1
|
Lu Y, You XY, Zhang Q, Lu QT, Hou JL, Cai Q. Enantioselective Total Synthesis of Fortimicin B. Angew Chem Int Ed Engl 2025; 64:e202424235. [PMID: 40079796 DOI: 10.1002/anie.202424235] [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: 12/11/2024] [Revised: 02/17/2025] [Accepted: 03/13/2025] [Indexed: 03/15/2025]
Abstract
Fortimicins, featuring a pseudodisaccharide scaffold, are an unusual class of aminoglycosides (AGs) with potent efficacy against several aminoglycoside-resistant bacterial strains. Notably, these molecules also exhibit lower inherent ototoxicity and nephrotoxicity than common aminoglycosides. Consequently, fortimicins are a promising type of protoypical molecules for the development of the next generation of aminoglycoside antibiotics. Here, we report the asymmetric total synthesis of fortimicin B in 12 steps (longest linear sequence, LLS) from readily available starting materials. An enantioselective Cu(II)-catalyzed inverse-electron-demand Diels-Alder (IEDDA) reaction of 2-pyrones and N-substituted 2-oxazolones was developed for the efficient synthesis of the fortamine fragment, which previously required a lengthy multistep synthesis owing to its complex stereochemistry. The 6-epi-purpurosamine B fragment was efficiently synthesized through a Cr(II)/Co(I)-mediated C─C bond coupling between aldehydes and alkyl halides. Within these two fragments, the stereoselective construction of the α-glycosidic bond of fortimicin B was realized via the gold(I)-catalyzed glycosylation. Overall, this study provides an efficient synthetic platform for future investigations into the structure-activity relationships of fortimicins.
Collapse
Affiliation(s)
- Yang Lu
- Department of Chemistry, Fudan University, 220 Handan Rd., Shanghai, 200433, China
| | - Xin-Yu You
- Department of Chemistry, Fudan University, 220 Handan Rd., Shanghai, 200433, China
| | - Qianwei Zhang
- Department of Chemistry, Fudan University, 220 Handan Rd., Shanghai, 200433, China
| | - Qi-Tao Lu
- Department of Chemistry, Fudan University, 220 Handan Rd., Shanghai, 200433, China
| | - Jun-Li Hou
- Department of Chemistry, Fudan University, 220 Handan Rd., Shanghai, 200433, China
| | - Quan Cai
- Department of Chemistry, Fudan University, 220 Handan Rd., Shanghai, 200433, China
| |
Collapse
|
2
|
Kushwaha D, Kushwaha AK, Kumar R, Chauhan D. Recent advances in the synthesis of Glycoconjugated heterocycles: A promising strategy for accessing bioactive compounds. Bioorg Chem 2025; 162:108559. [PMID: 40413973 DOI: 10.1016/j.bioorg.2025.108559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/27/2025]
Abstract
Glycoconjugation of biologically relevant heterocycles and natural products to create glycohybrids, combining the unique features of both structures, has emerged as a promising approach for the creation of carbohydrate-based therapeutics. This review presents a comprehensive overview of the glyco-heterocycles synthesized primarily over the past decade, offering in-depth insights into the synthetic methods employed. Additionally, the review delves into the biological activities exhibited by these molecules, with particular emphasis on the structural elements that influence their therapeutic potential. It covers the molecular hybridization of biologically privileged heterocycles-including thiadiazole, oxadiazole, pyrazole, imidazole, thiazolidine, pyridine, pyrimidine, indole, benzimidazole, benzothiazole, coumarin, quinoline, etc. with carbohydrates and explores their biological activity. By integrating insights into both synthetic strategies and bioactivity, this work aims to enhance the understanding of glyco-heterocycles as a versatile class of compounds for medicinal chemistry and drug development.
Collapse
Affiliation(s)
- Divya Kushwaha
- Department of Chemistry, MMV, Banaras Hindu University, Varanasi UP-221005, India.
| | - Ambuj Kumar Kushwaha
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi UP-221005, India
| | - Ravendra Kumar
- Department of Chemistry, MMV, Banaras Hindu University, Varanasi UP-221005, India
| | - Deepanshi Chauhan
- Department of Chemistry, MMV, Banaras Hindu University, Varanasi UP-221005, India
| |
Collapse
|
3
|
Shapovalova KS, Zatonsky GV, Razumova EA, Ipatova DA, Lukianov DA, Sergiev PV, Grammatikova NE, Tikhomirov AS, Shchekotikhin AE. Synthesis and Antibacterial Activity of New 6″-Modified Tobramycin Derivatives. Antibiotics (Basel) 2024; 13:1191. [PMID: 39766581 PMCID: PMC11672562 DOI: 10.3390/antibiotics13121191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Objectives: Aminoglycosides are one of the first classes of natural antibiotics which have not lost relevance due to their broad spectrum of action against Gram-positive, Gram-negative bacteria and mycobacteria. The high growth rate of antimicrobial resistance (AMR) together with the severe side effects of aminoglycosides increase the importance of developing improved semisynthetic derivatives. Methods: In this work, we proposed a synthetic route to new tobramycin derivatives modified at the 6″-position with aminoalkylamine or guanidinoalkylamine residues. Results: The antibacterial activity of the new compounds against reference strains of microorganisms was comparable to the parental tobramycin. In striking contrast to tobramycin (resistance index, >256), its 6″-modified derivatives were significantly more potent against resistant clinical isolates of P. aeruginosa strains (resistance index = 4-16) and they demonstrated a promising AMR circumvention in E. coli strains associated with mutations in the fusA gene encoding elongation factor G. All the obtained tobramycin derivatives exhibited reduced cytotoxicity for the eukaryotic HEK293T cells compared to the tobramycin and thereby they potentially may have improved therapeutic index. The proposed modification of the 6″-position of tobramycin does not change the mechanism of aminoglycoside's antibacterial activity: new compounds induced translation errors which resulted in the inhibition of protein synthesis in bacterial cells. Conclusions: Taken together, we can suggest that further modifications of the 6″-position of tobramycin may be beneficial for circumvention of AMR to aminoglycosides or used for conjugation with other molecules of interest.
Collapse
Affiliation(s)
- Kseniya S. Shapovalova
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow 119021, Russia; (K.S.S.); (G.V.Z.); (N.E.G.); (A.S.T.)
| | - Georgy V. Zatonsky
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow 119021, Russia; (K.S.S.); (G.V.Z.); (N.E.G.); (A.S.T.)
| | - Elizaveta A. Razumova
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119991, Russia; (E.A.R.); (D.A.I.); (D.A.L.); (P.V.S.)
| | - Daria A. Ipatova
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119991, Russia; (E.A.R.); (D.A.I.); (D.A.L.); (P.V.S.)
| | - Dmitrii A. Lukianov
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119991, Russia; (E.A.R.); (D.A.I.); (D.A.L.); (P.V.S.)
- Center for Molecular and Cellular Biology, Moscow 121205, Russia
| | - Petr V. Sergiev
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, Moscow 119991, Russia; (E.A.R.); (D.A.I.); (D.A.L.); (P.V.S.)
- Center for Molecular and Cellular Biology, Moscow 121205, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Natalia E. Grammatikova
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow 119021, Russia; (K.S.S.); (G.V.Z.); (N.E.G.); (A.S.T.)
| | - Alexander S. Tikhomirov
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow 119021, Russia; (K.S.S.); (G.V.Z.); (N.E.G.); (A.S.T.)
| | - Andrey E. Shchekotikhin
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow 119021, Russia; (K.S.S.); (G.V.Z.); (N.E.G.); (A.S.T.)
| |
Collapse
|
4
|
Ramirez DM, Dhiman S, Mukherjee A, Wimalasekara R, Schweizer F. Application of tobramycin benzyl ether as an antibiotic adjuvant capable of sensitizing multidrug-resistant Gram-negative bacteria to rifampicin. RSC Med Chem 2024; 15:1055-1065. [PMID: 38516601 PMCID: PMC10953491 DOI: 10.1039/d3md00602f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
The emergence of aminoglycoside resistance has prompted the development of amphiphilic aminoglycoside derivatives which target bacterial membranes. Tobramycin and nebramine ether derivatives initially designed for this purpose were optimized and screened for their potential application as outer membrane (OM) permeabilizing adjuvants. Structure-activity relationship (SAR) studies revealed that the tobramycin benzyl ether was the most optimal OM permeabilizer, capable of potentiating rifampicin, novobiocin, vancomycin, minocycline, and doxycycline against Gram-negative bacteria. The innovative use of this compound as an adjuvant is highlighted by its ability to sensitize multidrug-resistant (MDR) Gram-negative bacteria to rifampicin and restore the susceptibility of MDR Escherichia coli to minocycline.
Collapse
Affiliation(s)
| | - Shiv Dhiman
- Department of Chemistry, University of Manitoba Winnipeg MB R3T 2N2 Canada
| | - Ayan Mukherjee
- Department of Chemistry, University of Manitoba Winnipeg MB R3T 2N2 Canada
| | - Ruwani Wimalasekara
- Department of Microbiology, University of Manitoba Winnipeg MB R3T 2N2 Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba Winnipeg MB R3T 2N2 Canada
| |
Collapse
|
5
|
Sardzikova S, Andrijkova K, Svec P, Beke G, Klucar L, Minarik G, Bielik V, Kolenova A, Soltys K. Gut diversity and the resistome as biomarkers of febrile neutropenia outcome in paediatric oncology patients undergoing hematopoietic stem cell transplantation. Sci Rep 2024; 14:5504. [PMID: 38448687 PMCID: PMC10918076 DOI: 10.1038/s41598-024-56242-8] [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: 11/13/2023] [Accepted: 03/04/2024] [Indexed: 03/08/2024] Open
Abstract
The gut microbiota of paediatric oncology patients undergoing a conditioning regimen before hematopoietic stem cell transplantation is recently considered to play role in febrile neutropenia. Disruption of commensal microbiota and evolution of opportune pathogens community carrying a plethora of antibiotic-resistance genes play crucial role. However, the impact, predictive role and association of patient´s gut resistome in the course of the therapy is still to be elucidated. We analysed gut microbiota composition and resistome of 18 paediatric oncology patients undergoing hematopoietic stem cell transplantation, including 12 patients developing febrile neutropenia, hospitalized at The Bone Marrow Transplantation Unit of the National Institute of Children´s disease in Slovak Republic and healthy individuals (n = 14). Gut microbiome of stool samples obtained in 3 time points, before hematopoietic stem cell transplantation (n = 16), one week after hematopoietic stem cell transplantation (n = 16) and four weeks after hematopoietic stem cell transplantation (n = 14) was investigated using shotgun metagenome sequencing and bioinformatical analysis. We identified significant decrease in alpha-diversity and nine antibiotic-resistance genes msr(C), dfrG, erm(T), VanHAX, erm(B), aac(6)-aph(2), aph(3)-III, ant(6)-Ia and aac(6)-Ii, one week after hematopoietic stem cell transplantation associated with febrile neutropenia. Multidrug-resistant opportune pathogens of ESKAPE, Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae and Escherichia coli found in the gut carried the significant subset of patient's resistome. Over 50% of patients treated with trimethoprim/sulfamethoxazole, piperacillin/tazobactam and amikacin carried antibiotic-resistance genes to applied treatment. The alpha diversity and the resistome of gut microbiota one week after hematopoietic stem cell transplantation is relevant predictor of febrile neutropenia outcome after hematopoietic stem cell transplantation. Furthermore, the interindividual diversity of multi-drug resistant opportunistic pathogens with variable portfolios of antibiotic-resistance genes indicates necessity of preventive, personalized approach.
