Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Org Chem ; 88(9): 5572-5585, 2023 05 05.
Article in English | MEDLINE | ID: mdl-37083436

ABSTRACT

Organoselenium compounds are important scaffolds in pharmaceutical molecules. Herein, we report metal-free, electrochemical, highly chemo- and regioselective synthesis of gem-diselenides through the coupling of α-keto sulfoxonium ylides with diselenides. The versatility of the electrochemical manifold enabled the selenylation with ample scope and broad functional group tolerance, as well as setting the stage for modification of complex bioactive molecules. Detailed mechanistic studies revealed that the key C-Se bond was constructed using n-Bu4NI as an electrolyte and catalyst through the electrosynthetic protocol. Finally, the desired α-keto gem-diselenides showed excellent antimicrobial activity against Candida albicans, which can be identified as the lead compounds for further exploration.


Subject(s)
Anti-Infective Agents , Anti-Infective Agents/pharmacology , Pharmaceutical Preparations , Fungi
2.
Acc Chem Res ; 54(1): 104-119, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33346639

ABSTRACT

The alarming rate at which pathogens are developing resistance to conventional antibiotics represents one of the major global challenges to public health care. The prevalence of multidrug-resistant microorganisms is a major impetus for the discovery and development of new antimicrobials. Nature has, to date, been the source of most of the antibiotics discovered and used, including cationic antimicrobial peptides (CAMPs). CAMPs are key components of the innate immune system that are widely found in humans, animals, plants, and microorganisms and that serve as a first line of defense for the host. The attractive features of CAMPs have led to their recognition as potential new antimicrobials. However, they possess several inherent flaws that limit their clinical application including low stability, poor oral bioavailability, poor in vivo efficacy, and a high production cost. To address these issues, small molecule-based peptidomimetic antimicrobials have been designed to biomimic the structural features and biological function of CAMPs. Plant-derived flavonoids (e.g., xanthones and flavones) are active components in traditional herbal medicines and have been reported to contain a variety of significant pharmacological actions including antibacterial, antiviral, antioxidant, and anticancer activities. Over the past decade, we have developed a new chemical strategy to design, discover, and develop xanthone- or flavone-based peptidomimetics and have designed, synthesized, and biologically evaluated a library of approximately 450 new xanthone or flavone derivatives. The designed, structurally diverse compounds can be generally classified into two subfamilies, namely, peptidic and nonpeptidic amphiphilic xanthone or flavone derivatives. In this Account, we describe our efforts on the design, synthesis, biological property evaluation, and mechanism of action model studies of synthetic mimics of CAMPs. The flavonoid compounds are an important component of these rationally designed mimics because they function as hydrophobic aromatic moieties conjugated with different length lipid moieties, behave like an unnatural hydrophobic residue, and provide a rigid scaffold, with the reduced conformational flexibility more likely to provide an active conformation. The mimics can effectively disrupt the integrity of the bacterial membranes. Our endeavors encompass design principles, chemical synthesis, in vitro screening, structural optimization, extensive structural-activity relationship analysis, and a mechanism of action study through biophysical technologies including NMR spectroscopy techniques and computer dynamics simulations, drug resistance assays, in vivo pharmaceutical kinetics (PK) analyses, and in vivo efficacy evaluations of selected promising compounds against drug-resistant bacteria and fungi. Our major contributions to the discovery and development of flavonoid-based mimics as antimicrobials include effectively addressing several limitations associated with CAMPs and have led to promising compounds with a notable potential for further development as new therapeutic antimicrobial agents for the treatment of drug-resistant bacteria- or fungi-induced infections.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Drug Design , Flavonoids/chemistry , Antimicrobial Cationic Peptides/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Gram-Positive Bacteria/drug effects , Hemolysis/drug effects , Microbial Sensitivity Tests , Peptidomimetics/chemical synthesis , Peptidomimetics/chemistry , Structure-Activity Relationship , Xanthones/chemistry
3.
Amino Acids ; 50(10): 1329-1345, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30066172

