1
|
Inci TG, Ugurel E, Orlenco M, Akar S, Atlı R, Danis O, Turgut-Balik D. Cloning, Expression, Purification and Biological Activity Analysis of Recombinant Helicobacter pylori FabI as a Drug Target. Mol Biotechnol 2025:10.1007/s12033-025-01411-x. [PMID: 40016568 DOI: 10.1007/s12033-025-01411-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
Abstract
Helicobacter pylori (H. pylori) is an infectious agent colonized in gastric epithelium and leads to serious diseases such as ulcers and gastric carcinoma. H. pylori infection requires rapid and effective treatment options however existing therapies gradually diminish in efficacy due to the development of resistance. Type II fatty acid synthesis (FAS-II) pathway is a potent target for drug discovery studies because of its absence in humans and vital necessity for bacteria. In the last step of the synthesis, trans-2-enoyl-ACP is reduced to acyl-ACP with cofactor of NADH by enoyl-ACP reductase, FabI. In this study, recombinant HpFabI was successfully produced using an aLICator ligation-independent cloning and expression vector system for the first time. HpFabI gene was cloned, and then expressed, and the protein was purified in high yield. Recombinant HpFabI with a molecular mass of ~ 30 kDa was confirmed with Western Blot analysis and its concentration was determined in the range of 1.406-3.9495 mg/ml by Bradford Assay. The enzyme-specific activity of HpFabI was determined as 1.5871 nmol min-1 μg-1 by using NADH and crotonoyl-CoA as cofactor and substrate, respectively. HpFabI was produced in high yield to facilitate future inhibition studies including high throughput screening studies for FabI inhibition to contribute novel drug development studies fighting against H. pylori infection.
Collapse
Affiliation(s)
- Tugba Gul Inci
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, Davutpasa Campus, Esenler, 34220, Istanbul, Turkey
| | - Erennur Ugurel
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, Davutpasa Campus, Esenler, 34220, Istanbul, Turkey
| | - Maria Orlenco
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, Davutpasa Campus, Esenler, 34220, Istanbul, Turkey
| | - Selcan Akar
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, Davutpasa Campus, Esenler, 34220, Istanbul, Turkey
| | - Recepcan Atlı
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, Davutpasa Campus, Esenler, 34220, Istanbul, Turkey
| | - Ozkan Danis
- Department of Chemistry, Faculty of Science, Marmara University, Istanbul, Turkey
| | - Dilek Turgut-Balik
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, Davutpasa Campus, Esenler, 34220, Istanbul, Turkey.
| |
Collapse
|
2
|
Wang L, Liu X, Chen W, Sun Z. Non-targeted metabonomics reveals the effect of linalyl alcohol on Brochothrix thermophile and its potential application. Food Res Int 2025; 201:115549. [PMID: 39849689 DOI: 10.1016/j.foodres.2024.115549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/12/2024] [Accepted: 12/28/2024] [Indexed: 01/25/2025]
Abstract
Brochothrix thermophcta (B. thermophcta) is a pathogenic microorganism associated with food contamination. Linalyl alcohol, owing to its broad spectrum and exceptional antibacterial properties, is regarded as a potent natural antimicrobial agent. This is to elucidate the cellular-level mechanism of linalyl alcohol on B. thermophacta and investigate, for the first time, its regulatory effect on the metabolic pathway of B. thermophacta through metabonomics analysis. The results demonstrated that treatment with linalyl alcohol led to a reduction in bacterial metabolic capacity, while simultaneously promoting an increase in membrane fluidity through damage to the bacterial cell membrane. A total of 201 differential metabolites were identified at the metabolic level, with 50 showing significant up-regulation and 151 displaying significant down-regulation. The differential metabolites primarily participate in the tRNA cycle, amino acid metabolism, nucleotide metabolism, and aminoacyl-tRNA biosynthesis, with a particular emphasis on the significant impairment of amino acid metabolism. The application results demonstrated that linalyl alcohol exhibited a significant antibacterial effect on B. thermosphacta, as evidenced by the negligible changes observed in the color, smell, and tissue state of pork even after 8 days of treatment. In summary, linalyl alcohol exhibits multi-target and multi-pathway inhibition against B. thermosphacta, leading to disruption of cell morphology and metabolic processes. These findings provide a novel theoretical foundation for understanding the inhibitory mechanism of linalyl alcohol on B. thermosphacta, highlighting its potential as an effective alternative to food additives in the preservation industry of livestock products.