Collapse
Affiliation(s)
- Sara Sardzikova
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - Kristina Andrijkova
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - Peter Svec
- Department of Paediatric Haematology and Oncology, Children's Haematology and Oncology Clinic and Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Gabor Beke
- Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lubos Klucar
- Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - Viktor Bielik
- Department of Biological and Medical Science, Faculty of Physical Education and Sport, Comenius University in Bratislava, Bratislava, Slovakia
| | - Alexandra Kolenova
- Department of Paediatric Haematology and Oncology, Children's Haematology and Oncology Clinic and Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Katarina Soltys
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia.
| |
Collapse
|
6
|
Mehta D, Saini V, Bajaj A. Recent developments in membrane targeting antifungal agents to mitigate antifungal resistance. RSC Med Chem 2023; 14:1603-1628. [PMID: 37731690 PMCID: PMC10507810 DOI: 10.1039/d3md00151b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/22/2023] [Indexed: 09/22/2023] Open
Abstract
Fungal infections cause severe and life-threatening complications especially in immunocompromised individuals. Antifungals targeting cellular machinery and cell membranes including azoles are used in clinical practice to manage topical to systemic fungal infections. However, continuous exposure to clinically used antifungal agents in managing the fungal infections results in the development of multi-drug resistance via adapting different kinds of intrinsic and extrinsic mechanisms. The unique chemical composition of fungal membranes presents attractive targets for antifungal drug discovery as it is difficult for fungal cells to modify the membrane targets for emergence of drug resistance. Here, we discussed available antifungal drugs with their detailed mechanism of action and described different antifungal resistance mechanisms. We further emphasized structure-activity relationship studies of membrane-targeting antifungal agents, and classified membrane-targeting antifungal agents on the basis of their core scaffold with detailed pharmacological properties. This review aims to pique the interest of potential researchers who could explore this interesting and intricate fungal realm.
Collapse
Affiliation(s)
- Devashish Mehta
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology Faridabad-121001 Haryana India
| | - Varsha Saini
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology Faridabad-121001 Haryana India
| | - Avinash Bajaj
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology Faridabad-121001 Haryana India
| |
Collapse
|
7
|
Dhiman S, Ramirez D, Li Y, Kumar A, Arthur G, Schweizer F. Chimeric Tobramycin-Based Adjuvant TOB-TOB-CIP Potentiates Fluoroquinolone and β-Lactam Antibiotics against Multidrug-Resistant Pseudomonas aeruginosa. ACS Infect Dis 2023; 9:864-885. [PMID: 36917096 DOI: 10.1021/acsinfecdis.2c00549] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
According to the World Health Organization, antibiotic resistance is a global health threat. Of particular importance are infections caused by multidrug-resistant Gram-negative bacteria including Escherichia coli, Acinetobacter baumannii, Klebsiella pneumoniae, and Pseudomonas aeruginosa for which limited treatment options exist. Multiple and simultaneously occurring resistance mechanisms including outer membrane impermeability, overexpression of efflux pumps, antibiotic-modifying enzymes, and modification of genes and antibiotic targets have made antibiotic drug development more difficult against these pathogens. One strategy to cope with these challenges is the use of outer membrane permeabilizers that increase the intracellular concentration of antibiotics when used in combination. In some circumstances, this approach can rescue antibiotics from resistance or repurpose currently marketed antibiotics. Tobramycin-based hybrid antibiotic adjuvants that combine two outer membrane-active components have been previously shown to potentiate antibiotics by facilitating transit through the outer membrane, resulting in increased antibiotic accumulation within the cell. Herein, we extended the concept of tobramycin-based hybrid antibiotic adjuvants to tobramycin-based chimeras by engineering up to three different membrane-active antibiotic warheads such as tobramycin, 1-(1-naphthylmethyl)-piperazine, ciprofloxacin, and cyclam into a central 1,3,5-triazine scaffold. Chimera 4 (TOB-TOB-CIP) consistently synergized with ciprofloxacin, levofloxacin, and moxifloxacin against wild-type and fluoroquinolone-resistant P. aeruginosa. Moreover, the susceptibility breakpoints of ceftazidime, aztreonam, and imipenem were reached using the triple combination of chimera 4 with ceftazidime/avibactam, aztreonam/avibactam, and imipenem/relebactam, respectively, against β-lactamase-harboring P. aeruginosa. Our findings demonstrate that tobramycin-based chimeras form a novel class of antibiotic potentiators capable of restoring the activity of antibiotics against P. aeruginosa.
Collapse
Affiliation(s)
- Shiv Dhiman
- Department of Chemistry, University of Manitoba, Winnipeg R3T 2N2, Manitoba, Canada
| | - Danyel Ramirez
- Department of Chemistry, University of Manitoba, Winnipeg R3T 2N2, Manitoba, Canada
| | - Yanqi Li
- Department of Microbiology, University of Manitoba, Winnipeg R3T 2N2, Manitoba, Canada
| | - Ayush Kumar
- Department of Microbiology, University of Manitoba, Winnipeg R3T 2N2, Manitoba, Canada
| | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg R3E 0J9, Manitoba, Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba, Winnipeg R3T 2N2, Manitoba, Canada
| |
Collapse
|
8
|
Nivedhita S, Shyni Jasmin P, Sarvajith M, Nancharaiah YV. Effects of oxytetracycline on aerobic granular sludge process: Granulation, biological nutrient removal and microbial community structure. CHEMOSPHERE 2022; 307:136103. [PMID: 35995202 DOI: 10.1016/j.chemosphere.2022.136103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/01/2022] [Accepted: 08/15/2022] [Indexed: 06/15/2023]
Abstract
Formation of aerobic granular sludge (AGS), process performance and microbial community structure were investigated in lab-scale sequencing batch reactors (SBR) operated without and with oxytetracycline (OTC). Granulation of activated sludge and appearance of AGS was observed in parallel SBRs operated without and with OTC. However, formation of well-settling aerobic granules was relatively faster in the SBR fed with 100 μg/L OTC and observed within 2 weeks of start-up. Ammonium, total nitrogen, and phosphorus removals were quickly established in the AGS cultivated without OTC. In contrast, nitrogen and phosphorus removals were lower in the OTC fed SBR. But, a gradual improvement in nitrogen and phosphorus removals was observed. After 45 days, nitrogen and phosphorous removals were stabilized at 99% and 70%, respectively, due to establishment of OTC-tolerant community. qPCR revealed the impact of OTC on ammonium oxidizing bacteria, polyphosphate accumulating organisms and their enrichment during exposure to OTC. Ammonium and phosphorus were majorly removed via nitritation-denitritation and enhanced biological phosphorus removal (EBPR) pathways, respectively, in the presence of OTC. Brevundimonas (35%), Thaurea (14%) sp. Ca. Competibacter (5.6%), and Ca. Accumulibacter (4.2%) were enriched in OTC-fed AGS. Of the two OTC-tolerant strains isolated, Micrococcus luteus exhibited growth and efficient OTC biotransformation at different OTC concentrations. Moreover, M. luteus was predominantly growing in the form of aggregates. Key traits such as tolerance, biotransformation and high autoaggregation ability allowed a niche for this strain in the granules. This work has important implications in understanding the effect of antibiotics on AGS and designing AGS based treatment for antibiotic-laden wastewaters.
Collapse
Affiliation(s)
- S Nivedhita
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Chemistry Group, Bhabha Atomic Research Centre, Kalpakkam, 603 102, Tamil Nadu, India
| | - P Shyni Jasmin
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Chemistry Group, Bhabha Atomic Research Centre, Kalpakkam, 603 102, Tamil Nadu, India
| | - M Sarvajith
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Chemistry Group, Bhabha Atomic Research Centre, Kalpakkam, 603 102, Tamil Nadu, India; Homi Bhabha National Institute, BARC Training School Complex, Anushakti Nagar, Trombay, Mumbai, 400 094, India
| | - Y V Nancharaiah
- Biofouling and Biofilm Processes Section, Water and Steam Chemistry Division, Chemistry Group, Bhabha Atomic Research Centre, Kalpakkam, 603 102, Tamil Nadu, India; Homi Bhabha National Institute, BARC Training School Complex, Anushakti Nagar, Trombay, Mumbai, 400 094, India.
| |
Collapse
|
9
|
Takemoto JY, Altenberg GA, Poudyal N, Subedi YP, Chang CWT. Amphiphilic aminoglycosides: Modifications that revive old natural product antibiotics. Front Microbiol 2022; 13:1000199. [PMID: 36212866 PMCID: PMC9537547 DOI: 10.3389/fmicb.2022.1000199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Widely-used Streptomyces-derived antibacterial aminoglycosides have encountered challenges because of antibiotic resistance and toxicity. Today, they are largely relegated to medicinal topical applications. However, chemical modification to amphiphilic aminoglycosides can revive their efficacy against bacterial pathogens and expand their targets to other pathogenic microbes and disorders associated with hyperactive connexin hemichannels. For example, amphiphilic versions of neomycin and neamine are not subject to resistance and have expanded antibacterial spectra, and amphiphilic kanamycins are effective antifungals and have promising therapeutic uses as connexin hemichannel inhibitors. With further research and discoveries aimed at improved formulations and delivery, amphiphilic aminoglycosides may achieve new horizons in pharmacopeia and agriculture for Streptomyces aminoglycosides beyond just serving as topical antibacterials.
