors to confront terrific drug resistance. Some developed BSTIs exerted effectively antimicrobial efficacy against the tested strains. Notably, 2-pyridyl BSTI 14d exhibited good antibacterial activity against E. faecalis with MIC value of 1 μg/mL, which was superior to sulfathiazole and norfloxacin. The most active compound 14d not only showed rapid bactericidal properties and impeded E. faecalis biofilm formation to effectually relieve the development of drug resistance, but also performed low toxicity toward human red blood cells, human normal squamous epithelial cells and human non-neoplastic colon epithelial cells. Mechanistic investigation demonstrated that molecule 14d could exert efficient membrane destruction leading to the leakage of intracellular materials and metabolism inhibition, cause oxidative damage of E. faecalis through accumulation of excess reactive oxygen species and reduction of glutathione activity, and intercalate into DNA to hinder replication of DNA. Molecular docking indicated that the formation of 14d-dihydrofolate synthetase supramolecular complex could hinder the function of this enzyme. ADME analysis displayed that compound 14d possessed promising pharmacokinetic properties. These findings suggested that the newly developed benzenesulfonyl thiazoloimines with multitargeting antibacterial potential provided a new possibility for evading resistance.
Collapse
Affiliation(s)
- Xue-Mei Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Yuan-Yuan Hu
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Bo Fang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, PR China.
| | - Cheng-He Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
13
|
Odžak R, Crnčević D, Sabljić A, Primožič I, Šprung M. Synthesis and Biological Evaluation of 3-Amidoquinuclidine Quaternary Ammonium Compounds as New Soft Antibacterial Agents. Pharmaceuticals (Basel) 2023; 16:187. [PMID: 37259335 PMCID: PMC9966435 DOI: 10.3390/ph16020187] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 08/27/2023] Open
Abstract
Quaternary ammonium compounds (QACs) are among the most effective antimicrobial agents that have been used for more than a century. However, due to the growing trend of bacterial resistance and high toxicity of QACs, research in this field remains a pressing matter. Recent studies of the structure-activity relationship suggest that the introduction of the amide functional group into QAC structures results in soft variants that retain their antimicrobial properties while opening the possibility of fine-tuned activity regulation. Here, we report the synthesis and structure-function study of three structurally distinct series of naturally derived soft QACs. The obtained 3-amidoquinuclidine QACs showed a broad range of antibacterial activities related to the hydrophobic-hydrophilic balance of the QAC structures. All three series yielded candidates with minimal inhibitory concentrations (MIC) in the single-digit μM range. Time-resolved growth analysis revealed subtle differences in the antibacterial activity of the selected candidates. The versatile MIC values were recorded in different nutrient media, suggesting that the media composition may have a dramatic impact on the antibacterial potential. The new QACs were found to have excellent potential to suppress bacterial biofilm formation while exhibiting low ability to induce bacterial resistance. In addition, the selected candidates were found to be less toxic than commercially available QACs and proved to be potential substrates for protease degradation. These data suggest that 3-amidoquinuclidine QACs could be considered as novel antimicrobial agents that pose a low threat to ecosystems and human health.
Collapse
Affiliation(s)
- Renata Odžak
- Department of Chemistry, Faculty of Science, University of Split, R. Bošković 33, 21000 Split, Croatia
| | - Doris Crnčević
- Department of Chemistry, Faculty of Science, University of Split, R. Bošković 33, 21000 Split, Croatia
- Doctoral Study of Biophysics, Faculty of Science, University of Split, R. Bošković 33, 21000 Split, Croatia
| | - Antonio Sabljić
- Department of Chemistry, Faculty of Science, University of Split, R. Bošković 33, 21000 Split, Croatia
- Doctoral Study of Biophysics, Faculty of Science, University of Split, R. Bošković 33, 21000 Split, Croatia
| | - Ines Primožič
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, 10000 Zagreb, Croatia
| | - Matilda Šprung
- Department of Chemistry, Faculty of Science, University of Split, R. Bošković 33, 21000 Split, Croatia
| |
Collapse
|
14
|
Quinones as an Efficient Molecular Scaffold in the Antibacterial/Antifungal or Antitumoral Arsenal. Int J Mol Sci 2022; 23:ijms232214108. [PMID: 36430585 PMCID: PMC9697455 DOI: 10.3390/ijms232214108] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
Quinone-based compounds constitute several general classes of antibiotics that have long shown unwavering efficiency against both Gram-positive and Gram-negative microbial infections. These quinone-based antibiotics are increasingly popular due to their natural origins and are used in natural beverages from herbs or plants in African, Chinese and Indian traditional medicines to treat and prevent various diseases. Quinone-based antibiotics display different bioactive profiles depending on their structures and exert specific biocidal and anti-biofilm properties, and based on recent literature, will be discussed herein.
Collapse
|
15
|
Chug M, Brisbois EJ. Recent Developments in Multifunctional Antimicrobial Surfaces and Applications toward Advanced Nitric Oxide-Based Biomaterials. ACS MATERIALS AU 2022; 2:525-551. [PMID: 36124001 PMCID: PMC9479141 DOI: 10.1021/acsmaterialsau.2c00040] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 02/08/2023]
Abstract
Implant-associated infections arising from biofilm development are known to have detrimental effects with compromised quality of life for the patients, implying a progressing issue in healthcare. It has been a struggle for more than 50 years for the biomaterials field to achieve long-term success of medical implants by discouraging bacterial and protein adhesion without adversely affecting the surrounding tissue and cell functions. However, the rate of infections associated with medical devices is continuously escalating because of the intricate nature of bacterial biofilms, antibiotic resistance, and the lack of ability of monofunctional antibacterial materials to prevent the colonization of bacteria on the device surface. For this reason, many current strategies are focused on the development of novel antibacterial surfaces with dual antimicrobial functionality. These surfaces are based on the combination of two components into one system that can eradicate attached bacteria (antibiotics, peptides, nitric oxide, ammonium salts, light, etc.) and also resist or release adhesion of bacteria (hydrophilic polymers, zwitterionic, antiadhesive, topography, bioinspired surfaces, etc.). This review aims to outline the progress made in the field of biomedical engineering and biomaterials for the development of multifunctional antibacterial biomedical devices. Additionally, principles for material design and fabrication are highlighted using characteristic examples, with a special focus on combinational nitric oxide-releasing biomedical interfaces. A brief perspective on future research directions for engineering of dual-function antibacterial surfaces is also presented.