Collapse
Affiliation(s)
- Longteng Wang
- College of Food Sciences & Engineering, Hainan University, 58 People Road, Haikou 570228, China.
| | - Xing Liu
- College of Food Sciences & Engineering, Hainan University, 58 People Road, Haikou 570228, China.
| | - Wenxue Chen
- College of Food Sciences & Engineering, Hainan University, 58 People Road, Haikou 570228, China.
| | - Zhichang Sun
- College of Food Sciences & Engineering, Hainan University, 58 People Road, Haikou 570228, China.
| |
Collapse
|
3
|
Hernández-Vázquez E, Ramírez-Trinidad Á, Tovar-Román CE, Rivera Chávez JA, Huerta-Salazar E. N-acyl-4-arylaminopiperidines: Design and synthesis of a potential antimicrobial scaffold. Bioorg Med Chem Lett 2024; 112:129936. [PMID: 39214507 DOI: 10.1016/j.bmcl.2024.129936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/01/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
We report a concise synthesis of N-acylated piperidines through a Knoevenagel-Doebner condensation/amide construction/ amination sequence. The design of the piperidines considered the pharmacophoric features found in previously reported inhibitors of FabI, an enzyme implicated in bacterial fatty acid biosynthesis. After the microbiological evaluation at 50 μM, the analogs displayed moderate activity against some pathogens from the ESKAPE group, reaching up to 42 % of growth inhibition for MRSA, 54 % for K. pneumoniae, and 37 % for P. aeruginosa (multiresistant strains). Docking studies demonstrate that almost all of them docked satisfactorily into the catalytic domain of S. aureus FabI, maintaining a similar pose as other reported inhibitors. The results shown herein propose the N-acyl-4-arylaminopiperidines as the basis for the development of more active candidates.
Collapse
Affiliation(s)
- Eduardo Hernández-Vázquez
- Departmento de Química Orgánica. Instituto de Química, Universidad Nacional Autónoma de México, Mexico.
| | - Ángel Ramírez-Trinidad
- Departmento de Química Orgánica. Instituto de Química, Universidad Nacional Autónoma de México, Mexico
| | - César E Tovar-Román
- Departmento de Química Orgánica. Instituto de Química, Universidad Nacional Autónoma de México, Mexico
| | - José A Rivera Chávez
- Departmento de Productos Naturales, Instituto de Química, Universidad Nacional Autónoma de México, Mexico
| | - Elizabeth Huerta-Salazar
- Departmento de Química Orgánica. Instituto de Química, Universidad Nacional Autónoma de México, Mexico
| |
Collapse
|
4
|
Zhang L, Yang J, Xu X, Zhang J, Qiu Z, Ju Y, Luo B, Liu Y, Gou X, Sui J, Chen B, Wang Y, Tao T, He L, Yang T, Luo Y. Discovery and Optimization of Novel SaFabI Inhibitors as Specific Therapeutic Agents for MRSA Infection. J Med Chem 2024; 67:10096-10134. [PMID: 38845361 DOI: 10.1021/acs.jmedchem.4c00320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
As the rate-limiting enzyme in fatty acid biosynthesis, Staphylococcus aureus enoyl-acyl carrier protein reductase (SaFabI) emerges as a compelling target for combating methicillin-resistant S. aureus (MRSA) infections. Herein, compound 1, featuring a 4-(1H-benzo[d]imidazol-2-yl)pyrrolidin-2-one scaffold, was identified as a potent SaFabI inhibitor (IC50 = 976.8 nM) from an in-house library. Subsequent optimization yielded compound n31, with improved inhibitory efficacy on enzymatic activity (IC50 = 174.2 nM) and selective potency against S. aureus (MIC = 1-2 μg/mL). Mechanistically, n31 directly inhibited SaFabI in cellular contexts. Moreover, n31 exhibited favorable safety and pharmacokinetic profiles, and dose-dependently treated MRSA-induced skin infections, outperforming the approved drug, linezolid. The chiral separation of n31 resulted in (S)-n31, with superior activities (IC50 = 94.0 nM, MIC = 0.25-1 μg/mL) and in vivo therapeutic efficacy. In brief, our research proposes (S)-n31 as a promising candidate for SaFabI-targeted therapy, offering specific anti-S. aureus efficacy and potential for further development.