Collapse
Affiliation(s)
- Jon Y. Takemoto
- Department of Biology, Utah State University, Logan, UT, United States
| | - Guillermo A. Altenberg
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Naveena Poudyal
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, United States
| | - Yagya P. Subedi
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, United States
| | - Cheng-Wei T. Chang
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, United States
- *Correspondence: Cheng-Wei T. Chang,
| |
Collapse
|
10
|
Wang N, Luo J, Deng F, Huang Y, Zhou H. Antibiotic Combination Therapy: A Strategy to Overcome Bacterial Resistance to Aminoglycoside Antibiotics. Front Pharmacol 2022; 13:839808. [PMID: 35281905 PMCID: PMC8905495 DOI: 10.3389/fphar.2022.839808] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 12/15/2022] Open
Abstract
After the first aminoglycoside antibiotic streptomycin being applied in clinical practice in the mid-1940s, aminoglycoside antibiotics (AGAs) are widely used to treat clinical bacterial infections and bacterial resistance to AGAs is increasing. The bacterial resistance to AGAs is owed to aminoglycoside modifying enzyme modification, active efflux pump gene overexpression and 16S rRNA ribosomal subunit methylation, leading to modification of AGAs' structures and decreased concentration of drugs within bacteria. As AGAs's side effects and bacterial resistance, the development of AGAs is time-consuming and difficult. Because bacterial resistance may occur in a short time after application in clinical practice, it was found that the antibacterial effect of the combination was not only better than that of AGAs alone but also reduce the dosage of antibiotics, thereby reducing the occurrence of side effects. This article reviews the clinical use of AGAs, the antibacterial mechanisms, the molecular mechanisms of bacterial resistance, and especially focuses a recent development of the combination of AGAs with other drugs to exert a synergistic antibacterial effect to provide a new strategy to overcome bacterial resistance to AGAs.
Collapse
Affiliation(s)
| | | | | | | | - Hong Zhou
- Key Laboratory of Basic Pharmacology, Ministry of Education and Joint Laboratory of International Cooperation, Ministry of Education of Characteristic Ethnic Medicine, School of Pharmacy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
11
|
Dangi M, Khichi A, Jakhar R, Chhillar AK. Growing Preferences towards Analog-based Drug Discovery. Curr Pharm Biotechnol 2021; 22:1030-1045. [PMID: 32900347 DOI: 10.2174/1389201021666200908121409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/29/2020] [Accepted: 08/21/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The major concern of today's time is the developing resistance in most of the clinically derived pathogenic micro-organisms for available drugs through several mechanisms. Therefore, there is a dire need to develop novel molecules with drug-like properties that can be effective against the otherwise resistant micro-organisms. METHODS New drugs can be developed using several methods like structure-based drug design, ligandbased drug design, or by developing analogs of the available drugs to further improve their effects. However, the smartness is to opt for the techniques that have comparatively less expenditure, lower failure rates, and faster discovery rates. RESULTS Analog-Based Drug Design (ABDD) is one such technique that researchers worldwide are opting to develop new drug-like molecules with comparatively lower market values. They start by first designing the analogs sharing structural and pharmacological similarities to the existing drugs. This method embarks on scaffold structures of available drugs already approved by the clinical trials, but are left ineffective because of resistance developed by the pathogens. CONCLUSION In this review, we have discussed some recent examples of anti-fungal and anti-bacterial (antimicrobial) drugs that were designed based on the ABDD technique. Also, we have tried to focus on the in silico tools and techniques that can contribute to the designing and computational screening of the analogs, so that these can be further considered for in vitro screening to validate their better biological activities against the pathogens with comparatively reduced rates of failure.
Collapse
Affiliation(s)
- Mehak Dangi
- Centre for Bioinformatics, M.D. University, Rohtak-124001, Haryana, India
| | - Alka Khichi
- Centre for Bioinformatics, M.D. University, Rohtak-124001, Haryana, India
| | - Ritu Jakhar
- Centre for Bioinformatics, M.D. University, Rohtak-124001, Haryana, India
| | - Anil K Chhillar
- Centre for Bioinformatics, M.D. University, Rohtak-124001, Haryana, India
| |
Collapse
|
12
|
Green KD, Punetha A, Chandrika NT, Hou C, Garneau-Tsodikova S, Tsodikov OV. Development of Single-Stranded DNA Bisintercalating Inhibitors of Primase DnaG as Antibiotics. ChemMedChem 2021; 16:1986-1995. [PMID: 33711198 DOI: 10.1002/cmdc.202100001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 03/02/2021] [Indexed: 11/07/2022]
Abstract
Many essential enzymes in bacteria remain promising potential targets of antibacterial agents. In this study, we discovered that dequalinium, a topical antibacterial agent, is an inhibitor of Staphylococcus aureus primase DnaG (SaDnaG) with low-micromolar minimum inhibitory concentrations against several S. aureus strains, including methicillin-resistant bacteria. Mechanistic studies of dequalinium and a series of nine of its synthesized analogues revealed that these compounds are single-stranded DNA bisintercalators that penetrate a bacterium by compromising its membrane. The best compound of this series likely interacts with DnaG directly, inhibits both staphylococcal cell growth and biofilm formation, and displays no significant hemolytic activity or toxicity to mammalian cells. This compound is an excellent lead for further development of a novel anti-staphylococcal therapeutic.
Collapse
Affiliation(s)
- Keith D Green
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536-0596, USA
| | - Ankita Punetha
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536-0596, USA
| | | | - Caixia Hou
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536-0596, USA
| | | | - Oleg V Tsodikov
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536-0596, USA
| |
Collapse
|
13
|
Jaber QZ, Fridman M. Fresh Molecular Concepts to Extend the Lifetimes of Old Antimicrobial Drugs. CHEM REC 2021; 21:631-645. [PMID: 33605532 DOI: 10.1002/tcr.202100014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/04/2021] [Accepted: 02/04/2021] [Indexed: 11/09/2022]
Abstract
Antimicrobial drug development generally initiates with target identification and mode of action studies. Often, emergence of resistance and/or undesired side effects that are discovered only after prolonged clinical use, result in discontinuation of clinical use. Since the cost and time required for improvement of existing drugs are considerably lower than those required for the development of novel drugs, academic and pharmaceutical company researchers pursue this direction. In this account we describe selected examples of how chemical probes generated from antimicrobial drugs and chemical and enzymatic modifications of these drugs have been used to modify modes of action, block mechanisms of resistance, or reduce side effects, improving performance. These examples demonstrate how new and comprehensive mechanistic insights can be translated into fresh concepts for development of next-generation antimicrobial agents.
Collapse
Affiliation(s)
- Qais Z Jaber
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Micha Fridman
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| |
Collapse
|
14
|
Logviniuk D, Fridman M. Serum Prevents Interactions between Antimicrobial Amphiphilic Aminoglycosides and Plasma Membranes. ACS Infect Dis 2020; 6:3212-3223. [PMID: 33174428 DOI: 10.1021/acsinfecdis.0c00588] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Antimicrobial cationic amphiphiles have broad-spectrum activity, and microbes do not readily develop resistance to these agents, highlighting their clinical and industrial potential. Cationic amphiphiles perturb the integrity of membranes leading to cell death, and the lack of discrimination between microbial and mammalian plasma membranes is thought to be one of the main barriers of using these agents for the treatment of systemic infections. Here, we describe the synthesis and study of 20 antimicrobial cationic amphiphiles that are derivatives of the aminoglycoside nebramine with different numbers of alkyl chain ethers that differ in length and degree of unsaturation. We determined antifungal activities and evaluated hemoglobin release from red blood cells as a measure of membrane selectivity and analyzed how serum influences these activities. Microscopic images revealed morphological transformations of red blood cells from the normal double-disc shape to empty ghost cells upon treatment with the cationic amphiphiles. Antifungal activity, hemolysis, and morphological changes in red blood cells decreased as the percentage of serum in the culture medium was increased. In images of red blood cells treated with fluorescently labeled amphiphilic nebramine probes, the accumulation of the cationic amphiphiles in the membranes decreased as serum concentration increased. This suggests that, in addition to its known effect of preventing the deformability of red blood cells, serum prevents interactions between cationic amphiphiles and the plasma membrane. The results of this study indicate that biological activities of cationic amphiphiles are abrogated in serum. Thus, these agents are suitable for external and industrial uses but probably not for effective treatment of systemic infections.
Collapse
Affiliation(s)
- Dana Logviniuk
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Micha Fridman
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
15
|
Dezanet C, Kempf J, Mingeot-Leclercq MP, Décout JL. Amphiphilic Aminoglycosides as Medicinal Agents. Int J Mol Sci 2020; 21:E7411. [PMID: 33049963 PMCID: PMC7583001 DOI: 10.3390/ijms21197411] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 09/27/2020] [Accepted: 10/02/2020] [Indexed: 12/25/2022] Open
Abstract
The conjugation of hydrophobic group(s) to the polycationic hydrophilic core of the antibiotic drugs aminoglycosides (AGs), targeting ribosomal RNA, has led to the development of amphiphilic aminoglycosides (AAGs). These drugs exhibit numerous biological effects, including good antibacterial effects against susceptible and multidrug-resistant bacteria due to the targeting of bacterial membranes. In the first part of this review, we summarize our work in identifying and developing broad-spectrum antibacterial AAGs that constitute a new class of antibiotic agents acting on bacterial membranes. The target-shift strongly improves antibiotic activity against bacterial strains that are resistant to the parent AG drugs and to antibiotic drugs of other classes, and renders the emergence of resistant Pseudomonas aeruginosa strains highly difficult. Structure-activity and structure-eukaryotic cytotoxicity relationships, specificity and barriers that need to be crossed in their development as antibacterial agents are delineated, with a focus on their targets in membranes, lipopolysaccharides (LPS) and cardiolipin (CL), and the corresponding mode of action against Gram-negative bacteria. At the end of the first part, we summarize the other recent advances in the field of antibacterial AAGs, mainly published since 2016, with an emphasis on the emerging AAGs which are made of an AG core conjugated to an adjuvant or an antibiotic drug of another class (antibiotic hybrids). In the second part, we briefly illustrate other biological and biochemical effects of AAGs, i.e., their antifungal activity, their use as delivery vehicles of nucleic acids, of short peptide (polyamide) nucleic acids (PNAs) and of drugs, as well as their ability to cleave DNA at abasic sites and to inhibit the functioning of connexin hemichannels. Finally, we discuss some aspects of structure-activity relationships in order to explain and improve the target selectivity of AAGs.