Collapse
Affiliation(s)
- Manjyot
Kaur Chug
- School of Chemical, Materials
and Biomedical Engineering, University of
Georgia, Athens, Georgia 30602, United States
| | - Elizabeth J. Brisbois
- School of Chemical, Materials
and Biomedical Engineering, University of
Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
16
|
Dan W, Gao J, Qi X, Wang J, Dai J. Antibacterial quaternary ammonium agents: Chemical diversity and biological mechanism. Eur J Med Chem 2022; 243:114765. [PMID: 36116235 DOI: 10.1016/j.ejmech.2022.114765] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 01/04/2023]
Abstract
Bacterial infections have seriously threatened public health especially with the increasing resistance and the cliff-like decline of the number of newly approved antibacterial agents. Quaternary ammonium compounds (QACs) possess potent medicinal properties with 95 successfully marketed drugs, which also have a long history as antibacterial agents. In this review, we summarize the chemical diversity of antibacterial QACs, divided into chain-like and aromatic ring, reported over the past decade (2012 to mid-2022). Additionally, the structure-activity relationships, mainly covering hydrophobicity, charges and skeleton features, are discussed. In the cases where sufficient information is available, antibacterial mechanisms including biofilm, cell membrane, and intracellular targets are presented. It is hoped that this review will provide sufficient information for medicinal chemists to discover the new generation of antibacterial agents based on QACs.
Collapse
Affiliation(s)
- Wenjia Dan
- School of Life Science and Technology, Weifang Medical University, Shandong, China
| | - Jixiang Gao
- School of Life Science and Technology, Weifang Medical University, Shandong, China
| | - Xiaohui Qi
- School of Life Science and Technology, Weifang Medical University, Shandong, China
| | - Junru Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Shaanxi, China.
| | - Jiangkun Dai
- School of Life Science and Technology, Weifang Medical University, Shandong, China.
| |
Collapse
|
17
|
Zhuang Z, Meng Z, Li J, Shen P, Dai J, Lou X, Xia F, Tang BZ, Zhao Z. Antibacterial Theranostic Agents with Negligible Living Cell Invasiveness: AIE-Active Cationic Amphiphiles Regulated by Alkyl Chain Engineering. ACS NANO 2022; 16:11912-11930. [PMID: 35917549 DOI: 10.1021/acsnano.2c01721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
To address the threat of bacterial infection in the following post-antibiotic era, developing effective antibacterial approaches is of utmost urgency. Theranostic medicine integrating diagnosis and therapy is a promising protocol to fight against pathogenic bacteria. But numerous reported antibacterial theranostic materials are disclosed to be trapped in the excessive invasiveness to living mammal cells, leading to false positives and possible biosafety risks. Herein, a series of cationic pyridinium-substituted phosphindole oxide derivatives featuring aggregation-induced emission are designed, and alkyl chain engineering is conducted to finely tune their hydrophobicity and investigate their bioaffinity preference for living mammal cells and pathogenic bacteria. Most importantly, an efficient theranostic agent (PyBu-PIO) is acquired that is free from living cell invasiveness with negligible cytotoxicity and yet holds a good affinity for Gram-positive bacteria, including drug-resistant strains, with a superior inactivating effect. Externally applying PyBu-PIO onto Gram-positive bacteria-infected skin wounds can achieve creditable imaging effects and successfully accelerate the healing processes with reliable biosafety. This work proposes living cell invasiveness as a criterion for antibacterial theranostic materials and provides important enlightenment for the design of antibacterial theranostic materials.
Collapse
Affiliation(s)
- Zeyan Zhuang
- State Key Laboratory of Luminescent Materials and Devices, Key Laboratory of Luminescence from Molecular Aggregates of Guangdong Province, South China University of Technology, Guangzhou 510640, China
| | - Zijuan Meng
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Jianqing Li
- State Key Laboratory of Luminescent Materials and Devices, Key Laboratory of Luminescence from Molecular Aggregates of Guangdong Province, South China University of Technology, Guangzhou 510640, China
| | - Pingchuan Shen
- State Key Laboratory of Luminescent Materials and Devices, Key Laboratory of Luminescence from Molecular Aggregates of Guangdong Province, South China University of Technology, Guangzhou 510640, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaoding Lou
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Fan Xia
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Zujin Zhao
- State Key Laboratory of Luminescent Materials and Devices, Key Laboratory of Luminescence from Molecular Aggregates of Guangdong Province, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
18
|
Crnčević D, Krce L, Cvitković M, Brkljača Z, Sabljić A, Vuko E, Primožič I, Odžak R, Šprung M. New Membrane Active Antibacterial and Antiviral Amphiphiles Derived from Heterocyclic Backbone of Pyridinium-4-Aldoxime. Pharmaceuticals (Basel) 2022; 15:ph15070775. [PMID: 35890073 PMCID: PMC9315884 DOI: 10.3390/ph15070775] [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: 05/31/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
Quaternary ammonium salts (QAS) are irreplaceable membrane-active antimicrobial agents that have been widely used for nearly a century. Cetylpyridinium chloride (CPC) is one of the most potent QAS. However, recent data from the literature indicate that CPC activity against resistant bacterial strains is decreasing. The major QAS resistance pathway involves the QacR dimer, which regulates efflux pump expression. A plausible approach to address this issue is to structurally modify the CPC structure by adding other biologically active functional groups. Here, a series of QAS based on pyridine-4-aldoxime were synthesized, characterized, and tested for antimicrobial activity in vitro. Although we obtained several potent antiviral candidates, these candidates had lower antibacterial activity than CPC and were not toxic to human cell lines. We found that the addition of an oxime group to the pyridine backbone resulted in derivatives with large topological polar surfaces and with unfavorable cLog P values. Investigation of the antibacterial mode of action, involving the cell membrane, revealed altered cell morphologies in terms of corrugated and/or disrupted surface, while 87% of the cells studied exhibited a permeabilized membrane after 3 h of treatment at 4 × minimum inhibitory concentration (MIC). Molecular dynamic (MD) simulations of the interaction of QacR with a representative candidate showed rapid dimer disruption, whereas this was not observed for QacR and QacR bound to the structural analog CPC. This might explain the lower bioactivity of our compounds, as they are likely to cause premature expression of efflux pumps and thus activation of resistance.