ABSTRACT

Multi-drug resistant pathogens have been of increasing concern today. There is an urgent need for the discovery of more potent antibiotics. Cationic antimicrobial peptides (CAMPs) are known to be effective antimicrobial agents against resistant pathogens. However, poor activity under physiological conditions is one of the major limitations of CAMPS in clinical applications. In this study, a series of oligo-lipidated arginyl peptide OLAP dimers comprised of a saturated fatty acid chain (with m number of carbon units) and p repeating units of arginyl fatty acid chains (with n number of carbon units) were designed and studied for their antimicrobial activities as well as their physico-chemical property in various physiological conditions, such as in human serum albumin and high salt conditions. Our results showed that OLAP-11 exhibits potent antimicrobial activity against Gram-positive bacteria with improved physico-chemical activity in various physiological conditions. OLAP-11 is also less susceptible to human serum and trypsin degradation. The HPLC-MS analysis showed that the lipid-arginine bond is very stable. SYTOX Green assay and scanning electron microscopy both show that the OLAP-11 killed bacteria via inner membrane disruption. In addition, OLAP-11 is inner membrane targeting, making it difficult for bacteria to develop resistance. Overall, the design of the OLAP dimers provides an alternative approach to improve the physicochemical activity, peptide stability of CAMPs with potent inner membrane disruption and low in vitro toxicity to increase their potential for clinical applications in the future.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Fatty Acids/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/microbiology , Antimicrobial Cationic Peptides/chemical synthesis , Dimerization , Drug Design , Drug Stability , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , Humans , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Structure-Activity Relationship
4.
Amino Acids ; 49(10): 1653-1677, 2017 10.
Article in English | MEDLINE | ID: mdl-28823054

ABSTRACT

Infectious diseases impose serious public health burdens and continue to be a global public health crisis. The treatment of infections caused by multidrug-resistant pathogens is challenging because only a few viable therapeutic options are clinically available. The emergence and risk of drug-resistant superbugs and the dearth of new classes of antibiotics have drawn increasing awareness that we may return to the pre-antibiotic era. To date, lipopeptides have been received considerable attention because of the following properties: They exhibit potent antimicrobial activities against a broad spectrum of pathogens, rapid bactericidal activity and have a different antimicrobial action compared with most of the conventional antibiotics used today and very slow development of drug resistance tendency. In general, lipopeptides can be structurally classified into two parts: a hydrophilic peptide moiety and a hydrophobic fatty acyl chain. To date, a significant amount of design and synthesis of lipopeptides have been done to improve the therapeutic potential of lipopeptides. This review will present the current knowledge and the recent research in design and synthesis of new lipopeptides and their derivatives in the last 5 years.


Subject(s)
Anti-Infective Agents , Lipopeptides , Animals , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/chemistry , Anti-Infective Agents/therapeutic use , Humans , Lipopeptides/chemical synthesis , Lipopeptides/chemistry , Lipopeptides/therapeutic use
5.
Antimicrob Agents Chemother ; 60(1): 24-35, 2016 01.
Article in English | MEDLINE | ID: mdl-26459903