Collapse
Affiliation(s)
- Laiying Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaxing Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Xu
- Editorial Office of Chinese Journal of Medical Genetics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Jiangnan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiqiang Qiu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Ju
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Baozhu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xupeng Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Sui
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Baoyi Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanmei Wang
- Institute of traditional Chinese medicine, Sichuan College of Traditional Chinese Medicine, The Second Hospital of Traditional Chinese Medicine in Sichuan Province, Chengdu 610041, China
| | - Tao Tao
- Institute of traditional Chinese medicine, Sichuan College of Traditional Chinese Medicine, The Second Hospital of Traditional Chinese Medicine in Sichuan Province, Chengdu 610041, China
| | - Lei He
- Institute of traditional Chinese medicine, Sichuan College of Traditional Chinese Medicine, The Second Hospital of Traditional Chinese Medicine in Sichuan Province, Chengdu 610041, China
| | - Tao Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Youfu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Rutherford J, Avad K, Dureja C, Norseeda K, GC B, Wu C, Sun D, Hevener KE, Hurdle JG. Evaluation of Fusobacterium nucleatum Enoyl-ACP Reductase (FabK) as a Narrow-Spectrum Drug Target. ACS Infect Dis 2024; 10:1612-1623. [PMID: 38597503 PMCID: PMC11091888 DOI: 10.1021/acsinfecdis.3c00710] [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: 12/20/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/11/2024]
Abstract
Fusobacterium nucleatum, a pathobiont inhabiting the oral cavity, contributes to opportunistic diseases, such as periodontal diseases and gastrointestinal cancers, which involve microbiota imbalance. Broad-spectrum antimicrobial agents, while effective against F. nucleatum infections, can exacerbate dysbiosis. This necessitates the discovery of more targeted narrow-spectrum antimicrobial agents. We therefore investigated the potential for the fusobacterial enoyl-ACP reductase II (ENR II) isoenzyme FnFabK (C4N14_ 04250) as a narrow-spectrum drug target. ENRs catalyze the rate-limiting step in the bacterial fatty acid synthesis pathway. Bioinformatics revealed that of the four distinct bacterial ENR isoforms, F. nucleatum specifically encodes FnFabK. Genetic studies revealed that fabK was indispensable for F. nucleatum growth, as the gene could not be deleted, and silencing of its mRNA inhibited growth under the test conditions. Remarkably, exogenous fatty acids failed to rescue growth inhibition caused by the silencing of fabK. Screening of synthetic phenylimidazole analogues of a known FabK inhibitor identified an inhibitor (i.e., 681) of FnFabK enzymatic activity and F. nucleatum growth, with an IC50 of 2.1 μM (1.0 μg/mL) and a MIC of 0.4 μg/mL, respectively. Exogenous fatty acids did not attenuate the activity of 681 against F. nucleatum. Furthermore, FnFabK was confirmed as the intracellular target of 681 based on the overexpression of FnFabK shifting MICs and 681-resistant mutants having amino acid substitutions in FnFabK or mutations in other genetic loci affecting fatty acid biosynthesis. 681 had minimal activity against a range of commensal flora, and it was less active against streptococci in physiologic fatty acids. Taken together, FnFabK is an essential enzyme that is amenable to drug targeting for the discovery and development of narrow-spectrum antimicrobial agents.
Collapse
Affiliation(s)
- Jacob
T. Rutherford
- Center
for Infectious and Inflammatory Diseases, Institute of Biosciences
and Technology, Department of Translational Medical Sciences, Texas A&M Health Science Center, Houston, Texas 77030, United States
| | - Kristiana Avad
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Chetna Dureja
- Center
for Infectious and Inflammatory Diseases, Institute of Biosciences
and Technology, Department of Translational Medical Sciences, Texas A&M Health Science Center, Houston, Texas 77030, United States
| | - Krissada Norseeda
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii 96720, United States
| | - Bibek GC
- Department
of Microbiology & Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Chenggang Wu
- Department
of Microbiology & Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Dianqing Sun
- Department
of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii 96720, United States
| | - Kirk E. Hevener
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Julian G. Hurdle
- Center
for Infectious and Inflammatory Diseases, Institute of Biosciences
and Technology, Department of Translational Medical Sciences, Texas A&M Health Science Center, Houston, Texas 77030, United States
| |
Collapse
|
6
|
Khaled M, Ouache R, Pale P, Harkat H. Phytochemical Profiles and Biological Activities of Frankenia Species: A Review. Molecules 2024; 29:980. [PMID: 38474492 DOI: 10.3390/molecules29050980] [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: 01/11/2024] [Revised: 02/12/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
The relatively small Frankeniaceae family is represented by halophyte plants, growing in arid and semi-arid climates in saline, alkaline or calcareous soils. Due to their living conditions, they usually produce a large diversity of compounds, which often exhibit bioactivities. Some species of this genus have long been used as traditional herbal medicines to treat dysentery, diarrhea, gonorrhea, vaginal leucorrhea, respiratory diseases and wounds. To date, several studies on either phytochemical or pharmacological aspects, or both, have revealed that this genus is a rich source of diverse and novel bioactive chemicals, including phenolics, flavonoids, alkaloids and fatty acids. This review describes all the reported chemical profiles of Frankenia species, as well as the corresponding biological properties, when available. The aim of this review is to show the potential of these plants for various applications, especially therapeutic ones.