Collapse
Affiliation(s)
- Doris Crnčević
- Department of Chemistry, Faculty of Science, University of Split, R. Bošković 33, 21 000 Split, Croatia; (D.C.); (A.S.)
- Doctoral Study of Biophysics, Faculty of Science, University of Split, R. Bošković 33, 21 000 Split, Croatia
| | - Lucija Krce
- Department of Physics, Faculty of Science, University of Split, R. Bošković 33, 21 000 Split, Croatia; (L.K.); (M.C.)
| | - Mislav Cvitković
- Department of Physics, Faculty of Science, University of Split, R. Bošković 33, 21 000 Split, Croatia; (L.K.); (M.C.)
| | - Zlatko Brkljača
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička c. 54, 10 000 Zagreb, Croatia;
- Selvita Ltd., Prilaz Baruna Filipovića 29, 10 000 Zagreb, Croatia
| | - Antonio Sabljić
- Department of Chemistry, Faculty of Science, University of Split, R. Bošković 33, 21 000 Split, Croatia; (D.C.); (A.S.)
- Doctoral Study of Biophysics, Faculty of Science, University of Split, R. Bošković 33, 21 000 Split, Croatia
| | - Elma Vuko
- Department of Biology, Faculty of Science, University of Split, R. Bošković 33, 21 000 Split, Croatia;
| | - Ines Primožič
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, 10 000 Zagreb, Croatia;
| | - Renata Odžak
- Department of Chemistry, Faculty of Science, University of Split, R. Bošković 33, 21 000 Split, Croatia; (D.C.); (A.S.)
- Correspondence: (R.O.); (M.Š.)
| | - Matilda Šprung
- Department of Chemistry, Faculty of Science, University of Split, R. Bošković 33, 21 000 Split, Croatia; (D.C.); (A.S.)
- Correspondence: (R.O.); (M.Š.)
| |
Collapse
|
19
|
Fischer M, Müller P, Scheidt HA, Luck M. Drug-Membrane Interactions: Effects of Virus-Specific RNA-Dependent RNA Polymerase Inhibitors Remdesivir and Favipiravir on the Structure of Lipid Bilayers. Biochemistry 2022; 61:1392-1403. [PMID: 35731976 DOI: 10.1021/acs.biochem.2c00042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The two RNA-dependent RNA polymerase inhibitors remdesivir and favipiravir were originally developed and approved as broad-spectrum antiviral drugs for the treatment of harmful viral infections such as Ebola and influenza. With the outbreak of the global SARS-CoV-2 pandemic, the two drugs were repurposed for the treatment of COVID-19 patients. Clinical studies suggested that the efficacy of the drugs is enhanced in the case of an early or even prophylactic application. Because the contact between drug molecules and the plasma membrane is essential for a successful permeation process of the substances and therefore for their intracellular efficiency, drug-induced effects on the membrane structure are likely and have already been shown for other substances. We investigated the impact of remdesivir and favipiravir on lipid bilayers in model and cell membranes via several biophysical approaches. The measurements revealed that the embedding of remdesivir molecules in the lipid bilayer results in a disturbance of the membrane structure of the tested phospholipid vesicles. Nevertheless, in a cell-based assay, the presence of remdesivir induced only weak hemolysis of the treated erythrocytes. In contrast, no experimental indication for an effect on the structure and integrity of the membrane was detected in the case of favipiravir. Regarding potential prophylactic or accompanying use of the drugs in the therapy of COVID-19, the physiologically relevant impacts associated with the drug-induced structural modifications of the membrane might be important to understand side effects and/or low effectivities.
Collapse
Affiliation(s)
- Markus Fischer
- Institute for Medical Physics and Biophysics, Leipzig University, 04107 Leipzig, Germany
| | - Peter Müller
- Institute of Biology, Biophysical Chemistry, Humboldt-Universität zu Berlin, 10115 Berlin, Germany
| | - Holger A Scheidt
- Institute for Medical Physics and Biophysics, Leipzig University, 04107 Leipzig, Germany
| | - Meike Luck
- Institute of Biology, Biophysical Chemistry, Humboldt-Universität zu Berlin, 10115 Berlin, Germany
| |
Collapse
|
20
|
Isoquinolinedione-urea hybrids: Synthesis, antibacterial evaluation, drug-likeness, molecular docking and DFT studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
21
|
Khan NA, Kaur N, Owens P, Thomas OP, Boyd A. Bis-Indole Alkaloids Isolated from the Sponge Spongosorites calcicola Disrupt Cell Membranes of MRSA. Int J Mol Sci 2022; 23:ijms23041991. [PMID: 35216106 PMCID: PMC8874442 DOI: 10.3390/ijms23041991] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 02/01/2023] Open
Abstract
Antimicrobial resistance (AMR) is a global health challenge with methicillin resistant Staphylococcus aureus (MRSA), a leading cause of nosocomial infection. In the search for novel antibiotics, marine sponges have become model organisms as they produce diverse bioactive compounds. We investigated and compared the antibacterial potential of 3 bis-indole alkaloids—bromodeoxytopsentin, bromotopsentin and spongotine A—isolated from the Northeastern Atlantic sponge Spongosorites calcicola. Antimicrobial activity was determined by MIC and time-kill assays. The mechanism of action of bis-indoles was assessed using bacterial cytological profiling via fluorescence microscopy. Finally, we investigated the ability of bis-indole alkaloids to decrease the cytotoxicity of pathogens upon co-incubation with HeLa cells through the measurement of mammalian cell lysis. The bis-indoles were bactericidal to clinically relevant Gram-positive pathogens including MRSA and to the Gram-negative gastroenteric pathogen Vibrio parahaemolyticus. Furthermore, the alkaloids were synergistic in combination with conventional antibiotics. Antimicrobial activity of the bis-indole alkaloids was due to rapid disruption and permeabilization of the bacterial cell membrane. Significantly, the bis-indoles reduced pathogen cytotoxicity toward mammalian cells, indicating their ability to prevent bacterial virulence. In conclusion, sponge bis-indole alkaloids are membrane-permeabilizing agents that represent good antibiotic candidates because of their potency against Gram-positive and Gram-negative bacterial pathogens.