ABSTRACT

Microbial infections of the cornea are potentially devastating and can result in permanent visual loss or require vision-rescuing surgery. In recent years, there has been an increasing number of reports on nontuberculous mycobacterial infections of the cornea. Challenges to the management of nontuberculous mycobacterial keratitis include delayed laboratory detection, low index of clinical suspicion, poor drug penetration, slow response to therapy, and prolonged use of antibiotic combinations. The ability of nontuberculous mycobacteria to evade the host immune response and the ability to adhere and to form biofilms on biological and synthetic substrates contribute to the issue. Therefore, there is an urgent need for new antimicrobial compounds that can overcome these problems. In this study, we evaluated the biofilm architectures for Mycobacterium chelonae and Mycobacterium fortuitum in dynamic flow cell chamber and 8-well chamber slide models. Our results showed that mycobacterial biofilms were quite resistant to conventional antibiotics. However, DNase treatment could be used to overcome biofilm resistance. Moreover, we successfully evaluated a new antimicrobial compound (AM-228) that was effective not only for planktonic mycobacterial cells but also for biofilm treatment and was compared favorably with the most successful "fourth-generation" fluoroquinolone, gatifloxacin. Finally, a new treatment strategy emerged: a combination of DNase with an antibiotic was more effective than an antibiotic alone.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Deoxyribonucleases/pharmacology , Mycobacterium chelonae/drug effects , Mycobacterium fortuitum/drug effects , Xanthones/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Biofilms/growth & development , Cornea/drug effects , Cornea/microbiology , Diffusion Chambers, Culture , Drug Synergism , Drug Therapy, Combination , Fluoroquinolones/pharmacology , Gatifloxacin , Mycobacterium chelonae/physiology , Mycobacterium fortuitum/physiology , Plankton/drug effects , Plankton/growth & development , Rabbits , Rheology , Wound Healing/drug effects , Xanthones/chemical synthesis
6.
Eur J Med Chem ; 274: 116544, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38850855

ABSTRACT

Antibiotic resistance is becoming increasingly severe. The development of small molecular antimicrobial peptides is regarded as a promising design strategy for antibiotics. Here, a series of bisphenol derivatives with amphiphilic structures were designed and synthesized as antibacterial agents by imitating the design strategy of antimicrobial peptides. After a series of structural optimizations, lead compound 43 was identified, which exhibited excellent antibacterial activity against Gram-positive bacterial strains (MICs = 0.78-1.56 µg/mL), poor hemolytic activity (HC50 > 200 µg/mL), and low cytotoxicity (CC50 > 100 µg/mL). Further biological evaluation results indicated that 43 exerted antibacterial effects by directly destroying bacterial cell membranes and displayed rapid bactericidal properties (within 0.5-1 h), leading to a very low probability of drug resistance. Moreover, in a murine model of corneal infection, 43 exhibited a strong in vivo antibacterial efficacy. These findings indicate that 43 is a promising candidate compound for the treatment of bacterial infections.


Subject(s)
Anti-Bacterial Agents , Benzhydryl Compounds , Gram-Positive Bacteria , Microbial Sensitivity Tests , Phenols , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Phenols/pharmacology , Phenols/chemistry , Phenols/chemical synthesis , Animals , Gram-Positive Bacteria/drug effects , Mice , Structure-Activity Relationship , Benzhydryl Compounds/pharmacology , Benzhydryl Compounds/chemistry , Benzhydryl Compounds/chemical synthesis , Molecular Structure , Cell Membrane/drug effects , Dose-Response Relationship, Drug , Humans , Hemolysis/drug effects , Drug Development
7.
Eur J Med Chem ; 266: 116149, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38266554

ABSTRACT

The emergence of antibiotic resistance has brought a significant burden to public health. Here, we designed and synthesized a series of cannabidiol derivatives by biomimicking the structure and function of cationic antibacterial peptides. This is the first report on the design of cannabidiol derivatives as broad-spectrum antibacterial agents. Through the structure-activity relationship (SAR) study, we found a lead compound 23 that killed both Gram-negative and Gram-positive bacteria via a membrane-targeting mechanism of action with low resistance frequencies. Compound 23 also exhibited very weak hemolytic activity, low toxicity toward mammalian cells, and rapid bactericidal properties. To further validate the membrane action mechanism of compound 23, we performed transcriptomic analysis using RNA-seq, which revealed that treatment with compound 23 altered many cell wall/membrane/envelope biogenesis-related genes in Gram-positive and Gram-negative bacteria. More importantly, compound 23 showed potent in vivo antibacterial efficacy in murine corneal infection models caused by Staphylococcus aureus or Pseudomonas aeruginosa. These findings would provide a new design idea for the discovery of novel broad-spectrum antibacterial agents to overcome the antibiotic resistance crisis.