Collapse
Affiliation(s)
- Clément Dezanet
- Molecular Pharmacochemistry Department, University Grenoble Alpes, CNRS, 470 Rue de la Chimie, F-38000 Grenoble, France; (C.D.); (J.K.)
| | - Julie Kempf
- Molecular Pharmacochemistry Department, University Grenoble Alpes, CNRS, 470 Rue de la Chimie, F-38000 Grenoble, France; (C.D.); (J.K.)
| | - Marie-Paule Mingeot-Leclercq
- Cellular and Molecular Pharmacology Unit, Louvain Drug Research Institute, Catholic University of Louvain, Avenue E. Mounier 73, UCL B1.73.05, 1200 Brussels, Belgium
| | - Jean-Luc Décout
- Molecular Pharmacochemistry Department, University Grenoble Alpes, CNRS, 470 Rue de la Chimie, F-38000 Grenoble, France; (C.D.); (J.K.)
| |
Collapse
|
16
|
Idowu T, Ammeter D, Arthur G, Zhanel GG, Schweizer F. Potentiation of β-lactam antibiotics and β-lactam/β-lactamase inhibitor combinations against MDR and XDR Pseudomonas aeruginosa using non-ribosomal tobramycin-cyclam conjugates. J Antimicrob Chemother 2020; 74:2640-2648. [PMID: 31139830 DOI: 10.1093/jac/dkz228] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES To develop a multifunctional adjuvant molecule that can rescue β-lactam antibiotics and β-lactam/β-lactamase inhibitor combinations from resistance in carbapenem-resistant Pseudomonas aeruginosa clinical isolates. METHODS Preparation of adjuvant was guided by structure-activity relationships, following standard protocols. Susceptibility and chequerboard studies were assessed using serial 2-fold dilution assays. Toxicity was evaluated against porcine erythrocytes, human embryonic kidney (HEK293) cells and liver carcinoma (HepG2) cells via MTS assay. Preliminary in vivo efficacy was evaluated using a Galleria mellonella infection model. RESULTS Conjugation of tobramycin and cyclam abrogates the ribosomal effects of tobramycin but confers a potent adjuvant property that restores full antibiotic activity of meropenem and aztreonam against carbapenem-resistant P. aeruginosa. Therapeutic levels of susceptibility, as determined by CLSI susceptibility breakpoints, were attained in several MDR clinical isolates, and time-kill assays revealed a synergistic dose-dependent pharmacodynamic relationship. A triple combination of the adjuvant with ceftazidime/avibactam (approved), aztreonam/avibactam (Phase III) and meropenem/avibactam enhances the efficacies of β-lactam/β-lactamase inhibitors against recalcitrant strains, suggesting rapid access of the combination to their periplasmic targets. The newly developed adjuvants, and their combinations, were non-haemolytic and non-cytotoxic, and preliminary in vivo evaluation in G. mellonella suggests therapeutic potential for the double and triple combinations. CONCLUSIONS Non-ribosomal tobramycin-cyclam conjugate mitigates the effect of OprD/OprF porin loss in P. aeruginosa and potentiates β-lactam/β-lactamase inhibitors against carbapenem-resistant clinical isolates, highlighting the complexity of resistance to β-lactam antibiotics. Our strategy presents an avenue to further preserve the therapeutic utility of β-lactam antibiotics.
Collapse
Affiliation(s)
- Temilolu Idowu
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Derek Ammeter
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - George G Zhanel
- Department of Medical Microbiology/Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Medical Microbiology/Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
17
|
Ying L, Zhu H, Fosso MY, Garneau-Tsodikova S, Fredrick K. Modified Aminoglycosides Bind Nucleic Acids in High-Molecular-Weight Complexes. Antibiotics (Basel) 2020; 9:antibiotics9020093. [PMID: 32098020 PMCID: PMC7168264 DOI: 10.3390/antibiotics9020093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/05/2022] Open
Abstract
Aminoglycosides represent a large group of antibiotics well known for their ability to target the bacterial ribosome. In studying 6”-substituted variants of the aminoglycoside tobramycin, we serendipitously found that compounds with C12 or C14 linear alkyl substituents potently inhibit reverse transcription in vitro. Initial observations suggested specific inhibition of reverse transcriptase. However, further analysis showed that these and related compounds bind nucleic acids with high affinity, forming high-molecular weight complexes. Stable complex formation is observed with DNA or RNA in single- or double-stranded form. Given the amphiphilic nature of these aminoglycoside derivatives, they likely form micelles and/or vesicles with surface-bound nucleic acids. Hence, these compounds may be useful tools to localize nucleic acids to surfaces or deliver nucleic acids to cells or organelles.
Collapse
Affiliation(s)
- Lanqing Ying
- Department of Microbiology and Center for RNA Biology, The Ohio State University, Columbus, OH 43210-1292, USA; (L.Y.); (H.Z.)
| | - Hongkun Zhu
- Department of Microbiology and Center for RNA Biology, The Ohio State University, Columbus, OH 43210-1292, USA; (L.Y.); (H.Z.)
| | - Marina Y. Fosso
- Department of Pharmaceutical Sciences in the College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA; (M.Y.F.); (S.G.-T.)
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences in the College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA; (M.Y.F.); (S.G.-T.)
| | - Kurt Fredrick
- Department of Microbiology and Center for RNA Biology, The Ohio State University, Columbus, OH 43210-1292, USA; (L.Y.); (H.Z.)
- Correspondence: ; Tel.: +1-614-292-6679
| |
Collapse
|
18
|
Zada SL, Baruch BB, Simhaev L, Engel H, Fridman M. Chemical Modifications Reduce Auditory Cell Damage Induced by Aminoglycoside Antibiotics. J Am Chem Soc 2020; 142:3077-3087. [PMID: 31958945 DOI: 10.1021/jacs.9b12420] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although aminoglycoside antibiotics are effective against Gram-negative infections, these drugs often cause irreversible hearing damage. Binding to the decoding site of the eukaryotic ribosomes appears to result in ototoxicity, but there is evidence that other effects are involved. Here, we show how chemical modifications of apramycin and geneticin, considered among the least and most toxic aminoglycosides, respectively, reduce auditory cell damage. Using molecular dynamics simulations, we studied how modified aminoglycosides influence the essential freedom of movement of the decoding site of the ribosome, the region targeted by aminoglycosides. By determining the ratio of a protein translated in mitochondria to that of a protein translated in the cytoplasm, we showed that aminoglycosides can paradoxically elevate rather than reduce protein levels. We showed that certain aminoglycosides induce rapid plasma membrane permeabilization and that this nonribosomal effect can also be reduced through chemical modifications. The results presented suggest a new paradigm for the development of safer aminoglycoside antibiotics.
Collapse
Affiliation(s)
- Sivan Louzoun Zada
- School of Chemistry, Raymond and Beverley Sackler Faculty of Exact Sciences , Tel Aviv University , Tel Aviv , Israel , 6997801
| | - Bar Ben Baruch
- School of Chemistry, Raymond and Beverley Sackler Faculty of Exact Sciences , Tel Aviv University , Tel Aviv , Israel , 6997801
| | - Luba Simhaev
- Blavatnik Center for Drug Discovery , Tel Aviv University , Tel Aviv , 6997801 , Israel
| | - Hamutal Engel
- Blavatnik Center for Drug Discovery , Tel Aviv University , Tel Aviv , 6997801 , Israel
| | - Micha Fridman
- School of Chemistry, Raymond and Beverley Sackler Faculty of Exact Sciences , Tel Aviv University , Tel Aviv , Israel , 6997801
| |
Collapse
|
19
|
Idowu T, Ammeter D, Rossong H, Zhanel GG, Schweizer F. Homodimeric Tobramycin Adjuvant Repurposes Novobiocin as an Effective Antibacterial Agent against Gram-Negative Bacteria. J Med Chem 2019; 62:9103-9115. [PMID: 31557020 DOI: 10.1021/acs.jmedchem.9b00876] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Low permeability across the outer membrane is a major reason why most antibiotics are ineffective against Gram-negative bacteria. Agents that permeabilize the outer membrane are typically toxic at their effective concentrations. Here, we report the development of a broad-spectrum homodimeric tobramycin adjuvant that is nontoxic and more potent than the gold standard permeabilizing agent, polymyxin B nonapeptide. In pilot studies, the adjuvant confers potent bactericidal activity on novobiocin against Gram-negative bacteria, including carbapenem-resistant and colistin-resistant strains bearing plasmid-borne mcr-1 genes. Resistance development to the combination was significantly reduced, relative to novobiocin alone, and there was no induction of cross-resistance to other antibiotics, including the gyrase-acting fluoroquinolones. Tobramycin homodimer may allow the use of lower doses of novobiocin, overcoming its twin problem of efficacy and toxicity.
Collapse
Affiliation(s)
- Temilolu Idowu
- Department of Chemistry , University of Manitoba , Winnipeg , MB R3T 2N2 , Canada
| | - Derek Ammeter
- Department of Chemistry , University of Manitoba , Winnipeg , MB R3T 2N2 , Canada
| | - Heather Rossong
- Department of Chemistry , University of Manitoba , Winnipeg , MB R3T 2N2 , Canada
| | - George G Zhanel
- Department of Medical Microbiology/Infectious Diseases , University of Manitoba , Winnipeg , MB R3T 1R9 , Canada
| | - Frank Schweizer
- Department of Chemistry , University of Manitoba , Winnipeg , MB R3T 2N2 , Canada.,Department of Medical Microbiology/Infectious Diseases , University of Manitoba , Winnipeg , MB R3T 1R9 , Canada
| |
Collapse
|
20
|
Subedi YP, Pandey U, Alfindee MN, Montgomery H, Roberts P, Wight J, Nichols G, Grilley M, Takemoto JY, Chang CWT. Scalable and cost-effective tosylation-mediated synthesis of antifungal and fungal diagnostic 6″-Modified amphiphilic kanamycins. Eur J Med Chem 2019; 182:111639. [PMID: 31470306 DOI: 10.1016/j.ejmech.2019.111639] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022]
Abstract
Amphiphilic kanamycins bearing hydrophobic modifications at the 6″ position have attracted interest due to remarkable antibacterial-to-antifungal switches in bioactivity. In this report, we investigate a hurdle that hinders practical applications of these amphiphilic kanamycins: a cost-effective synthesis that allows the incorporation of various connecting functionalities to which the hydrophobic moieties are connected to the kanamycin core. A cost-effective tosylation enables various modifications at the 6″ position, which is scalable to a 90-g scale. The connecting functionalities, such as amine and thiol, were not the dominant factor for biological activity. Instead, the linear chain length played the decisive role. Amphiphilic kanamycin attached with tetradecyl (C14) or hexadecyl (C16) showed strong antifungal and modest antibacterial activities than with shorter chains (C6-C10). However, increases in chain length were closely correlated with an increase in HeLa cell toxicity. Thus, a compromise between the antimicrobial activities and cytotoxicities, for optimal efficacy of amphiphilic kanamycins may contain chain lengths between C8 and C12. Finally, the described synthetic protocol also allows the preparation of a fluorescent amphiphilic kanamycin selective toward fungi.