Collapse
Affiliation(s)
- Neyaz A. Khan
- Discipline of Microbiology, School of Natural Sciences and Ryan Institute, National University of Ireland Galway, H91 TK33 Galway, Ireland;
| | - Navdeep Kaur
- Discipline of Chemistry, School of Biological and Chemical Science and Ryan Institute, National University of Ireland Galway, H91 TK33 Galway, Ireland; (N.K.); (O.P.T.)
| | - Peter Owens
- Centre for Microscopy and Imaging, National University of Ireland, Galway, H91 TK33 Galway, Ireland;
| | - Olivier P. Thomas
- Discipline of Chemistry, School of Biological and Chemical Science and Ryan Institute, National University of Ireland Galway, H91 TK33 Galway, Ireland; (N.K.); (O.P.T.)
| | - Aoife Boyd
- Discipline of Microbiology, School of Natural Sciences and Ryan Institute, National University of Ireland Galway, H91 TK33 Galway, Ireland;
- Correspondence: ; Tel.: +353-(0)91-492404
| |
Collapse
|
22
|
Limwongyut J, Moreland AS, Nie C, Read de Alaniz J, Bazan GC. Amide Moieties Modulate the Antimicrobial Activities of Conjugated Oligoelectrolytes against Gram-negative Bacteria. Chemistry 2022; 11:e202100260. [PMID: 35133087 PMCID: PMC8822875 DOI: 10.1002/open.202100260] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/23/2021] [Indexed: 11/18/2022]
Abstract
Cationic conjugated oligoelectrolytes (COEs) are a class of compounds that can be tailored to achieve relevant in vitro antimicrobial properties with relatively low cytotoxicity against mammalian cells. Three distyrylbenzene‐based COEs were designed containing amide functional groups on the side chains. Their properties were compared to two representative COEs with only quaternary ammonium groups. The optimal compound, COE2−3C−C3‐Apropyl, has an antimicrobial efficacy against Escherichia coli with an MIC=2 μg mL−1, even in the presence of human serum albumin low cytotoxicity (IC50=740 μg mL−1) and minimal hemolytic activity. Moreover, we find that amide groups increase interactions between COEs and a bacterial lipid mimic based on calcein leakage assay and allow COEs to readily permeabilize the cytoplasmic membrane of E. coli. These findings suggest that hydrogen bond forming moieties can be further applied in the molecular design of antimicrobial COEs to further improve their selectivity towards bacteria.
Collapse
Affiliation(s)
- Jakkarin Limwongyut
- Center for Polymers and Organic Solids, Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Alex S Moreland
- Center for Polymers and Organic Solids, Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Chenyao Nie
- Department of Chemistry and Chemical Engineering, National University of Singapore, Singapore, 117543, Singapore
| | - Javier Read de Alaniz
- Center for Polymers and Organic Solids, Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Guillermo C Bazan
- Center for Polymers and Organic Solids, Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.,Department of Chemistry and Chemical Engineering, National University of Singapore, Singapore, 117543, Singapore
| |
Collapse
|
23
|
Li D, Gao S, Ye K, Wang Q, Xie C, Wu W, Feng L, Jiang L, Zheng K, Pang Q. Membrane-active La(III) and Ce(III) complexes as potent antibacterial agents: synthesis, characterization, in vitro, in silico, and in vivo studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131595] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
24
|
Pang Z, Li Q, Jia Y, Yan W, Qi J, Guo Y, Hu F, Zhou D, Jiang X. Controlling the pyridinium-zwitterionic ligand ratio on atomically precise gold nanoclusters allowing for eradicating Gram-positive drug-resistant bacteria and retaining biocompatibility. Chem Sci 2021; 12:14871-14882. [PMID: 34820103 PMCID: PMC8597861 DOI: 10.1039/d1sc03056f] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/24/2021] [Indexed: 01/09/2023] Open
Abstract
Infections caused by multidrug-resistant (MDR) bacteria are an increasing global healthcare concern. In this study, we developed a dual-ligand-functionalised Au25(SR1) x (SR2)18-x -type gold nanocluster and determined its antibacterial activity against MDR bacterial strains. The pyridinium ligand (SR1) provided bactericidal potency and the zwitterionic ligand (SR2) enhanced the stability and biocompatibility. By optimising the ligand ratio, our gold nanocluster could effectively kill MDR Gram-positive bacteria via multiple antibacterial actions, including inducing bacterial aggregation, disrupting bacterial membrane integrity and potential, and generating reactive oxygen species. Moreover, combining the optimised gold nanocluster with common antibiotics could significantly enhance the antibacterial activity against MDR bacteria both in in vitro and animal models of skin infections. Furthermore, the fluorescence of the gold nanocluster at the second near-infrared (NIR-II) biological window allowed for the monitoring of its biodistribution and body clearance, which confirmed that the gold nanoclusters had good renal clearance and biocompatibility. This study provides a new strategy to combat the MDR challenge using multifunctional gold nanomaterials.