Subject(s)
Anti-Bacterial Agents , Cannabidiol , Animals , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Cannabidiol/pharmacology , Gram-Negative Bacteria , Gram-Positive Bacteria , Mammals , Microbial Sensitivity Tests , Peptides/chemistry , Peptides/pharmacology
8.
Eur J Med Chem ; 260: 115733, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37643545

ABSTRACT

Multidrug-resistant bacteria infections pose an increasingly serious threat to human health, and the development of antimicrobials is far from meeting the clinical demand. It is urgent to discover and develop novel antibiotics to combat bacterial resistance. Currently, the development of membrane active antimicrobial agents is an attractive strategy to cope with antimicrobial resistance issues. In this study, the synthesis and biological evaluation of cationic amphiphilic phenothiazine-based derivatives were reported. Among them, the most promising compound 30 bearing a n-heptyl group and two arginine residues displayed potent bactericidal activity against both Gram-positive (MICs = 1.56 µg/mL) and Gram-negative bacteria (MICs = 3.125-6.25 µg/mL). Compound 30 showed low hemolysis activity (HC50 = 281.4 ± 1.6 µg/mL) and low cytotoxicity (CC50 > 50 µg/mL) toward mammalian cells, as well as excellent salt resistance. Compound 30 rapidly killed bacteria by acting on the bacterial cell membrane and appeared less prone to resistance. Importantly, compound 30 showed potent in vivo efficacy in a murine model of bacterial keratitis. Hence, the results suggested compound 30 has a promising prospect as a broad-spectrum antibacterial agent for the treatment of drug-resistant bacterial infections.


Subject(s)
Anti-Infective Agents , Antipsychotic Agents , Heterocyclic Compounds , Humans , Animals , Mice , Phenothiazines/pharmacology , Bacteria , Anti-Bacterial Agents/pharmacology , Arginine , Cations , Mammals
9.
J Med Chem ; 65(20): 14221-14236, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36256884

ABSTRACT

Due to the emergence of antimicrobial resistance and the lack of new antibacterial agents, it has become urgent to discover and develop new antibacterial agents against multidrug-resistant pathogens. Antimicrobial peptides (AMPs) serve as the first line of defense for the host. In this work, we have designed, synthesized, and biologically evaluated a series of phenyl sulfide derivatives by biomimicking the structural features and biological functions of AMPs. Among these derivatives, the most promising compound 17 exhibited potent antibacterial activity against Gram-positive bacteria (minimum inhibitory concentrations = 0.39-1.56 µg/mL), low hemolytic activity (HC50 > 200 µg/mL), and high membrane selectivity. In addition, 17 can rapidly kill Gram-positive bacteria within 0.5 h through membrane-targeting action and avoid antibiotic resistance. More importantly, 17 showed high in vivo efficacy against Staphylococcus aureus in a murine corneal infection model. Therefore, 17 has great potential as a lead compound for the treatment of Gram-positive bacterial infections.


Subject(s)
Anti-Infective Agents , Gram-Positive Bacteria , Mice , Animals , Microbial Sensitivity Tests , Anti-Bacterial Agents/chemistry , Cations , Sulfides/pharmacology , Sulfides/therapeutic use
10.
Eur J Med Chem ; 243: 114734, 2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36088756

ABSTRACT

The rising prevalence of drug-resistant pathogens is one of the biggest threats to human health. The development of new antibiotics that can overcome drug resistance is in urgent need. Herein, we designed and synthesized a series of amphiphilic tetrahydroquinoline derivatives as small-molecule-based antimicrobial peptidomimetics. Two lead compounds 36 and 52 which contained the tetrahydroquinoline core, hydrophobic alkyl chains (n-nonyl or isoprenyl group), different spacer lengths (n = 4 or 8), and cationic guanidine moiety, showed poor hemolytic activity, low cytotoxicity, and potent broad-spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria, as well as fungi. The further biological evaluation revealed that compounds 36 and 52 can kill bacteria and fungi rapidly via membrane-targeting action and avoid drug resistance development. More importantly, compounds 36 and 52 exhibited similarly potent in vivo antimicrobial activities in a murine corneal infection caused by Staphylococcus aureus ATCC29213 or Pseudomonas aeruginosa ATCC9027, as compared to vancomycin or gatifloxacin. These results suggest that compounds 36 and 52 have great potential as new broad-spectrum antimicrobial agents to combat microbial resistance.