Collapse
Affiliation(s)
- Yagya Prasad Subedi
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT, 84322-0300, USA
| | - Uddav Pandey
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT, 84322-0300, USA
| | - Madher N Alfindee
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT, 84322-0300, USA
| | - Heath Montgomery
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT, 84322-0300, USA
| | - Paul Roberts
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT, 84322-0300, USA
| | - Jeffrey Wight
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT, 84322-0300, USA
| | - Gavin Nichols
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT, 84322-0300, USA
| | - Michell Grilley
- Department of Biology, Utah State University, 5305 Old Main Hill, Logan, UT, 84322-5305, USA
| | - Jon Y Takemoto
- Department of Biology, Utah State University, 5305 Old Main Hill, Logan, UT, 84322-5305, USA
| | - Cheng-Wei Tom Chang
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT, 84322-0300, USA.
| |
Collapse
|
21
|
Louzoun‐Zada S, Jaber QZ, Fridman M. Guiding Drugs to Target‐Harboring Organelles: Stretching Drug‐Delivery to a Higher Level of Resolution. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201906284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Sivan Louzoun‐Zada
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences Tel Aviv University Tel Aviv 6997801 Israel
| | - Qais Z. Jaber
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences Tel Aviv University Tel Aviv 6997801 Israel
| | - Micha Fridman
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences Tel Aviv University Tel Aviv 6997801 Israel
| |
Collapse
|
22
|
Louzoun-Zada S, Jaber QZ, Fridman M. Guiding Drugs to Target-Harboring Organelles: Stretching Drug-Delivery to a Higher Level of Resolution. Angew Chem Int Ed Engl 2019; 58:15584-15594. [PMID: 31237741 DOI: 10.1002/anie.201906284] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Indexed: 01/04/2023]
Abstract
The ratio between the dose of drug required for optimal efficacy and the dose that causes toxicity is referred to as the therapeutic window. This ratio can be increased by directing the drug to the diseased tissue or pathogenic cell. For drugs targeting fungi and malignant cells, the therapeutic window can be further improved by increasing the resolution of drug delivery to the specific organelle that harbors the drug's target. Organelle targeting is challenging and is, therefore, an under-exploited strategy. Here we provide an overview of recent advances in control of the subcellular distribution of small molecules with the focus on chemical modifications. Highlighted are recent examples of active and passive organelle-specific targeting by incorporation of organelle-directing molecular determinants or by chemical modifications of the pharmacophore. The outstanding potential that lies in the development of organelle-specific drugs is becoming increasingly apparent.
Collapse
Affiliation(s)
- Sivan Louzoun-Zada
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Qais Z Jaber
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Micha Fridman
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| |
Collapse
|
23
|
Idowu T, Arthur G, Zhanel GG, Schweizer F. Heterodimeric Rifampicin-Tobramycin conjugates break intrinsic resistance of Pseudomonas aeruginosa to doxycycline and chloramphenicol in vitro and in a Galleria mellonella in vivo model. Eur J Med Chem 2019; 174:16-32. [PMID: 31022550 DOI: 10.1016/j.ejmech.2019.04.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/01/2019] [Accepted: 04/13/2019] [Indexed: 11/25/2022]
Abstract
Intrinsic resistance in Pseudomonas aeruginosa, defined by chromosomally encoded low outer membrane permeability and constitutively over-expressed efflux pumps, is a major reason why the pathogen is refractory to many antibiotics. Herein, we report that heterodimeric rifampicin-tobramycin conjugates break this intrinsic resistance and sensitize multidrug and extensively drug-resistant P. aeruginosa to doxycycline and chloramphenicol in vitro and in vivo. Tetracyclines and chloramphenicol are model compounds for bacteriostatic effects, but when combined with rifampicin-tobramycin adjuvants, their effects became bactericidal at sub MIC levels. Potentiation of tetracyclines correlates with the SAR of this class of drugs and is consistent with outer membrane permeabilization and efflux pump inhibition. Overall, this strategy finds new uses for old drugs and presents an avenue to expand the therapeutic utility of legacy antibiotics to recalcitrant pathogens such as P. aeruginosa.
Collapse
Affiliation(s)
- Temilolu Idowu
- Department of Chemistry, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada
| | - George G Zhanel
- Department of Medical Microbiology/Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 1R9, Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada; Department of Medical Microbiology/Infectious Diseases, University of Manitoba, Winnipeg, MB, R3T 1R9, Canada.
| |
Collapse
|
24
|
Subedi YP, Roberts P, Grilley M, Takemoto JY, Chang CWT. Development of Fungal Selective Amphiphilic Kanamycin: Cost-Effective Synthesis and Use of Fluorescent Analogs for Mode of Action Investigation. ACS Infect Dis 2019; 5:473-483. [PMID: 30674192 DOI: 10.1021/acsinfecdis.8b00327] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Amphiphilic aminoglycosides have attracted interest due to their novel antifungal activities. A crucial but often neglected factor for drug development in academia is cost of production. Herein is reported a one-step, inexpensive synthesis of amphiphilic alkyl kanamycins constituted with only natural components. The synthetic methodology also enabled the preparation of a series fluorescent amphiphilic aryl kanamycins for direct structure-activity mode of action studies. The lead compounds showed prominent antifungal activities against a panel of fungi, including Fusarium graminearum, Cryptococcus neoformans, and several Candida sp., and also significant antibacterial activities. With fluorescence-based whole cell assays, the aryl amphiphilic kanamycins were observed to permeabilize fungal surface membranes at faster rates than bacterial surface membranes. Also, the antifungal action of the amphiphilic kanamycins was observed to occur in a biphasic mode with an initial fast phase correlated with rapid membrane permeabilization at subminimal inhibitory concentrations and a slower phase membrane permeabilization that elevates the reactive oxygen species production leading to cell death. Inactive hydrophobic amphiphilic kanamycins displayed no membrane permeabilization. The results offer cost-effective methods for producing amphiphilic kanamycins and reveal insights into how nonfungal specific amphiphilic kanamycins can be employed for fungal specific diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Yagya Prasad Subedi
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, Utah 84322-0300, United States
| | - Paul Roberts
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, Utah 84322-0300, United States
| | - Michelle Grilley
- Department of Biology, Utah State University, 5305 Old Main Hill, Logan, Utah 84322-5305, United States
| | - Jon Y. Takemoto
- Department of Biology, Utah State University, 5305 Old Main Hill, Logan, Utah 84322-5305, United States
| | - Cheng-Wei Tom Chang
- Department of Chemistry and Biochemistry, Utah State University, 0300 Old Main Hill, Logan, Utah 84322-0300, United States
| |
Collapse
|
25
|
Synthesis, antimicrobial activity, attenuation of aminoglycoside resistance in MRSA, and ribosomal A-site binding of pyrene-neomycin conjugates. Eur J Med Chem 2018; 163:381-393. [PMID: 30530174 DOI: 10.1016/j.ejmech.2018.11.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 01/27/2023]
Abstract
The development of new ligands that have comparable or enhanced therapeutic efficacy relative to current drugs is vital to the health of the global community in the short and long term. One strategy to accomplish this goal is to functionalize sites on current antimicrobials to enhance specificity and affinity while abating resistance mechanisms of infectious organisms. Herein, we report the synthesis of a series of pyrene-neomycin B (PYR-NEO) conjugates, their binding affinity to A-site RNA targets, resistance to aminoglycoside-modifying enzymes (AMEs), and antibacterial activity against a wide variety of bacterial strains of clinical relevance. PYR-NEO conjugation significantly alters the affinities of NEO for bacterial A-site targets. The conjugation of PYR to NEO significantly increased the resistance of NEO to AME modification. PYR-NEO conjugates exhibited broad-spectrum activity towards Gram-positive bacteria, including improved activity against NEO-resistant methicillin-resistant Staphylococcus aureus (MRSA) strains.
Collapse
|
26
|
Jaber QZ, Benhamou RI, Herzog IM, Ben Baruch B, Fridman M. Cationic Amphiphiles Induce Macromolecule Denaturation and Organelle Decomposition in Pathogenic Yeast. Angew Chem Int Ed Engl 2018; 57:16391-16395. [PMID: 30307679 DOI: 10.1002/anie.201809410] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/27/2018] [Indexed: 11/12/2022]
Abstract
Cationic amphiphiles are a large and diverse class of antimicrobial agents. Although their mode of action is not fully resolved, it is generally accepted that these antimicrobials perturb the structural integrity of the plasma membrane leading to the microbial cell disruption. Here we report on the development of inherently fluorescent antifungal cationic amphiphiles and on the study of their effects on cells of Candida, one of the most common fungal pathogens in humans. Fluorescent images of Candida yeast cells that express a fluorescent reporter protein revealed that the cationic amphiphiles rapidly accumulated in the cytosol and led to structural changes in proteins and DNA. Using fluorescent organelle-specific dyes, we showed that these antifungal agents also caused organelle disassembly in Candida cells. The results of this study indicate that, in designing antifungal cationic amphiphiles for clinical use, the intracellular activities of these molecules must be addressed to avoid undesired side effects to mammalian cells.
Collapse
Affiliation(s)
- Qais Z Jaber
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Raphael I Benhamou
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Ido M Herzog
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Bar Ben Baruch
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel
| | - Micha Fridman
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| |
Collapse
|
27
|
Jaber QZ, Benhamou RI, Herzog IM, Ben Baruch B, Fridman M. Cationic Amphiphiles Induce Macromolecule Denaturation and Organelle Decomposition in Pathogenic Yeast. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201809410] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Qais Z. Jaber
- School of Chemistry; Raymond and Beverly Sackler Faculty of Exact Sciences; Tel Aviv University; Tel Aviv 6997801 Israel
| | - Raphael I. Benhamou
- School of Chemistry; Raymond and Beverly Sackler Faculty of Exact Sciences; Tel Aviv University; Tel Aviv 6997801 Israel
| | - Ido M. Herzog
- School of Chemistry; Raymond and Beverly Sackler Faculty of Exact Sciences; Tel Aviv University; Tel Aviv 6997801 Israel
| | - Bar Ben Baruch
- Department of Neurobiology; George S. Wise Faculty of Life Sciences; Tel Aviv University; Ramat Aviv Tel Aviv 6997801 Israel
| | - Micha Fridman
- School of Chemistry; Raymond and Beverly Sackler Faculty of Exact Sciences; Tel Aviv University; Tel Aviv 6997801 Israel
| |
Collapse
|
28
|
Kukielski C, Maiti K, Bhaduri S, Story S, Arya DP. Rapid solid-phase syntheses of a peptidic-aminoglycoside library. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.07.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
29
|
Ezelarab HAA, Abbas SH, Hassan HA, Abuo-Rahma GEDA. Recent updates of fluoroquinolones as antibacterial agents. Arch Pharm (Weinheim) 2018; 351:e1800141. [DOI: 10.1002/ardp.201800141] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Hend A. A. Ezelarab
- Faculty of Pharmacy, Department of Medicinal Chemistry; Minia University; Minia Egypt
| | - Samar H. Abbas
- Faculty of Pharmacy, Department of Medicinal Chemistry; Minia University; Minia Egypt
| | - Heba A. Hassan
- Faculty of Pharmacy, Department of Medicinal Chemistry; Minia University; Minia Egypt
| | | |
Collapse
|
30
|
Louzoun Zada S, Green KD, Shrestha SK, Herzog IM, Garneau-Tsodikova S, Fridman M. Derivatives of Ribosome-Inhibiting Antibiotic Chloramphenicol Inhibit the Biosynthesis of Bacterial Cell Wall. ACS Infect Dis 2018; 4:1121-1129. [PMID: 29714997 DOI: 10.1021/acsinfecdis.8b00078] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Here, we describe the preparation and evaluation of α,β-unsaturated carbonyl derivatives of the bacterial translation inhibiting antibiotic chloramphenicol (CAM). Compared to the parent antibiotic, two compounds containing α,β-unsaturated ketones (1 and 4) displayed a broader spectrum of activity against a panel of Gram-positive pathogens with a minimum inhibitory concentration range of 2-32 μg/mL. Interestingly, unlike the parent CAM, these compounds do not inhibit bacterial translation. Microscopic evidence and metabolic labeling of a cell wall peptidoglycan suggested that compounds 1 and 4 caused extensive damage to the envelope of Staphylococcus aureus cells by inhibition of the early stage of cell wall peptidoglycan biosynthesis. Unlike the effect of membrane-disrupting antimicrobial cationic amphiphiles, these compounds did not rapidly permeabilize the bacterial membrane. Like the parent antibiotic CAM, compounds 1 and 4 had a bacteriostatic effect on S. aureus. Both compounds 1 and 4 were cytotoxic to immortalized nucleated mammalian cells; however, neither caused measurable membrane damage to mammalian red blood cells. These data suggest that the reported CAM-derived antimicrobial agents offer a new molecular scaffold for development of novel bacterial cell wall biosynthesis inhibiting antibiotics.