Collapse
Affiliation(s)
- Zeyang Pang
- Department of Biomedical Engineering, Southern University of Science and Technology No 1088, Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Qizhen Li
- Department of Biomedical Engineering, Southern University of Science and Technology No 1088, Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Yuexiao Jia
- Department of Biomedical Engineering, Southern University of Science and Technology No 1088, Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Weixiao Yan
- Department of Biomedical Engineering, Southern University of Science and Technology No 1088, Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Jie Qi
- Department of Biomedical Engineering, Southern University of Science and Technology No 1088, Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Yuan Guo
- School of Food Science and Nutrition and Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Fupin Hu
- Institute of Antibiotics, Huashan Hospital, Fudan University Shanghai 200040 P. R. China
| | - Dejian Zhou
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology No 1088, Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| |
Collapse
|
25
|
Volynsky PE, Smirnova AI, Akimov SA, Sokolov SS, Severin FF. The Membrane-Water Partition Coefficients of Antifungal, but Not Antibacterial, Membrane-Active Compounds Are Similar. Front Microbiol 2021; 12:756408. [PMID: 34803981 PMCID: PMC8602886 DOI: 10.3389/fmicb.2021.756408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Pavel E. Volynsky
- Laboratory of Biomolecular Modeling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (RAS), Moscow, Russia
| | - Alexandra I. Smirnova
- Department of Molecular Energetics of Microorganisms, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Sergey A. Akimov
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences (RAS), Moscow, Russia
| | - Svyatoslav S. Sokolov
- Department of Molecular Energetics of Microorganisms, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Fedor F. Severin
- Department of Molecular Energetics of Microorganisms, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
26
|
S-alkylated thiosemicarbazone derivatives: Synthesis, crystal structure determination, antimicrobial activity evaluation and molecular docking studies. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130674] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
27
|
Pearce AN, Chen D, Edmeades LR, Cadelis MM, Troudi A, Brunel JM, Bourguet-Kondracki ML, Copp BR. Repurposing primaquine as a polyamine conjugate to become an antibiotic adjuvant. Bioorg Med Chem 2021; 38:116110. [PMID: 33831695 DOI: 10.1016/j.bmc.2021.116110] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/04/2021] [Accepted: 03/06/2021] [Indexed: 11/17/2022]
Abstract
In our search for new antibiotic adjuvants as a novel strategy to deal with the emergence of multi-drug resistant (MDR) bacteria, a series of succinylprimaquine-polyamine (SPQ-PA) conjugates and derivatives of a cationic amphiphilic nature have been prepared. Evaluation of these primaquine conjugates for intrinsic antimicrobial properties and the ability to restore the antibiotic activity of doxycycline identified two derivatives, SPQ-PA3-8-3 and SPQ-PA3-10-3 that exhibited intrinsic activity against the Gram-positive bacteria Staphylococcus aureus and the yeast Cryptococcus neoformans. None of the analogues were active against the Gram-negative bacterium Pseudomonas aeruginosa. However, in the presence of a sub-therapeutic amount of doxycycline (4.5 µM), both SPQ-PA3-4-3 and SPQ-PA3-10-3 compounds displayed potent antibiotic adjuvant properties against P. aeruginosa, with MIC's of 6.25 µM. A series of derivatives were prepared to investigate the structure-activity relationship that explored the influence of both a simplified aryl lipophilic substituent and variation of the length of the polyamine scaffold on observed intrinsic antimicrobial properties and the ability to potentiate the action of doxycycline against P. aeruginosa.
Collapse
Affiliation(s)
- A Norrie Pearce
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Dan Chen
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Liam R Edmeades
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Melissa M Cadelis
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Azza Troudi
- UMR_MD1, U-1261, Aix Marseille Universite, INSERM, SSA, MCT, 13385 Marseille, France
| | - Jean Michel Brunel
- UMR_MD1, U-1261, Aix Marseille Universite, INSERM, SSA, MCT, 13385 Marseille, France
| | - Marie-Lise Bourguet-Kondracki
- Laboratoire Molécules de Communication et Adaptation des Micro-organismes, UMR 7245 CNRS, Muséum National d'Histoire Naturelle, 57 rue Cuvier (C.P. 54), 75005 Paris, France
| | - Brent R Copp
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
28
|
Feliciano JA, Leitgeb AJ, Schrank CL, Allen RA, Minbiole KPC, Wuest WM, Carden RG. Trivalent sulfonium compounds (TSCs): Tetrahydrothiophene-based amphiphiles exhibit similar antimicrobial activity to analogous ammonium-based amphiphiles. Bioorg Med Chem Lett 2021; 37:127809. [PMID: 33516911 PMCID: PMC7965331 DOI: 10.1016/j.bmcl.2021.127809] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/28/2020] [Accepted: 01/15/2021] [Indexed: 10/22/2022]
Abstract
Recent advances in the development of quaternary ammonium compounds (QACs) have focused on new structural motifs to increase bioactivity, but significantly less studied has been the change from ammonium- to sulfonium-based disinfectants. Herein, we report the synthesis of structurally analogous series of quaternary ammonium and trivalent sulfonium compounds (TSCs). The bioactivity profiles of these compounds generally mirror each other, and the antibacterial activity of sulfonium-based THT-18 was found to be comparable to the commercial disinfectant, BAC. The development of these compounds presents a new avenue for further study of disinfectants to combat the growing threat of bacterial resistance.
Collapse
Affiliation(s)
- Javier A Feliciano
- Department of Chemistry, Villanova University, Villanova, PA 19085, United States
| | - Austin J Leitgeb
- Department of Chemistry, Villanova University, Villanova, PA 19085, United States
| | | | - Ryan A Allen
- Department of Chemistry, Emory University, Atlanta, GA 30322, Unites States
| | - Kevin P C Minbiole
- Department of Chemistry, Villanova University, Villanova, PA 19085, United States
| | - William M Wuest
- Department of Chemistry, Emory University, Atlanta, GA 30322, Unites States.
| | - Robert G Carden
- Department of Chemistry, Villanova University, Villanova, PA 19085, United States.
| |
Collapse
|
29
|
Meşeli T, Doğan ŞD, Gündüz MG, Kökbudak Z, Skaro Bogojevic S, Noonan T, Vojnovic S, Wolber G, Nikodinovic-Runic J. Design, synthesis, antibacterial activity evaluation and molecular modeling studies of new sulfonamides containing a sulfathiazole moiety. NEW J CHEM 2021. [DOI: 10.1039/d1nj00150g] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Molecular modifications on sulfathiazole to overcome sulfonamide resistance: the discovery of selective antibacterial agents against Staphylococcus aureus.