Collapse
Affiliation(s)
- Meyada Khaled
- Department of Pharmacy, Faculty of Medicine, Batna 2 University, Batna 05000, Algeria
| | - Rachid Ouache
- Laboratory of Physio-Toxicology, Cellular and Molecular Pathology-Biomolecules (LPTPCMB), Batna 2 University, Batna 05000, Algeria
| | - Patrick Pale
- Laboratory of Organic Synthesis & Catalysis, Institute of Chemistry (UMR-CNRS 7177), University of Strasbourg, 67000 Strasbourg, France
| | - Hassina Harkat
- Department of Pharmacy, Faculty of Medicine, Batna 2 University, Batna 05000, Algeria
- Laboratory of Physio-Toxicology, Cellular and Molecular Pathology-Biomolecules (LPTPCMB), Batna 2 University, Batna 05000, Algeria
| |
Collapse
|
7
|
Xu Z, Tian P. Rethinking Biosynthesis of Aclacinomycin A. Molecules 2023; 28:molecules28062761. [PMID: 36985733 PMCID: PMC10054333 DOI: 10.3390/molecules28062761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/22/2023] Open
Abstract
Aclacinomycin A (ACM-A) is an anthracycline antitumor agent widely used in clinical practice. The current industrial production of ACM-A relies primarily on chemical synthesis and microbial fermentation. However, chemical synthesis involves multiple reactions which give rise to high production costs and environmental pollution. Microbial fermentation is a sustainable strategy, yet the current fermentation yield is too low to satisfy market demand. Hence, strain improvement is highly desirable, and tremendous endeavors have been made to decipher biosynthesis pathways and modify key enzymes. In this review, we comprehensively describe the reported biosynthesis pathways, key enzymes, and, especially, catalytic mechanisms. In addition, we come up with strategies to uncover unknown enzymes and improve the activities of rate-limiting enzymes. Overall, this review aims to provide valuable insights for complete biosynthesis of ACM-A.
Collapse
|
8
|
Yan W, Zheng Y, Dou C, Zhang G, Arnaout T, Cheng W. The pathogenic mechanism of Mycobacterium tuberculosis: implication for new drug development. MOLECULAR BIOMEDICINE 2022; 3:48. [PMID: 36547804 PMCID: PMC9780415 DOI: 10.1186/s43556-022-00106-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is a tenacious pathogen that has latently infected one third of the world's population. However, conventional TB treatment regimens are no longer sufficient to tackle the growing threat of drug resistance, stimulating the development of innovative anti-tuberculosis agents, with special emphasis on new protein targets. The Mtb genome encodes ~4000 predicted proteins, among which many enzymes participate in various cellular metabolisms. For example, more than 200 proteins are involved in fatty acid biosynthesis, which assists in the construction of the cell envelope, and is closely related to the pathogenesis and resistance of mycobacteria. Here we review several essential enzymes responsible for fatty acid and nucleotide biosynthesis, cellular metabolism of lipids or amino acids, energy utilization, and metal uptake. These include InhA, MmpL3, MmaA4, PcaA, CmaA1, CmaA2, isocitrate lyases (ICLs), pantothenate synthase (PS), Lysine-ε amino transferase (LAT), LeuD, IdeR, KatG, Rv1098c, and PyrG. In addition, we summarize the role of the transcriptional regulator PhoP which may regulate the expression of more than 110 genes, and the essential biosynthesis enzyme glutamine synthetase (GlnA1). All these enzymes are either validated drug targets or promising target candidates, with drugs targeting ICLs and LAT expected to solve the problem of persistent TB infection. To better understand how anti-tuberculosis drugs act on these proteins, their structures and the structure-based drug/inhibitor designs are discussed. Overall, this investigation should provide guidance and support for current and future pharmaceutical development efforts against mycobacterial pathogenesis.