Collapse
Affiliation(s)
- Sivan Louzoun Zada
- Raymond and Beverly Sackler Faculty of Exact Sciences, School of Chemistry, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Keith D. Green
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536-0596, United States
| | - Sanjib K. Shrestha
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536-0596, United States
| | - Ido M. Herzog
- Raymond and Beverly Sackler Faculty of Exact Sciences, School of Chemistry, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536-0596, United States
| | - Micha Fridman
- Raymond and Beverly Sackler Faculty of Exact Sciences, School of Chemistry, Tel Aviv University, Tel Aviv, 6997801, Israel
| |
Collapse
|
31
|
Takemoto JY, Wegulo SN, Yuen GY, Stevens JA, Jochum CC, Chang CWT, Kawasaki Y, Miller GW. Suppression of wheat Fusarium head blight by novel amphiphilic aminoglycoside fungicide K20. Fungal Biol 2018; 122:465-470. [DOI: 10.1016/j.funbio.2017.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 11/25/2022]
|
32
|
Steinbuch KB, Benhamou RI, Levin L, Stein R, Fridman M. Increased Degree of Unsaturation in the Lipid of Antifungal Cationic Amphiphiles Facilitates Selective Fungal Cell Disruption. ACS Infect Dis 2018; 4:825-836. [PMID: 29419285 DOI: 10.1021/acsinfecdis.7b00272] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Antimicrobial cationic amphiphiles derived from aminoglycosides act through cell membrane permeabilization but have limited selectivity for microbial cell membranes. Herein, we report that an increased degree of unsaturation in the fatty acid segment of antifungal cationic amphiphiles derived from the aminoglycoside tobramycin significantly reduced toxicity to mammalian cells. A collection of tobramycin-derived cationic amphiphiles substituted with C18 lipid chains varying in degree of unsaturation and double bond configuration were synthesized. All had potent activity against a panel of important fungal pathogens including strains with resistance to a variety of antifungal drugs. The tobramycin-derived cationic amphiphile substituted with linolenic acid with three cis double bonds (compound 6) was up to an order of magnitude less toxic to mammalian cells than cationic amphiphiles composed of lipids with a lower degree of unsaturation and than the fungal membrane disrupting drug amphotericin B. Compound 6 was 12-fold more selective (red blood cell hemolysis relative to antifungal activity) than compound 1, the derivative with a fully saturated lipid chain. Notably, compound 6 disrupted the membranes of fungal cells without affecting the viability of cocultured mammalian cells. This study demonstrates that the degree of unsaturation and the configuration of the double bond in lipids of cationic amphiphiles are important parameters that, if optimized, result in compounds with broad spectrum and potent antifungal activity as well as reduced toxicity toward mammalian cells.
Collapse
Affiliation(s)
- Kfir B. Steinbuch
- School of Chemistry, Raymond and Beverley Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel 6997801
| | - Raphael I. Benhamou
- School of Chemistry, Raymond and Beverley Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel 6997801
| | - Lotan Levin
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel 6997801
| | - Reuven Stein
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel 6997801
| | - Micha Fridman
- School of Chemistry, Raymond and Beverley Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel 6997801
| |
Collapse
|
33
|
Fosso MY, Shrestha SK, Thamban Chandrika N, Dennis EK, Green KD, Garneau-Tsodikova S. Differential Effects of Linkers on the Activity of Amphiphilic Tobramycin Antifungals. Molecules 2018; 23:molecules23040899. [PMID: 29652845 PMCID: PMC5971061 DOI: 10.3390/molecules23040899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 01/08/2023] Open
Abstract
As the threat associated with fungal infections continues to rise and the availability of antifungal drugs remains a concern, it becomes obvious that the need to bolster the antifungal armamentarium is urgent. Building from our previous findings of tobramycin (TOB) derivatives with antifungal activity, we further investigate the effects of various linkers on the biological activity of these aminoglycosides. Herein, we analyze how thioether, sulfone, triazole, amide, and ether functionalities affect the antifungal activity of alkylated TOB derivatives against 22 Candida, Cryptococcus, and Aspergillus species. We also evaluate their impact on the hemolysis of murine erythrocytes and the cytotoxicity against mammalian cell lines. While the triazole linker appears to confer optimal activity overall, all of the linkers incorporated into the TOB derivatives resulted in compounds that are very effective against the Cryptococcus neoformans species, with MIC values ranging from 0.48 to 3.9 μg/mL.
Collapse
Affiliation(s)
- Marina Y Fosso
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA.
| | - Sanjib K Shrestha
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA.
| | - Nishad Thamban Chandrika
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA.
| | - Emily K Dennis
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA.
| | - Keith D Green
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA.
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA.
| |
Collapse
|
34
|
Niu Y, Wang M, Cao Y, Nimmagadda A, Hu J, Wu Y, Cai J, Ye XS. Rational Design of Dimeric Lysine N-Alkylamides as Potent and Broad-Spectrum Antibacterial Agents. J Med Chem 2018; 61:2865-2874. [PMID: 29569910 DOI: 10.1021/acs.jmedchem.7b01704] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibiotic resistance is one of the biggest threats to public health, and new antibacterial agents hence are in an urgent need to combat infectious diseases caused by multidrug-resistant (MDR) pathogens. Utilizing dimerization strategy, we rationally designed and efficiently synthesized a new series of small molecule dimeric lysine alkylamides as mimics of AMPs. Evaluation of these mimics against a panel of Gram-positive and Gram-negative bacteria including MDR strains was performed, and a broad-spectrum and potent compound 3d was identified. This compound displayed high specificity toward bacteria over mammalian cell. Time-kill kinetics and mechanistic studies suggest that compound 3d quickly eliminated bacteria in a bactericidal mode by disrupting bacterial cell membrane. In addition, lead compound 3d could inhibit biofilm formation and did not develop drug resistance in S. aureus and E. coli over 14 passages. These results suggested that dimeric lysine nonylamide has immense potential as a new type of novel small molecular agent to combat antibiotic resistance.
Collapse
Affiliation(s)
- Youhong Niu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xue Yuan Road No.38 , Beijing 100191 , China
| | - Minghui Wang
- Department of Chemistry , University of South Florida , 4202 E. Fowler Avenue , Tampa , Florida 33620 , United States
| | - Yafei Cao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xue Yuan Road No.38 , Beijing 100191 , China
| | - Alekhya Nimmagadda
- Department of Chemistry , University of South Florida , 4202 E. Fowler Avenue , Tampa , Florida 33620 , United States
| | - Jianxing Hu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xue Yuan Road No.38 , Beijing 100191 , China
| | - Yanfen Wu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xue Yuan Road No.38 , Beijing 100191 , China
| | - Jianfeng Cai
- Department of Chemistry , University of South Florida , 4202 E. Fowler Avenue , Tampa , Florida 33620 , United States
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xue Yuan Road No.38 , Beijing 100191 , China
| |
Collapse
|
35
|
Basak A, Abouelhassan Y, Zuo R, Yousaf H, Ding Y, Huigens RW. Antimicrobial peptide-inspired NH125 analogues: bacterial and fungal biofilm-eradicating agents and rapid killers of MRSA persisters. Org Biomol Chem 2018; 15:5503-5512. [PMID: 28534905 DOI: 10.1039/c7ob01028a] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During microbial infection, antimicrobial peptides are utilized by the immune response to rapidly eradicate microbial pathogens through the destruction of cellular membranes. Inspired by antimicrobial peptides, quaternary ammonium cationic (QAC) compounds have emerged as agents capable of destroying bacterial membranes leading to rapid bacterial death, including the eradication of persistent, surface-attached bacterial biofilms. NH125, an imidazolium cation with a sixteen membered fatty tail, was recently reported to eradicate persister cells and was our starting point for the development of novel antimicrobial agents. Here, we describe the design, chemical synthesis and biological investigations of a collection of 30 diverse NH125 analogues which provided critical insights into structural features that are important for antimicrobial activities in this class. From these studies, multiple NH125 analogues were identified to possess potent antibacterial and antifungal activities, eradicate both bacterial and fungal biofilms and rapidly eradicate MRSA persister cells in stationary phase. NH125 analogues also demonstrated more rapid persister cell killing activities against MRSA when tested alongside a panel of diverse membrane-active agents, including BAC-16 and daptomycin. NH125 analogues could have a significant impact on persister- and biofilm-related problems in numerous biomedical applications.
Collapse
Affiliation(s)
- Akash Basak
- Department of Chemistry, University of Florida, Gainesville, FL 32610, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Antibiotic Hybrids: the Next Generation of Agents and Adjuvants against Gram-Negative Pathogens? Clin Microbiol Rev 2018. [PMID: 29540434 DOI: 10.1128/cmr.00077-17] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The global incidence of drug-resistant Gram-negative bacillary infections has been increasing, and there is a dire need to develop novel strategies to overcome this problem. Intrinsic resistance in Gram-negative bacteria, such as their protective outer membrane and constitutively overexpressed efflux pumps, is a major survival weapon that renders them refractory to current antibiotics. Several potential avenues to overcome this problem have been at the heart of antibiotic drug discovery in the past few decades. We review some of these strategies, with emphasis on antibiotic hybrids either as stand-alone antibacterial agents or as adjuvants that potentiate a primary antibiotic in Gram-negative bacteria. Antibiotic hybrid is defined in this review as a synthetic construct of two or more pharmacophores belonging to an established agent known to elicit a desired antimicrobial effect. The concepts, advances, and challenges of antibiotic hybrids are elaborated in this article. Moreover, we discuss several antibiotic hybrids that were or are in clinical evaluation. Mechanistic insights into how tobramycin-based antibiotic hybrids are able to potentiate legacy antibiotics in multidrug-resistant Gram-negative bacilli are also highlighted. Antibiotic hybrids indeed have a promising future as a therapeutic strategy to overcome drug resistance in Gram-negative pathogens and/or expand the usefulness of our current antibiotic arsenal.