Subject(s)
Anti-Infective Agents , Gram-Negative Bacteria , Humans , Mice , Animals , Gram-Positive Bacteria , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests , Bacteria , Fungi
11.
J Med Chem ; 65(24): 16754-16773, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36510819

ABSTRACT

Antibiotic resistance is emerging as a "global public health concern". To address the growing epidemic of multidrug-resistant pathogens, the development of novel antimicrobials is urgently needed. In this study, by biomimicking cationic antibacterial peptides, we designed and synthesized a series of new membrane-active nonivamide and capsaicin derivatives as peptidomimetic antimicrobials. Through modulating charge/hydrophobicity balance and rationalizing structure-activity relationships of these peptidomimetics, compound 51 was identified as the lead compound. Compound 51 exhibited potent antibacterial activity against both Gram-positive bacteria (MICs = 0.39-0.78 µg/mL) and Gram-negative bacteria (MICs = 1.56-6.25 µg/mL), with low hemolytic activity and low cytotoxicity. Compound 51 displayed a faster bactericidal action through a membrane-disruptive mechanism and avoided bacterial resistance development. Furthermore, compound 51 significantly reduced the microbial burden in a murine model of keratitis infected by Staphylococcus aureus or Pseudomonas aeruginosa. Hence, this design strategy can provide a promising and effective solution to overcome antibiotic resistance.


Subject(s)
Anti-Infective Agents , Capsaicin , Animals , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Structure-Activity Relationship , Microbial Sensitivity Tests
12.
Front Chem ; 10: 816741, 2022.
Article in English | MEDLINE | ID: mdl-35211455

ABSTRACT

Bacteria have developed increasing resistance to currently used antimicrobial agents. New classes of antimicrobial drugs are urgently required to fight drug-resistant pathogens. Here, we designed and synthesized a series of calix[4]arene derivatives as antibacterial agents by biomimicking the structural properties and biological functions of antibacterial peptides. After introducing cationic hydrophilic moieties and preliminary structural optimization, we obtained a lead compound (16) that exhibited excellent antibacterial activity against Gram-positive bacteria, low toxicity toward mammalian cells and poor hemolytic activity. The antibacterial mechanism studies showed that compound 16 can destroy bacterial cell membrane directly, leading to bacterial death and a low tendency to develop bacterial resistance.

13.
J Med Chem ; 64(9): 5603-5619, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33909443

ABSTRACT

Infections caused by drug-resistant bacteria seriously endanger human health and global public health. Therefore, it is urgent to discover and develop novel antimicrobial agents to combat multidrug-resistant bacteria. In this study, we designed and synthesized a series of new membrane-active bakuchiol derivatives by biomimicking the structure and function of cationic antibacterial peptides. The most promising compound 28 displayed potent antibacterial activity against both Gram-positive bacteria (minimum inhibitory concentration, MIC = 1.56-3.125 µg/mL) and Gram-negative bacteria (MIC = 3.125 µg/mL), very weak hemolytic activity, and low cytotoxicity. Compound 28 had rapid bactericidal properties and avoided bacterial resistance. More importantly, compound 28 showed strong in vivo antibacterial efficacy against Staphylococcus aureus and Pseudomonas aeruginosa in murine corneal infection models. This design strategy is expected to provide an effective solution to the antibiotic crisis.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Drug Design , Phenols/chemistry , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Cell Line , Cell Survival/drug effects , Corneal Diseases/drug therapy , Corneal Diseases/microbiology , Corneal Diseases/pathology , Disease Models, Animal , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Hemolysis/drug effects , Mice , Microbial Sensitivity Tests , Phenols/pharmacology , Phenols/therapeutic use , Staphylococcus aureus/physiology , Structure-Activity Relationship
14.
J Med Chem ; 64(14): 10429-10444, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34235929