Collapse
Affiliation(s)
- Weizhu Yan
- grid.412901.f0000 0004 1770 1022Division of Respiratory and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041 China
| | - Yanhui Zheng
- grid.412901.f0000 0004 1770 1022Division of Respiratory and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041 China
| | - Chao Dou
- grid.412901.f0000 0004 1770 1022Division of Respiratory and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041 China
| | - Guixiang Zhang
- grid.13291.380000 0001 0807 1581Division of Gastrointestinal Surgery, Department of General Surgery and Gastric Cancer center, West China Hospital, Sichuan University, No. 37. Guo Xue Xiang, Chengdu, 610041 China
| | - Toufic Arnaout
- Kappa Crystals Ltd., Dublin, Ireland ,MSD Dunboyne BioNX, Co. Meath, Ireland
| | - Wei Cheng
- grid.412901.f0000 0004 1770 1022Division of Respiratory and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041 China
| |
Collapse
|
9
|
Qiu D, Ke M, Zhang Q, Zhang F, Lu T, Sun L, Qian H. Response of microbial antibiotic resistance to pesticides: An emerging health threat. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 850:158057. [PMID: 35977623 DOI: 10.1016/j.scitotenv.2022.158057] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
The spread of microbial antibiotic resistance has seriously threatened public health globally. Non-antibiotic stressors have significantly contributed to the evolution of bacterial antibiotic resistance. Although numerous studies have been conducted on the potential risk of pesticide pollution for bacterial antibiotic resistance, a systematic review of these concerns is still lacking. In the present study, we elaborate the mechanism underlying the effects of pesticides on bacterial antibiotic resistance acquisition as well as the propagation of antimicrobial resistance. Pesticide stress enhanced the acquisition of antibiotic resistance in bacteria via various mechanisms, including the activation of efflux pumps, inhibition of outer membrane pores for resistance to antibiotics, and gene mutation induction. Horizontal gene transfer is a major mechanism whereby pesticides influence the transmission of antibiotic resistance genes (ARGs) in bacteria. Pesticides promoted the conjugation transfer of ARGs by increasing cell membrane permeability and increased the proportion of bacterial mobile gene elements, which facilitate the spread of ARGs. This review can improve our understanding regarding the pesticide-induced generation and spread of ARGs and antibiotic resistant bacteria. Moreover, it can be applied to reduce the ecological risks of ARGs in the future.
Collapse
Affiliation(s)
- Danyan Qiu
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Mingjing Ke
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Qi Zhang
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Fan Zhang
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Tao Lu
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Liwei Sun
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Haifeng Qian
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China.
| |
Collapse
|
10
|
Transcriptome Analyses of Prophage in Mediating Persistent Methicillin-Resistant Staphylococcus aureus Endovascular Infection. Genes (Basel) 2022; 13:genes13091527. [PMID: 36140695 PMCID: PMC9498598 DOI: 10.3390/genes13091527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 12/24/2022] Open
Abstract
Persistent methicillin-resistant Staphylococcus aureus (MRSA) endovascular infections represent a significant subset of S. aureus infections and correlate with exceptionally high mortality. We have recently demonstrated that the lysogenization of prophage ϕSA169 from a clinical persistent MRSA bacteremia isolate (300-169) into a clinical resolving bacteremia MRSA isolate (301-188) resulted in the acquisition of well-defined in vitro and in vivo phenotypic and genotypic profiles related to persistent outcome. However, the underlying mechanism(s) of this impact is unknown. In the current study, we explored the genetic mechanism that may contribute to the ϕSA169-correlated persistence using RNA sequencing. Transcriptomic analyses revealed that the most significant impacts of ϕSA169 were: (i) the enhancement of fatty acid biosynthesis and purine and pyrimidine metabolic pathways; (ii) the repression of galactose metabolism and phosphotransferase system (PTS); and (iii) the down-regulation of the mutual prophage genes in both 300-169 and 301-188 strains. In addition, the influence of different genetic backgrounds between 300-169 and 301-188 might also be involved in the persistent outcome. These findings may provide targets for future studies on the persistence of MRSA.
Collapse
|
11
|
Parker E, Cain BN, Hajian B, Ulrich RJ, Geddes EJ, Barkho S, Lee HY, Williams JD, Raynor M, Caridha D, Zaino A, Shekhar M, Muñoz KA, Rzasa KM, Temple ER, Hunt D, Jin X, Vuong C, Pannone K, Kelly AM, Mulligan MP, Lee KK, Lau GW, Hung DT, Hergenrother PJ. An Iterative Approach Guides Discovery of the FabI Inhibitor Fabimycin, a Late-Stage Antibiotic Candidate with In Vivo Efficacy against Drug-Resistant Gram-Negative Infections. ACS CENTRAL SCIENCE 2022; 8:1145-1158. [PMID: 36032774 PMCID: PMC9413440 DOI: 10.1021/acscentsci.2c00598] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Indexed: 05/13/2023]
Abstract
Genomic studies and experiments with permeability-deficient strains have revealed a variety of biological targets that can be engaged to kill Gram-negative bacteria. However, the formidable outer membrane and promiscuous efflux pumps of these pathogens prevent many candidate antibiotics from reaching these targets. One such promising target is the enzyme FabI, which catalyzes the rate-determining step in bacterial fatty acid biosynthesis. Notably, FabI inhibitors have advanced to clinical trials for Staphylococcus aureus infections but not for infections caused by Gram-negative bacteria. Here, we synthesize a suite of FabI inhibitors whose structures fit permeation rules for Gram-negative bacteria and leverage activity against a challenging panel of Gram-negative clinical isolates as a filter for advancement. The compound to emerge, called fabimycin, has impressive activity against >200 clinical isolates of Escherichia coli, Klebsiella pneumoniae, and Acinetobacter baumannii, and does not kill commensal bacteria. X-ray structures of fabimycin in complex with FabI provide molecular insights into the inhibition. Fabimycin demonstrates activity in multiple mouse models of infection caused by Gram-negative bacteria, including a challenging urinary tract infection model. Fabimycin has translational promise, and its discovery provides additional evidence that antibiotics can be systematically modified to accumulate in Gram-negative bacteria and kill these problematic pathogens.