Collapse
Affiliation(s)
- Tuğba Meşeli
- Department of Chemistry
- Faculty of Science
- Erciyes University
- Kayseri
- Turkey
| | - Şengül Dilem Doğan
- Department of Basic Sciences
- Faculty of Pharmacy
- Erciyes University
- Kayseri
- Turkey
| | - Miyase Gözde Gündüz
- Department of Pharmaceutical Chemistry
- Faculty of Pharmacy
- Hacettepe University
- Sıhhiye
- Turkey
| | - Zülbiye Kökbudak
- Department of Chemistry
- Faculty of Science
- Erciyes University
- Kayseri
- Turkey
| | - Sanja Skaro Bogojevic
- Institute of Molecular Genetics and Genetic Engineering
- University of Belgrade
- 11000 Belgrade
- Serbia
| | - Theresa Noonan
- Department of Pharmaceutical and Medicinal Chemistry
- Institute of Pharmacy
- Freie Universität Berlin
- 14195 Berlin
- Germany
| | - Sandra Vojnovic
- Institute of Molecular Genetics and Genetic Engineering
- University of Belgrade
- 11000 Belgrade
- Serbia
| | - Gerhard Wolber
- Department of Pharmaceutical and Medicinal Chemistry
- Institute of Pharmacy
- Freie Universität Berlin
- 14195 Berlin
- Germany
| | | |
Collapse
|
30
|
Nural Y, Ozdemir S, Doluca O, Demir B, Yalcin MS, Atabey H, Kanat B, Erat S, Sari H, Seferoglu Z. Synthesis, biological properties, and acid dissociation constant of novel naphthoquinone–triazole hybrids. Bioorg Chem 2020; 105:104441. [DOI: 10.1016/j.bioorg.2020.104441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/17/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022]
|
31
|
Gündüz MG, Uğur SB, Güney F, Özkul C, Krishna VS, Kaya S, Sriram D, Doğan ŞD. 1,3-Disubstituted urea derivatives: Synthesis, antimicrobial activity evaluation and in silico studies. Bioorg Chem 2020; 102:104104. [PMID: 32736149 DOI: 10.1016/j.bioorg.2020.104104] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/29/2020] [Accepted: 07/14/2020] [Indexed: 01/09/2023]
Abstract
The development of new antimicrobial compounds is in high demand to overcome the emerging drug resistance against infectious microbial pathogens. In the present study, we carried out the extensive antimicrobial screening of disubstituted urea derivatives. In addition to the classical synthesis of urea compounds by the reaction of amines and isocyanates, we also applied a new route including bromination, oxidation and azidination reactions, respectively, to convert 2-amino-3-methylpyridine to 1,3-disubstituted urea derivatives using various amines. The evaluation of antimicrobial activities against various bacterial strains, Candida albicans as well as Mycobacterium tuberculosis resulted in the discovery of new active molecules. Among them, two compounds, which have the lowest MIC values on Pseudomonas aeruginosa, were further evaluated for their inhibition capacities of biofilm formation. In order to evaluate their potential mechanism of biofilm inhibition, these two compounds were docked into the active site of LasR, which is the transcriptional regulator of bacterial signaling mechanism known as quorum sensing. Finally, the theoretical parameters of the bioactive molecules were calculated to establish their drug-likeness properties.
Collapse
Affiliation(s)
- Miyase Gözde Gündüz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Sıhhiye, 06100 Ankara, Turkey
| | - Sümeyye Buran Uğur
- Department of Basic Sciences, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Funda Güney
- Department of Basic Sciences, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Ceren Özkul
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Hacettepe University, Sıhhiye, 06100 Ankara, Turkey
| | - Vagolu Siva Krishna
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad 500078, India
| | - Serdal Kaya
- Department of Chemistry, Faculty of Arts and Sciences, Giresun University, 28200 Giresun, Turkey; Department of Aeronautical, Faculty of Aviation and Space Sciences, Necmettin Erbakan University, 42140 Konya, Turkey
| | - Dharmarajan Sriram
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad 500078, India
| | - Şengül Dilem Doğan
- Department of Basic Sciences, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey.
| |
Collapse
|
32
|
Karataş MO, Noma SAA, Gürses C, Balcıoğlu S, Ateş B, Alıcı B, Çakır Ü. Water Soluble Coumarin Quaternary Ammonium Chlorides: Synthesis and Biological Evaluation. Chem Biodivers 2020; 17:e2000258. [PMID: 32638471 DOI: 10.1002/cbdv.202000258] [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: 04/03/2020] [Accepted: 07/07/2020] [Indexed: 11/11/2022]
Abstract
In the present study, coumarin-bearing three pyridinium and three tetra-alkyl ammonium salts were synthesized. The compounds were fully characterized by 1 H- and 13 C-NMR, LC/MS and IR spectroscopic methods and elemental analyses. The cytotoxic properties of all compounds were tested against human liver cancer (HepG2), human colorectal cancer (Caco-2) and non-cancer mouse fibroblast (L-929) cell lines. Some compounds performed comparable cytotoxicity with standard drug cisplatin. Antibacterial properties of the compounds were tested against Gram-negative Escherichia coli and Gram-positive Bacillus subtilis bacteria, but the compounds did not have any antibacterial effect against both bacteria. Enzyme inhibitory properties of all compounds were tested on the activities of human carbonic anhydrase I and II, and xanthine oxidase. All compounds inhibited both enzymes more effectively than standard drugs, acetazolamide and allopurinol, respectively. The biological evaluation results showed that ionic and water soluble coumarin derivatives are promising structures for further investigations especially on enzyme inhibition field.