Collapse
|
37
|
Thamban Chandrika N, Garneau-Tsodikova S. Comprehensive review of chemical strategies for the preparation of new aminoglycosides and their biological activities. Chem Soc Rev 2018; 47:1189-1249. [PMID: 29296992 PMCID: PMC5818290 DOI: 10.1039/c7cs00407a] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A systematic analysis of all synthetic and chemoenzymatic methodologies for the preparation of aminoglycosides for a variety of applications (therapeutic and agricultural) reported in the scientific literature up to 2017 is presented. This comprehensive analysis of derivatization/generation of novel aminoglycosides and their conjugates is divided based on the types of modifications used to make the new derivatives. Both the chemical strategies utilized and the biological results observed are covered. Structure-activity relationships based on different synthetic modifications along with their implications for activity and ability to avoid resistance against different microorganisms are also presented.
Collapse
Affiliation(s)
- Nishad Thamban Chandrika
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA.
| | | |
Collapse
|
38
|
Salta J, Benhamou RI, Herzog IM, Fridman M. Tuning the Effects of Bacterial Membrane Permeability through Photo-Isomerization of Antimicrobial Cationic Amphiphiles. Chemistry 2017; 23:12724-12728. [PMID: 28727190 DOI: 10.1002/chem.201703010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Indexed: 11/08/2022]
Abstract
Several important antimicrobial drugs act by permeabilizing cell membranes. In this study, we showed that the intensity of membrane permeability caused by antimicrobial cationic amphiphiles can be modified not only by their concentration but also through light-induced isomerization of their lipid segment. Two types of photo-isomerizable cationic amphiphiles were developed and the effects of photo-isomerization on bacterial growth and membrane permeability were evaluated. One photo-isomer inhibited cell growth and division, whereas the other photo-isomer led to a rapid and lethal bacterial membrane-disrupting effect. The switch from "on" to "off" can be obtained by either the cis- or trans-isomer depending on the bacterial strain and the type of cationic amphiphile. These cationic amphiphiles offer a novel tool for research and industrial applications that require light-controlled bacterial membrane permeabilization.
Collapse
Affiliation(s)
- Joana Salta
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Raphael I Benhamou
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Ido M Herzog
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Micha Fridman
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| |
Collapse
|
39
|
Liu B, Liu X, Zhang JR, Liu G. A Natural Lipotrisaccharide and Its Derivatives Selectively Lyse Streptococcus pneumoniae via Interaction with Cell Membrane. ACS Infect Dis 2017; 3:438-453. [PMID: 28264558 DOI: 10.1021/acsinfecdis.7b00008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A natural lipotrisaccharide (NP000778, 1a), a new triglycosidic tri-O-substituted glycolipid isolated from the Morinda citrifolia plant, and its chemical derivatives were identified to be active against major Gram-positive pathogens, particularly Streptococcus pneumoniae. Additional evidence indicated that 1a and its synthetic derivatives exerted their bactericidal activities against S. pneumoniae by selectively targeting the bacterial membrane, leading to the rapid lysis of the pneumococci. Efficient synthesis of 1a and its derivatives was performed using an application of the intramolecular aglycon delivery (IAD) reaction to establish its structure-activity relationships (SARs). SAR analysis indicated that trisaccharide glycolipid compounds showed good selectivity and high potency against S. pneumoniae. These compounds contain a linear chain with a chain length from C3 to C9 at the 2-position (R1) and 4'-position (R3), as well as a 2-methyl butyryl group at the 3'-position (R2), without an aza substitution in the lipid chain. This is the first lipotrisaccharide identified with potent bactericidal activity via interaction with cell membrane. The results reported herein offer a valuable guideline for the design of glycolipid derivatives that selectively target pathogenic bacteria.
Collapse
Affiliation(s)
- Bo Liu
- School of Pharmaceutical Sciences, Beijing 100084, People’s Republic of China
- Beijing Institute of Petrochemical Technology, Beijing 102607, People’s Republic of China
| | - Xue Liu
- Center
for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Jing-Ren Zhang
- Center
for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Gang Liu
- School of Pharmaceutical Sciences, Beijing 100084, People’s Republic of China
| |
Collapse
|
40
|
Yang X, Goswami S, Gorityala BK, Domalaon R, Lyu Y, Kumar A, Zhanel GG, Schweizer F. A Tobramycin Vector Enhances Synergy and Efficacy of Efflux Pump Inhibitors against Multidrug-Resistant Gram-Negative Bacteria. J Med Chem 2017; 60:3913-3932. [DOI: 10.1021/acs.jmedchem.7b00156] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Xuan Yang
- Department
of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Sudeep Goswami
- Department
of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | | | - Ronald Domalaon
- Department
of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Yinfeng Lyu
- Department
of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Institute
of Animal Nutrition, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China
| | - Ayush Kumar
- Department
of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department
of Medical Microbiology, University of Manitoba, Winnipeg, MB R3T 1R9, Canada
| | - George G. Zhanel
- Department
of Medical Microbiology, University of Manitoba, Winnipeg, MB R3T 1R9, Canada
| | - Frank Schweizer
- Department
of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department
of Medical Microbiology, University of Manitoba, Winnipeg, MB R3T 1R9, Canada
| |
Collapse
|
41
|
Lyu Y, Yang X, Goswami S, Gorityala BK, Idowu T, Domalaon R, Zhanel GG, Shan A, Schweizer F. Amphiphilic Tobramycin-Lysine Conjugates Sensitize Multidrug Resistant Gram-Negative Bacteria to Rifampicin and Minocycline. J Med Chem 2017; 60:3684-3702. [PMID: 28409644 DOI: 10.1021/acs.jmedchem.6b01742] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chromosomally encoded low membrane permeability and highly efficient efflux systems are major mechanisms by which Pseudomonas aeruginosa evades antibiotic actions. Our previous reports have shown that amphiphilic tobramycin-fluoroquinolone hybrids can enhance efficacy of fluoroquinolone antibiotics against multidrug-resistant (MDR) P. aeruginosa isolates. Herein, we report on a novel class of tobramycin-lysine conjugates containing an optimized amphiphilic tobramycin-C12 tether that sensitize Gram-negative bacteria to legacy antibiotics. Combination studies indicate the ability of these conjugates to synergize rifampicin and minocycline against MDR and extensively drug resistant (XDR) P. aeruginosa isolates and enhance efficacy of both antibiotics in the Galleria mellonella larvae in vivo infection model. Mode of action studies indicate that the amphiphilic tobramycin-lysine adjuvants enhance outer membrane cell penetration and affect the proton motive force, which energizes efflux pumps. Overall, this study provides a strategy for generating effective antibiotic adjuvants that overcome resistance of rifampicin and minocycline in MDR and XDR Gram-negative bacteria including P. aeruginosa.
Collapse
Affiliation(s)
- Yinfeng Lyu
- Institute of Animal Nutrition, Northeast Agricultural University , Harbin, Heilongjiang 150030, P.R. China.,Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada
| | - Xuan Yang
- Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada
| | - Sudeep Goswami
- Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada
| | | | - Temilolu Idowu
- Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada
| | - Ronald Domalaon
- Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada
| | - George G Zhanel
- Department of Medical Microbiology/Infectious Diseases, University of Manitoba , Winnipeg, MB R3T 1R9, Canada
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University , Harbin, Heilongjiang 150030, P.R. China
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba , Winnipeg, MB R3T 2N2, Canada.,Department of Medical Microbiology/Infectious Diseases, University of Manitoba , Winnipeg, MB R3T 1R9, Canada
| |
Collapse
|
42
|
Degtyareva NN, Gong C, Story S, Levinson NS, Oyelere AK, Green KD, Garneau-Tsodikova S, Arya DP. Antimicrobial Activity, AME Resistance, and A-Site Binding Studies of Anthraquinone-Neomycin Conjugates. ACS Infect Dis 2017; 3:206-215. [PMID: 28103015 PMCID: PMC5971063 DOI: 10.1021/acsinfecdis.6b00176] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The antibacterial effects of aminoglycosides are based on their association with the A-site of bacterial rRNA and interference with the translational process in the bacterial cell, causing cell death. The clinical use of aminoglycosides is complicated by resistance and side effects, some of which arise from their interactions with the human mitochondrial 12S rRNA and its deafness-associated mutations, C1494U and A1555G. We report a rapid assay that allows screening of aminoglycoside compounds to these classes of rRNAs. These screening tools are important to find antibiotics that selectively bind to the bacterial A-site rather than human, mitochondrial A-sites and its mutant homologues. Herein, we report our preliminary work on the optimization of this screen using 12 anthraquinone-neomycin (AMA-NEO) conjugates against molecular constructs representing five A-site homologues, Escherichia coli, human cytosolic, mitochondrial, C1494U, and A1555G, using a fluorescent displacement screening assay. These conjugates were also tested for inhibition of protein synthesis, antibacterial activity against 14 clinically relevant bacterial strains, and the effect on enzymes that inactivate aminoglycosides. The AMA-NEO conjugates demonstrated significantly improved resistance against aminoglycoside-modifying enzymes (AMEs), as compared with NEO. Several compounds exhibited significantly greater inhibition of prokaryotic protein synthesis as compared to NEO and were extremely poor inhibitors of eukaryotic translation. There was significant variation in antibacterial activity and MIC of selected compounds between bacterial strains, with Escherichia coli, Enteroccocus faecalis, Citrobacter freundii, Shigella flexneri, Serratia marcescens, Proteus mirabilis, Enterobacter cloacae, Staphylococcus epidermidis, and Listeria monocytogenes exhibiting moderate to high sensitivity (50-100% growth inhibition) whereas Acinetobacter baumannii, Pseudomonas aeruginosa, Klebsiellla pneumoniae, and MRSA strains expressed low sensitivity, as compared to the parent aminoglycoside NEO.