Collapse
Affiliation(s)
- Erica
N. Parker
- Department
of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Brett N. Cain
- Department
of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Behnoush Hajian
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Rebecca J. Ulrich
- Department
of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Emily J. Geddes
- Department
of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Sulyman Barkho
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Hyang Yeon Lee
- Department
of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - John D. Williams
- Walter
Reed Army Institute of Research, Silver Spring, Maryland 20910 United States
| | - Malik Raynor
- Walter
Reed Army Institute of Research, Silver Spring, Maryland 20910 United States
| | - Diana Caridha
- Walter
Reed Army Institute of Research, Silver Spring, Maryland 20910 United States
| | - Angela Zaino
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Mrinal Shekhar
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Kristen A. Muñoz
- Department
of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Kara M. Rzasa
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Emily R. Temple
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Diana Hunt
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for Computational and Integrative
Biology, Massachusetts General Hospital, Boston, Massachusetts 02115, United States
| | - Xiannu Jin
- Walter
Reed Army Institute of Research, Silver Spring, Maryland 20910 United States
| | - Chau Vuong
- Walter
Reed Army Institute of Research, Silver Spring, Maryland 20910 United States
| | - Kristina Pannone
- Walter
Reed Army Institute of Research, Silver Spring, Maryland 20910 United States
| | - Aya M. Kelly
- Department
of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Michael P. Mulligan
- Department
of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Katie K. Lee
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Gee W. Lau
- Department
of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Deborah T. Hung
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for Computational and Integrative
Biology, Massachusetts General Hospital, Boston, Massachusetts 02115, United States
| | - Paul J. Hergenrother
- Department
of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
12
|
Wang Z, Liu X, Duan Y, Huang Y. Nanoparticle-Hydrogel Systems Containing Platensimycin for Local Treatment of Methicillin-Resistant Staphylococcus aureus Infection. Mol Pharm 2021; 18:4099-4110. [PMID: 34554755 DOI: 10.1021/acs.molpharmaceut.1c00523] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Skin and soft tissue infections require effective and sustained topical administration. Platensimycin (PTM) is a natural drug lead that targets bacterial fatty acid synthases and has a great potential to treat infections caused by methicillin-resistant Staphylococcus aureus (MRSA). To facilitate the use of PTM against local MRSA infections, we prepared polyacrylamide hydrogels containing polyamidoamine (PAMAM)/PTM nanoparticles (NP-gel(PTM)) for the controlled release of PTM. NP-gel(PTM) can continuously inhibit the growth of MRSA and its biofilm formation in simulated drug flow models in vitro. In situ implantation of NP-gel(PTM) could treat MRSA-infected subcutaneous soft tissues without toxicity. For MRSA-infected skin wounds, NP-gel(PTM) not only showed strong anti-MRSA activity but also accelerated more wound healing than the widely used antibiotic mupirocin. Collectively, PTM is expected to be used in this safe and effective NP-gel delivery platform for the treatment of local infections, which might help to alleviate the current antibiotic resistance crisis.
Collapse
Affiliation(s)
- Zhe Wang
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, Hunan 410013, China
| | - Xingyun Liu
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, Hunan 410013, China
| | - Yanwen Duan
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, Hunan 410013, China.,Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug Discover, Changsha, Hunan 410011, China.,National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, Hunan 410011, China
| | - Yong Huang
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, Hunan 410013, China.,National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, Hunan 410011, China
| |
Collapse
|
13
|
Alsenani F, Tupally KR, Chua ET, Eltanahy E, Alsufyani H, Parekh HS, Schenk PM. Evaluation of microalgae and cyanobacteria as potential sources of antimicrobial compounds. Saudi Pharm J 2020; 28:1834-1841. [PMID: 33424272 PMCID: PMC7783216 DOI: 10.1016/j.jsps.2020.11.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 11/21/2020] [Indexed: 12/12/2022] Open
Abstract
Three microalgal extracts show antimicrobial activity against gram-positive bacteria. Different purification techniques were used to reveal the bioactive compounds. Linoleic acid, oleic acid, DHA and EPA inhibited the growth of gram-positive bacteria.