Collapse
Affiliation(s)
- Mert O Karataş
- İnönü University, Faculty of Science, Department of Chemistry, 9044280, Malatya, Turkey
| | - Samir A A Noma
- İnönü University, Faculty of Science, Department of Chemistry, 9044280, Malatya, Turkey
| | - Canbolat Gürses
- İnönü University, Faculty of Science, Department of Molecular Biology and Genetics, 9044280, Malatya, Turkey
| | - Sevgi Balcıoğlu
- Fırat University, Department of Food Processing, 9023600, Elazığ, Turkey
| | - Burhan Ateş
- İnönü University, Faculty of Science, Department of Chemistry, 9044280, Malatya, Turkey
| | - Bülent Alıcı
- İnönü University, Faculty of Science, Department of Chemistry, 9044280, Malatya, Turkey
| | - Ümit Çakır
- Balıkesir University, Faculty of Science, Department of Chemistry, 9010440, Balıkesir, Turkey
| |
Collapse
|
33
|
The Neutrally Charged Diarylurea Compound PQ401 Kills Antibiotic-Resistant and Antibiotic-Tolerant Staphylococcus aureus. mBio 2020; 11:mBio.01140-20. [PMID: 32605985 PMCID: PMC7327171 DOI: 10.1128/mbio.01140-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Membrane-damaging antimicrobial agents have great potential to treat multidrug-resistant or multidrug-tolerant bacteria against which conventional antibiotics are not effective. However, their therapeutic applications are often hampered due to their low selectivity to bacterial over mammalian membranes or their potential for cross-resistance to a broad spectrum of cationic membrane-active antimicrobial agents. We discovered that the diarylurea derivative compound PQ401 has antimicrobial potency against multidrug-resistant and multidrug-tolerant Staphylococcus aureus. PQ401 selectively disrupts bacterial membrane lipid bilayers in comparison to mammalian membranes. Unlike cationic membrane-active antimicrobials, the neutral form of PQ401 rather than its cationic form exhibits maximum membrane activity. Overall, our results demonstrate that PQ401 could be a promising lead compound that overcomes the current limitations of membrane selectivity and cross-resistance. Also, this work provides deeper insight into the design and development of new noncharged membrane-targeting therapeutics to combat hard-to-cure bacterial infections. Resistance or tolerance to traditional antibiotics is a challenging issue in antimicrobial chemotherapy. Moreover, traditional bactericidal antibiotics kill only actively growing bacterial cells, whereas nongrowing metabolically inactive cells are tolerant to and therefore “persist” in the presence of legacy antibiotics. Here, we report that the diarylurea derivative PQ401, previously characterized as an inhibitor of the insulin-like growth factor I receptor, kills both antibiotic-resistant and nongrowing antibiotic-tolerant methicillin-resistant Staphylococcus aureus (MRSA) by lipid bilayer disruption. PQ401 showed several beneficial properties as an antimicrobial lead compound, including rapid killing kinetics, low probability for resistance development, high selectivity to bacterial membranes compared to mammalian membranes, and synergism with gentamicin. In contrast to well-studied membrane-disrupting cationic antimicrobial low-molecular-weight compounds and peptides, molecular dynamic simulations supported by efficacy data demonstrate that the neutral form of PQ401 penetrates and subsequently embeds into bacterial lipid bilayers more effectively than the cationic form. Lastly, PQ401 showed efficacy in both the Caenorhabditis elegans and Galleria mellonella models of MRSA infection. These data suggest that PQ401 may be a lead candidate for repurposing as a membrane-active antimicrobial and has potential for further development as a human antibacterial therapeutic for difficult-to-treat infections caused by both drug-resistant and -tolerant S. aureus.
Collapse
|
34
|
Alkhalifa S, Jennings MC, Granata D, Klein M, Wuest WM, Minbiole KPC, Carnevale V. Analysis of the Destabilization of Bacterial Membranes by Quaternary Ammonium Compounds: A Combined Experimental and Computational Study. Chembiochem 2020; 21:1510-1516. [PMID: 31859426 PMCID: PMC7237276 DOI: 10.1002/cbic.201900698] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Indexed: 12/24/2022]
Abstract
The mechanism of action of quaternary ammonium compound (QAC) antiseptics has long been assumed to be straightforward membrane disruption, although the process of approaching and entering the membrane has little modeling precedent. Furthermore, questions have more recently arisen regarding bacterial resistance mechanisms, and why select classes of QACs (specifically, multicationic QACs) are less prone to resistance. In order to better understand such subtleties, a series of molecular dynamics simulations were utilized to help identify these molecular determinants, directly comparing mono-, bis-, and triscationic QACs in simulated membrane intercalation models. Three distinct membranes were simulated, mimicking the surfaces of Escherichia coli and Staphylococcus aureus, as well as a neutral phospholipid control. By analyzing the resulting trajectories in the form of a timeseries analysis, insight was gleaned regarding the significant steps and interactions involved in the destabilization of phospholipid bilayers within the bacterial membranes. Finally, to more specifically probe the effect of the hydrophobic section of the amphiphile that presumably penetrates the membrane, a series of alkyl- and ester-based biscationic quaternary ammonium compounds were prepared, tested for antimicrobial activity against both Gram-positive and Gram-negative bacteria, and modeled.