ABSTRACT

Antibiotic resistance has become one of the most urgently important problems facing healthcare providers. A novel series of dipicolylamine-containing carbazole amphiphiles with strong Zn2+ chelating ability were synthesized, biomimicking cationic antimicrobial peptides. Effective broad-spectrum 16 combined with 12.5 µg/mL Zn2+ was identified as the most promising antimicrobial candidate. 16 combined with 12.5 µg/mL Zn2+ exhibited excellent antimicrobial activity against both Gram-positive and Gram-negative bacteria (MICs = 0.78-3.125 µg/mL), weak hemolytic activity, and low cytotoxicity. Time-kill kinetics and mechanism studies revealed 16 combined with 12.5 µg/mL Zn2+ had rapid bacterial killing properties, as evidenced by disruption of the integrity of bacterial cell membranes, effectively preventing bacterial resistance development. Importantly, 16 combined with 12.5 µg/mL Zn2+ showed excellent in vivo efficacy in a murine keratitis model caused by Staphylococcus aureus ATCC29213 or Pseudomonas aeruginosa ATCC9027. Therefore, 16 combined with 12.5 µg/mL Zn2+ could be a promising candidate for treating bacterial infections.


Subject(s)
Amines/pharmacology , Anti-Bacterial Agents/pharmacology , Carbazoles/pharmacology , Coordination Complexes/pharmacology , Drug Design , Picolinic Acids/pharmacology , Zinc/pharmacology , Amines/chemistry , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Carbazoles/chemistry , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Female , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Keratitis/drug therapy , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Molecular Structure , Picolinic Acids/chemistry , Structure-Activity Relationship , Zinc/chemistry
15.
ACS Infect Dis ; 7(10): 2864-2875, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34505771

ABSTRACT

Increases in drug-resistant pathogens are becoming a serious detriment to human health. To combat pathogen infections, a new series of amphiphilic coumarin derivatives were designed and synthesized as antimicrobial agents with membrane-targeting action. We herein report a lead compound, 25, that displayed potent antibacterial activity against Gram-positive bacteria, including MRSA. Compound 25 exhibited weak hemolytic activity and low toxicity to mammalian cells and can kill Gram-positive bacteria quickly (within 0.5 h) by directly disrupting the bacterial cell membranes. Additionally, compound 25 demonstrated excellent efficacy in a murine corneal infection caused by Staphylococcus aureus. These results suggest that 25 has great potential to be a potent antimicrobial agent for treating drug-resistant Gram-positive bacterial infections.


Subject(s)
Anti-Infective Agents , Coumarins , Animals , Anti-Bacterial Agents/pharmacology , Coumarins/pharmacology , Gram-Positive Bacteria , Humans , Mice , Microbial Sensitivity Tests , Staphylococcus aureus
16.
Eur J Med Chem ; 226: 113813, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34520955