In recent decades, marine microorganisms have become known for their ability to produce a wide variety of secondary bioactive metabolites. Several compounds have been isolated from marine microorganisms for the development of novel bioactives for the food and pharmaceutical industries. In this study, a number of microalgae were evaluated for their antimicrobial activity against gram-positive and gram-negative bacteria, including food and plant pathogens, using various extraction techniques and antimicrobial assays. Disc diffusion and spot-on-lawn assays were conducted to confirm the antimicrobial activity. To measure the potency of the extracts, minimum inhibition concentrations (MIultCs) were measured. Three microalgae, namely Isochrysis galbana, Scenedesmus sp. NT8c, and Chlorella sp. FN1, showed strong inhibitory activity preferentially against gram-positive bacteria. These microalgal species were then selected for further purification and analysis, leading to compound identification. By using a mixture of different chromatography techniques gas chromatography–mass spectrometry (GC–MS) and high-performance liquid chromatography (HPLC) and ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS), we were able to separate and identify the dominant compounds that are responsible for the inhibitory activity. Additionally, nuclear magnetic resonance (NMR) was used to confirm the presence of these compounds. The dominant compounds that were identified and purified in the extracts are linoleic acid, oleic acid, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA). These compounds are the potential candidates that inhibit the growth of gram-positive bacteria. This indicates the potential use of microalgae and their antimicrobial compounds as biocontrol agents against food and plant pathogens.
Collapse
Affiliation(s)
- Faisal Alsenani
- Department of Pharmacognosy, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia.,School of Pharmacy, Pharmacy Australia Centre of Excellence (PACE), The University of Queensland, Brisbane, QLD 4072, Australia.,Algae Biotechnology Laboratory, School of Agriculture and Food Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Karnaker R Tupally
- School of Pharmacy, Pharmacy Australia Centre of Excellence (PACE), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Elvis T Chua
- Algae Biotechnology Laboratory, School of Agriculture and Food Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Eladl Eltanahy
- Phycology Laboratory, Botany Department, Faculty of Science, Mansoura University, Egypt
| | - Hamed Alsufyani
- Department of Biology, Faculty of Science and Arts, University of Jeddah, Khulais 21921, Saudi Arabia
| | - Harendra S Parekh
- School of Pharmacy, Pharmacy Australia Centre of Excellence (PACE), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Peer M Schenk
- Algae Biotechnology Laboratory, School of Agriculture and Food Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
14
|
Thitiananpakorn K, Aiba Y, Tan XE, Watanabe S, Kiga K, Sato'o Y, Boonsiri T, Li FY, Sasahara T, Taki Y, Azam AH, Zhang Y, Cui L. Association of mprF mutations with cross-resistance to daptomycin and vancomycin in methicillin-resistant Staphylococcus aureus (MRSA). Sci Rep 2020; 10:16107. [PMID: 32999359 PMCID: PMC7527455 DOI: 10.1038/s41598-020-73108-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
We first reported a phenomenon of cross-resistance to vancomycin (VCM) and daptomycin (DAP) in methicillin-resistant Staphylococcus aureus (MRSA) in 2006, but mechanisms underlying the cross-resistance remain incompletely understood. Here, we present a follow-up study aimed to investigate genetic determinants associated with the cross-resistance. Using 12 sets of paired DAP susceptible (DAPS) and DAP non-susceptible (DAPR) MRSA isolates from 12 patients who had DAP therapy, we (i) assessed susceptibility to DAP and VCM, (ii) compared whole-genome sequences, (iii) identified mutations associated with cross-resistance to DAP and VCM, and (iv) investigated the impact of altered gene expression and metabolic pathway relevant to the cross-resistance. We found that all 12 DAPR strains exhibiting cross-resistance to DAP and VCM carried mutations in mprF, while one DAPR strain with reduced susceptibility to only DAP carried a lacF mutation. On the other hand, among the 32 vancomycin-intermediate S. aureus (VISA) strains isolated from patients treated with VCM, five out of the 18 strains showing cross-resistance to DAP and VCM carried a mprF mutation, while 14 strains resistant to only VCM had no mprF mutation. Moreover, substitution of mprF in a DAPS strain with mutated mprF resulted in cross-resistance and vice versa. The elevated lysyl-phosphatidylglycerol (L-PG) production, increased positive bacterial surface charges and activated cell wall (CW) synthetic pathways were commonly found in both clinical isolates and laboratory-developed mutants that carry mprF mutations. We conclude that mprF mutation is responsible for the cross-resistance of MRSA to DAP and VCM, and treatment with DAP is more likely to select for mprF-mediated cross-resistance than is with VCM.