Collapse
Affiliation(s)
| | - Changjun Gong
- Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Sandra Story
- NUBAD, LLC, Greenville, South Carolina 29605, United States
| | - Nathanael S. Levinson
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| | - Adegboyega K. Oyelere
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| | - Keith D. Green
- College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0596, United States
| | | | - Dev P. Arya
- NUBAD, LLC, Greenville, South Carolina 29605, United States
- Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| |
Collapse
|
43
|
Allam A, Maigre L, Alves de Sousa R, Dumont E, Vergalli J, Pagès JM, Artaud I. New amphiphilic neamine conjugates bearing a metal binding motif active against MDR E. aerogenes Gram-negative bacteria. Eur J Med Chem 2017; 127:748-756. [DOI: 10.1016/j.ejmech.2016.10.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/29/2016] [Accepted: 10/23/2016] [Indexed: 01/22/2023]
|
44
|
Abouelhassan Y, Basak A, Yousaf H, Huigens RW. Identification of N-Arylated NH125 Analogues as Rapid Eradicating Agents against MRSA Persister Cells and Potent Biofilm Killers of Gram-Positive Pathogens. Chembiochem 2017; 18:352-357. [PMID: 27925693 DOI: 10.1002/cbic.201600622] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Indexed: 11/11/2022]
Abstract
Bacterial biofilms housing dormant persister cells are innately tolerant to antibiotics and disinfectants, yet several membrane-active agents are known to eradicate tolerant bacterial cells. NH125, a membrane-active persister killer and starting point for development, led to the identification of two N-arylated analogues (1 and 2) that displayed improved biofilm eradication potencies compared to the parent compound and rapid persister-cell-killing activities in stationary cultures of methicillin-resistant Staphylococcus aureus (MRSA). We found 1 and 2 to be superior to other membrane-active agents in biofilm eradication assays, with 1 demonstrating minimum biofilm eradication concentrations (MBEC) of 23.5, 11.7, and 2.35 μm against MRSA, methicillin-resistant Staphylococcus epidermidis (MRSE), and vancomycin-resistant Enterococcus faecium (VRE) biofilms, respectively. We tested our panel of membrane-active agents against MRSA stationary cultures and found 1 to rapidly eradicate MRSA stationary cells by 4 log units (99.99 %) in 30 min. The potent biofilm eradication and rapid persister-cell-killing activities exhibited by N-arylated NH125 analogues could have significant impact in addressing biofilm-associated problems.
Collapse
Affiliation(s)
- Yasmeen Abouelhassan
- Department of Medicinal Chemistry, Center for Natural Products Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, FL, 32610, USA
| | - Akash Basak
- Department of Chemistry, University of Florida, 1600 SW Archer Road, Gainesville, FL, 32610, USA
| | - Hussain Yousaf
- Department of Medicinal Chemistry, Center for Natural Products Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, FL, 32610, USA
| | - Robert W Huigens
- Department of Medicinal Chemistry, Center for Natural Products Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, FL, 32610, USA.,Department of Chemistry, University of Florida, 1600 SW Archer Road, Gainesville, FL, 32610, USA
| |
Collapse
|
45
|
Ngo HX, Shrestha SK, Green KD, Garneau-Tsodikova S. Development of ebsulfur analogues as potent antibacterials against methicillin-resistant Staphylococcus aureus. Bioorg Med Chem 2016; 24:6298-6306. [PMID: 27073054 PMCID: PMC5045767 DOI: 10.1016/j.bmc.2016.03.060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/26/2016] [Accepted: 03/30/2016] [Indexed: 11/21/2022]
Abstract
Antibiotic resistance is a worldwide problem that needs to be addressed. Staphylococcus aureus is one of the dangerous "ESKAPE" pathogens that rapidly evolve and evade many current FDA-approved antibiotics. Thus, there is an urgent need for new anti-MRSA compounds. Ebselen (also known as 2-phenyl-1,2-benzisoselenazol-3(2H)-one) has shown promising activity in clinical trials for cerebral ischemia, bipolar disorder, and noise-induced hearing loss. Recently, there has been a renewed interest in exploring the antibacterial properties of ebselen. In this study, we synthesized an ebselen-inspired library of 33 compounds where the selenium atom has been replaced by sulfur (ebsulfur derivatives) and evaluated them against a panel of drug-sensitive and drug-resistant S. aureus and non-S. aureus strains. Within our library, we identified three outstanding analogues with potent activity against all S. aureus strains tested (MIC values mostly ⩽2μg/mL), and numerous additional ones with overall very good to good antibacterial activity (1-7.8μg/mL). We also characterized the time-kill analysis, anti-biofilm ability, hemolytic activity, mammalian cytotoxicity, membrane-disruption ability, and reactive oxygen species (ROS) production of some of these analogues.
Collapse
Affiliation(s)
- Huy X Ngo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536-0596, USA
| | - Sanjib K Shrestha
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536-0596, USA
| | - Keith D Green
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536-0596, USA
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536-0596, USA.
| |
Collapse
|
46
|
Zhang Q, Alfindee MN, Shrestha JP, Nziko VDPN, Kawasaki Y, Peng X, Takemoto JY, Chang CWT. Divergent Synthesis of Three Classes of Antifungal Amphiphilic Kanamycin Derivatives. J Org Chem 2016; 81:10651-10663. [DOI: 10.1021/acs.joc.6b01189] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Qian Zhang
- Department
of Chemistry and Biochemistry, Utah State University, 0300 Old
Main Hill, Logan, Utah 84322-0300, United States
| | - Madher N. Alfindee
- Department
of Chemistry and Biochemistry, Utah State University, 0300 Old
Main Hill, Logan, Utah 84322-0300, United States
| | - Jaya P. Shrestha
- Department
of Chemistry and Biochemistry, Utah State University, 0300 Old
Main Hill, Logan, Utah 84322-0300, United States
| | - Vincent de Paul Nzuwah Nziko
- Department
of Chemistry and Biochemistry, Utah State University, 0300 Old
Main Hill, Logan, Utah 84322-0300, United States
| | - Yukie Kawasaki
- Department
of Biology, Utah State University, 5305 Old Main Hill, Logan, Utah 84322-5305, United States
| | - Xinrui Peng
- Department
of Chemistry and Biochemistry, Utah State University, 0300 Old
Main Hill, Logan, Utah 84322-0300, United States
| | - Jon Y. Takemoto
- Department
of Biology, Utah State University, 5305 Old Main Hill, Logan, Utah 84322-5305, United States
| | - Cheng-Wei Tom Chang
- Department
of Chemistry and Biochemistry, Utah State University, 0300 Old
Main Hill, Logan, Utah 84322-0300, United States
| |
Collapse
|
47
|
Zimmermann L, Das I, Désiré J, Sautrey G, Barros R. S. V, El Khoury M, Mingeot-Leclercq MP, Décout JL. New Broad-Spectrum Antibacterial Amphiphilic Aminoglycosides Active against Resistant Bacteria: From Neamine Derivatives to Smaller Neosamine Analogues. J Med Chem 2016; 59:9350-9369. [DOI: 10.1021/acs.jmedchem.6b00818] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Louis Zimmermann
- Département
de Pharmacochimie Moléculaire, ICMG FR 2607, University Grenoble Alpes/CNRS, UMR 5063, 470 Rue de la Chimie, BP 53, F-38041 Grenoble, France
| | - Indrajit Das
- Département
de Pharmacochimie Moléculaire, ICMG FR 2607, University Grenoble Alpes/CNRS, UMR 5063, 470 Rue de la Chimie, BP 53, F-38041 Grenoble, France
| | - Jérôme Désiré
- Département
de Pharmacochimie Moléculaire, ICMG FR 2607, University Grenoble Alpes/CNRS, UMR 5063, 470 Rue de la Chimie, BP 53, F-38041 Grenoble, France
| | - Guillaume Sautrey
- Unité
de Pharmacologie Cellulaire et Moléculaire, Louvain Drug Research
Institute, Université Catholique de Louvain, Avenue E.
Mounier 73, B1.73.05, B-1200 Brussels, Belgium
| | - Vinicius Barros R. S.
- Département
de Pharmacochimie Moléculaire, ICMG FR 2607, University Grenoble Alpes/CNRS, UMR 5063, 470 Rue de la Chimie, BP 53, F-38041 Grenoble, France
| | - Micheline El Khoury
- Unité
de Pharmacologie Cellulaire et Moléculaire, Louvain Drug Research
Institute, Université Catholique de Louvain, Avenue E.
Mounier 73, B1.73.05, B-1200 Brussels, Belgium
| | - Marie-Paule Mingeot-Leclercq
- Unité
de Pharmacologie Cellulaire et Moléculaire, Louvain Drug Research
Institute, Université Catholique de Louvain, Avenue E.
Mounier 73, B1.73.05, B-1200 Brussels, Belgium
| | - Jean-Luc Décout
- Département
de Pharmacochimie Moléculaire, ICMG FR 2607, University Grenoble Alpes/CNRS, UMR 5063, 470 Rue de la Chimie, BP 53, F-38041 Grenoble, France
| |
Collapse
|
48
|
Ghosh C, Konai MM, Sarkar P, Samaddar S, Haldar J. Designing Simple Lipidated Lysines: Bifurcation Imparts Selective Antibacterial Activity. ChemMedChem 2016; 11:2367-2371. [DOI: 10.1002/cmdc.201600400] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Chandradhish Ghosh
- Chemical Biology and Medicinal Chemistry Laboratory, New Chemistry Unit; Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru; 560064 Karnataka India
| | - Mohini Mohan Konai
- Chemical Biology and Medicinal Chemistry Laboratory, New Chemistry Unit; Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru; 560064 Karnataka India
| | - Paramita Sarkar
- Chemical Biology and Medicinal Chemistry Laboratory, New Chemistry Unit; Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru; 560064 Karnataka India
| | - Sandip Samaddar
- Chemical Biology and Medicinal Chemistry Laboratory, New Chemistry Unit; Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru; 560064 Karnataka India
| | - Jayanta Haldar
- Chemical Biology and Medicinal Chemistry Laboratory, New Chemistry Unit; Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru; 560064 Karnataka India
| |
Collapse
|
49
|
Benhamou RI, Steinbuch KB, Fridman M. Antifungal Imidazole-Decorated Cationic Amphiphiles with Markedly Low Hemolytic Activity. Chemistry 2016; 22:11148-51. [DOI: 10.1002/chem.201602198] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Raphael I. Benhamou
- School of Chemistry; Raymond & Beverly Sackler Faculty of Exact Sciences; Tel Aviv University; 6997801 Tel Aviv Israel
| | - Kfir B. Steinbuch
- School of Chemistry; Raymond & Beverly Sackler Faculty of Exact Sciences; Tel Aviv University; 6997801 Tel Aviv Israel
| | - Micha Fridman
- School of Chemistry; Raymond & Beverly Sackler Faculty of Exact Sciences; Tel Aviv University; 6997801 Tel Aviv Israel
| |
Collapse
|
50
|
Mingeot-Leclercq MP, Décout JL. Bacterial lipid membranes as promising targets to fight antimicrobial resistance, molecular foundations and illustration through the renewal of aminoglycoside antibiotics and emergence of amphiphilic aminoglycosides. MEDCHEMCOMM 2016. [DOI: 10.1039/c5md00503e] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Membrane anionic lipids as attractive targets in the design of amphiphilic antibacterial drugs active against resistant bacteria: molecular foundations and examples.
Collapse
Affiliation(s)
- Marie-Paule Mingeot-Leclercq
- Louvain Drug Research Institute
- Université catholique de Louvain
- Unité de Pharmacologie Cellulaire et Moléculaire
- Brussels
- Belgium
| | - Jean-Luc Décout
- Département de Pharmacochimie Moléculaire
- Université Grenoble Alpes/CNRS
- UMR 5063
- ICMG FR 2607
- F-38041 Grenoble
| |
Collapse
|