Collapse
Affiliation(s)
- Saleh Alkhalifa
- Department of Chemistry, Villanova University, 800 East Lancaster Avenue, Villanova, PA, 19085, USA
| | - Megan C Jennings
- Department of Chemistry, Temple University, 1901 North 13th Street, Philadelphia, PA, 19122, USA
| | - Daniele Granata
- Department of Chemistry, Temple University, 1901 North 13th Street, Philadelphia, PA, 19122, USA
| | - Michael Klein
- Department of Chemistry, Temple University, 1901 North 13th Street, Philadelphia, PA, 19122, USA
| | - William M Wuest
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, GA, 30322, USA
- Emory Antibiotic Resistance Center, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA, 30322, USA
| | - Kevin P C Minbiole
- Department of Chemistry, Villanova University, 800 East Lancaster Avenue, Villanova, PA, 19085, USA
| | - Vincenzo Carnevale
- Department of Chemistry, Temple University, 1901 North 13th Street, Philadelphia, PA, 19122, USA
| |
Collapse
|
35
|
Leitgeb AJ, Feliciano JA, Sanchez HA, Allen RA, Morrison KR, Sommers KJ, Carden RG, Wuest WM, Minbiole KPC. Further Investigations into Rigidity-Activity Relationships in BisQAC Amphiphilic Antiseptics. ChemMedChem 2020; 15:667-670. [PMID: 32022457 PMCID: PMC8322965 DOI: 10.1002/cmdc.201900662] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/09/2020] [Indexed: 12/11/2022]
Abstract
Thirty-six biscationic quaternary ammonium compounds were efficiently synthesized in one step to examine the effect of molecular geometry of two-carbon linkers on antimicrobial activity. The synthesized compounds showed strong antimicrobial activity against a panel of both Gram-positive and Gram-negative bacteria, including methicillin-resistant Staphylococcus aureus (MRSA). While the linker geometry showed only a modest correlation with antimicrobial activity, several of the synthesized bisQACs are promising potential antiseptics due to good antimicrobial activity (MIC≤2 μM) and their higher therapeutic indices compared to previously reported QACs.
Collapse
Affiliation(s)
- Austin J. Leitgeb
- Department of Chemistry, Villanova University, Villanova, PA, 19085 (USA)
| | | | - Hugo A. Sanchez
- Department of Chemistry, Villanova University, Villanova, PA, 19085 (USA)
| | - Ryan A. Allen
- Department of Chemistry, Villanova University, Villanova, PA, 19085 (USA)
| | | | - Kyle J. Sommers
- Department of Chemistry, Emory University, Atlanta, GA, 30322 (USA)
| | - Robert G. Carden
- Department of Chemistry, Villanova University, Villanova, PA, 19085 (USA)
| | - William M. Wuest
- Department of Chemistry, Emory University, Atlanta, GA, 30322 (USA)
| | | |
Collapse
|
36
|
Karataş MO, Günal S, Mansur A, Alıcı B, Özdemir İ. Catechol-bearing imidazolium and benzimidazolium chlorides as promising antimicrobial agents. Arch Pharm (Weinheim) 2020; 353:e2000013. [PMID: 32301169 DOI: 10.1002/ardp.202000013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 11/06/2022]
Abstract
Catechol-containing imidazolium (four) and benzimidazolium chlorides (eight) were synthesized to evaluate their antimicrobial properties. All the compounds were fully characterized using 1 H and 13 C nuclear magnetic resonance, liquid chromatography-mass spectrometry, infrared spectroscopic methods, and elemental analyses. Antimicrobial activities of the compounds were tested against the bacteria Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumannii, Klebsiella pneumoniae, Staphylococcus aureus, methicillin-resistant S. aureus (MRSA), Enterococcus faecalis, and the fungal strains Candida albicans and Candida glabrata, and promising results were achieved. The two most important benzyl-substituted benzimidazolium chlorides, 3l and 3k, showed comparable activity to vancomycin against MRSA.
Collapse
Affiliation(s)
- Mert O Karataş
- Department of Chemistry, Faculty of Science, İnönü University, Malatya, Turkey
| | - Selami Günal
- Department of Microbiology, Faculty of Pharmacy, İnönü University, Malatya, Turkey
| | - Ahmet Mansur
- Department of Microbiology, Faculty of Pharmacy, İnönü University, Malatya, Turkey
| | - Bülent Alıcı
- Department of Chemistry, Faculty of Science, İnönü University, Malatya, Turkey
| | - İsmail Özdemir
- Department of Chemistry, Faculty of Science, İnönü University, Malatya, Turkey.,Catalysis Research and Application Centre, İnönü University, Malatya, Turkey
| |
Collapse
|
37
|
Fan T, Guo W, Shao T, Zhou W, Hu P, Liu M, Chen Y, Yi Z. Design, synthesis and evaluation of phenylthiazole and phenylthiophene pyrimidindiamine derivatives targeting the bacterial membrane. Eur J Med Chem 2020; 190:112141. [PMID: 32078862 DOI: 10.1016/j.ejmech.2020.112141] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 01/17/2023]
Abstract
As the continuous rise in the incidence of antibiotic resistance, it is urgent to develop novel chemical scaffolds with antibacterial activities to control the spread of resistance to conventional antibiotics. In this study, a series of phenylthiazole and phenylthiophene pyrimidindiamine derivatives were designed and synthesized by modifying the hit compound (N2-isobutyl-N4-((4-methyl-2-phenylthiazol-5-yl)methyl) pyrimidine-2,4-diamine) and their antibacterial activities were evaluated both in vitro and in vivo. Among the tested compounds, compound 14g (N4-((5-(3-bromophenyl)thiophen-2-yl)methyl)-N2-isobutylpyrimidine-2,4-diamine) displayed the best antibacterial activities, which was not only capable of inhibiting E. coli and S. aureus growth at concentrations as low as 2 and 3 μg/mL in vitro, but also efficacious in a mice model of bacteremia in vivo. Unlike conventional antibiotics, compound 14g was elucidated to mainly destroy the bacterial cell membrane, with the dissipation of membrane potential and leakage of contents, ultimately leading to cell death. The destruction of cell structure is challenging to induce bacterial resistance, which suggested that compound 14g may be a kind of promising alternatives to antibiotics against bacteria.
Collapse
Affiliation(s)
- Tingting Fan
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 200241, Shanghai, China; Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Weikai Guo
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Ting Shao
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Wenbo Zhou
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Pan Hu
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Mingyao Liu
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Yihua Chen
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 200241, Shanghai, China.
| | - Zhengfang Yi
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 200241, Shanghai, China.
| |
Collapse
|