ABSTRACT

The emergence of bacterial multidrug resistance and the lack of new antimicrobial agents urgently demand the discovery and development of novel antibacterials that avoid bacterial resistance. Antimicrobial peptidomimetics represent a promising approach for overcoming antibiotic resistance. Herein we report the synthesis and evaluation of indole-based amphiphilic antimicrobial peptidomimetics, bearing hydrophobic side chains and hydrophilic cationic moieties. Among these derivatives, compound 28 demonstrated potent antimicrobial activity against Gram-positive bacteria, low hemolytic activity and low toxicity towards mammalian cells, as well as good stability in salt conditions. Moreover, compound 28 showed the rapid killing of bacteria via membrane-targeting action without developing bacterial resistance. More importantly, compound 28 displayed high antimicrobial potency against Gram-positive bacteria in a murine model of bacterial keratitis, and was found to be more efficient than vancomycin. Thus, compound 28 had great potential as a promising lead compound for the treatment of Gram-positive bacterial infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gram-Positive Bacteria/drug effects , Indoles/pharmacology , Peptidomimetics/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Indoles/chemistry , Microbial Sensitivity Tests , Molecular Structure , Peptidomimetics/chemical synthesis , Peptidomimetics/chemistry , Structure-Activity Relationship
17.
Chin Med ; 16(1): 116, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34758851

ABSTRACT

BACKGROUND: Arsenic (As3+) is a carcinogen with considerable environmental and occupational relevancy. Its mechanism of action and methods of prevention remain to be investigated. Previous studies have demonstrated that ROS is responsible for As3+-induced cell transformation, which is considered as the first stage of As3+ carcinogenesis. The NF-E2 p45-related factor-2 (Nrf2) signaling pathway regulates the cellular antioxidant response, and activation of Nrf2 has recently been shown to limit oxidative damage following exposure to As3+ METHODS AND RESULTS: In this study, molecular docking was used to virtually screen natural antioxidant chemical databases and identify molecules that interact with the ligand-binding site of Keap1 (PDB code 4L7B). The cell-based assays and molecular docking findings revealed that curcumin has the best inhibitory activity against Keap1-4L7B. Co-immunoprecipitation (Co-IP) results indicated that curcumin is a potent Keap1 Kelch domain-dependent Nrf2 activator that stabilizes Nrf2 by hindering its ubiquitination. The increased activation of Nrf2 and its target antioxidant genes by curcumin could significantly decrease As3+-generated ROS. Moreover, curcumin induced autophagy in As3+-treated BEAS-2B via inducing autophagy by the formation of a p62/LC-3 complex and increasing autophagic flux by promoting transcription factor EB (TFEB) and lysosome-associated membrane protein 1 (LAMP1) expression. Knockdown of Nrf2 abolished curcumin-induced autophagy and downregulated ROS. Further studies showed that inhibition of autophagosome and lysosome fusion with bafilomycin a1 (BafA1) could block curcumin and prevented As3+-induced cell transformation. These results demonstrated that curcumin prevents As3+-induced cell transformation by inducing autophagy via the activation of the Nrf2 signaling pathway in BEAS-2B cells. However, overexpression of Keap-1 showed a constitutively high level of Nrf2 in As3+-transformed BEAS-2B cells (AsT) is Keap1-independent regulation. Overexpression of Nrf2 in AsT demonstrated that curcumin increased ROS levels and induced cell apoptosis via the downregulation of Nrf2. Further studies showed that curcumin decreased the Nrf2 level in AsT by activating GSK-3ß to inhibit the activation of PI3K/AKT. Co-IP assay results showed that curcumin promoted the interaction of Nrf2 with the GSK-3ß/ß-TrCP axis and ubiquitin. Moreover, the inhibition of GSK-3ß reversed Nrf2 expression in curcumin-treated AsT, indicating that the decrease in Nrf2 is due to activation of the GSK-3ß/ß-TrCP ubiquitination pathway. Furthermore, in vitro and in vivo results showed that curcumin induced cell apoptosis, and had anti-angiogenesis and anti-tumorigenesis effects as a result of activating the GSK-3ß/ß-TrCP ubiquitination pathway and subsequent decrease in Nrf2. CONCLUSIONS: Taken together, in the first stage, curcumin activated Nrf2, decreased ROS, and induced autophagy in normal cells to prevent As3+-induced cell transformation. In the second stage, curcumin promoted ROS and apoptosis and inhibited angiogenesis via inhibition of constitutive expression of Nrf2 in AsT to prevent tumorigenesis. Our results suggest that antioxidant natural compounds such as curcumin can be evaluated as potential candidates for complementary therapies in the treatment of As3+-induced carcinogenesis.

18.
Eur J Med Chem ; 202: 112596, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32659547

ABSTRACT

New antimicrobial agents are urgently needed to overcome drug-resistant bacterial infections. Here we describe the design, synthesis and evaluation of a new class of amphiphilic sofalcone compounds as antimicrobial peptidomimetics. The most promising compound 14, bearing two arginine residues, showed poor hemolytic activity, low cytotoxicity, and excellent antimicrobial activity against Gram-positive bacteria, including MRSA. Compound 14, had good stability in various salt conditions, killed bacteria rapidly by directly disrupting bacterial cell membranes and was slow at developing bacterial resistance. Additionally, compound 14 exhibited effective in vivo efficacy in the murine model of bacterial keratitis caused by Staphylococcus aureus ATCC29213. Our studies suggested that compound 14 possessed promising potential to be used as a novel antimicrobial agent to combat drug-resistant Gram-positive bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Chalcones/pharmacology , Drug Design , Staphylococcus aureus/drug effects , Surface-Active Agents/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Cell Line , Cell Survival/drug effects , Chalcones/chemical synthesis , Chalcones/chemistry , Dose-Response Relationship, Drug , Humans , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Surface-Active Agents/chemical synthesis , Surface-Active Agents/chemistry
19.
Biochim Biophys Acta Biomembr ; 1862(9): 183353, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32407778

ABSTRACT

Semisynthetic γ-mangostin derivative LS02 is a novel cationic amphiphilic peptidomimetic antimicrobial agent containing a hydrophobic scaffold and three hydrophilic and positively charged residues of arginine. LS02 showed low in vitro toxicity, potent activities against Gram-positive bacteria including MRSA (MIC = 1.56-6.25 µg/mL) and avoidance of drug resistance. The mode of action studies indicated that LS02 killed bacteria by disrupting bacterial cell membranes. LS02 not only exhibited good water solubility, low hemolytic activity and cell cytotoxicity, but also displayed excellent in vitro and in vivo antibacterial activity, indicating its great potential of being a lead compound as a novel membrane-active antibacterial agent capable of combating bacterial resistance.


Subject(s)
Anti-Infective Agents , Methicillin-Resistant Staphylococcus aureus/growth & development , Peptidomimetics , Xanthones , Animals , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Female , Humans , MCF-7 Cells , Mice , Peptidomimetics/chemical synthesis , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Rabbits , Xanthones/chemical synthesis , Xanthones/chemistry , Xanthones/pharmacology
20.
J Med Chem ; 63(17): 9284-9299, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32787074

ABSTRACT

The development of new antimicrobial agents capable of curing drug-resistant bacteria-induced infections is becoming a major challenge to the global healthcare system. To develop antimicrobials with new molecular entities, a series of novel carbazole-based compounds were designed and synthesized by biomimicking the structural properties and biological function of antimicrobial peptides. Compound 29 was selected as a lead compound from the structure-activity relationship analyses and biological activity evaluation. Compound 29 showed excellent antimicrobial activity against Gram-positive bacteria (MICs = 0.78-1.56 µg/mL), poor hemolytic activity (HC50 > 200 µg/mL), and low cytotoxicity to mammalian cells. Compound 29 had fast bactericidal properties and effectively prevented bacterial resistance in laboratory simulations. Antibacterial mechanism studies revealed that compound 29 directly destroyed bacterial cell membranes, leading to bacterial deaths. Importantly, compound 29 displayed an excellent efficacy in a murine bacterial keratitis model caused by Staphylococcus aureus ATCC29213.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Carbazoles/chemistry , Carbazoles/pharmacology , Drug Design , Gram-Positive Bacteria/drug effects , Hydrophobic and Hydrophilic Interactions , Animals , Gram-Positive Bacteria/physiology , Kinetics , Mice
SELECTION OF CITATIONS
SEARCH DETAIL