Collapse
Affiliation(s)
- Kanate Thitiananpakorn
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Yoshifumi Aiba
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Xin-Ee Tan
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Shinya Watanabe
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Kotaro Kiga
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Yusuke Sato'o
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Tanit Boonsiri
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Feng-Yu Li
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Teppei Sasahara
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Yusuke Taki
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Aa Haeruman Azam
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Yuancheng Zhang
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan
| | - Longzhu Cui
- Division of Bacteriology, Department of Infection and Immunity, Faculty of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-shi, Tochigi, 329-0498, Japan.
| |
Collapse
|
15
|
Rana P, Ghouse SM, Akunuri R, Madhavi YV, Chopra S, Nanduri S. FabI (enoyl acyl carrier protein reductase) - A potential broad spectrum therapeutic target and its inhibitors. Eur J Med Chem 2020; 208:112757. [PMID: 32883635 DOI: 10.1016/j.ejmech.2020.112757] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/30/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022]
Abstract
Development of new anti-bacterial agents acting upon underexploited targets and thus evading known mechanisms of resistance is the need of the hour. The highly conserved and distinct bacterial fatty acid biosynthesis pathway (FAS-II), presents a validated and yet relatively underexploited target for drug discovery. FabI and its isoforms (FabL, FabK, FabV and InhA) are essential enoyl-ACP reductases present in several microorganisms. In addition, the components of the FAS-II pathway are distinct from the multi-enzyme FAS-I complex found in mammals. Thus, inhibition of FabI and its isoforms is anticipated to result in broad-spectrum antibacterial activity. Several research groups from industry and academic laboratories have devoted significant efforts to develop effective FabI-targeting antibiotics, which are currently in various stages of clinical development for the treatment of multi-drug resistant bacterial infections. This review summarizes all the natural as well as synthetic inhibitors of gram-positive and gram-negative enoyl ACP reductases (FabI). The knowledge of the reported inhibitors can aid in the development of broad-spectrum antibacterials specifically targeting FabI enzymes from S. aureus, S. epidermidis, B. anthracis, B. cereus, E. coli, P. aeruginosa, P. falciparum and M. tuberculosis.
Collapse
Affiliation(s)
- Preeti Rana
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Shaik Mahammad Ghouse
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Ravikumar Akunuri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Y V Madhavi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Sidharth Chopra
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, 226 031, Uttar Pradesh, India.
| | - Srinivas Nanduri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India.
| |
Collapse
|
16
|
Chung PY. Novel targets of pentacyclic triterpenoids in Staphylococcus aureus: A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 73:152933. [PMID: 31103429 DOI: 10.1016/j.phymed.2019.152933] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Staphylococcus aureus is an important pathogen both in community-acquired and healthcare-associated infections, and has successfully evolved numerous strategies for resisting the action to practically all antibiotics. Resistance to methicillin is now widely described in the community setting (CMRSA), thus the development of new drugs or alternative therapies is urgently necessary. Plants and their secondary metabolites have been a major alternative source in providing structurally diverse bioactive compounds as potential therapeutic agents for the treatment of bacterial infections. One of the classes of natural secondary metabolites from plants with the most bioactive compounds are the triterpenoids, which comprises structurally diverse organic compounds. In nature, triterpenoids are often found as tetra- or penta-cyclic structures. AIM This review highlights the anti-staphylococcal activities of pentacyclic triterpenoids, particularly α-amyrin (AM), betulinic acid (BA) and betulinaldehyde (BE). These compounds are based on a 30-carbon skeleton comprising five six-membered rings (ursanes and lanostanes) or four six-membered rings and one five-membered ring (lupanes and hopanes). METHODS Electronic databases such as ScienceDirect, PubMed and Scopus were used to search scientific contributions until March 2018, using relevant keywords. Literature focusing on the antimicrobial and antibiofilms of effects of pentacyclic triterpenoids on S. aureus were identified and summarized. RESULTS Pentacyclic triterpenoids can be divided into three representative classes, namely ursane, lupane and oleananes. This class of compounds have been shown to exhibit analgesic, immunomodulatory, anti-inflammatory, anticancer, antioxidant, antifungal and antibacterial activities. In studies of the antimicrobial activities and targets of AM, BA and BE in sensitive and multidrug-resistant S. aureus, these compounds acted synergistically and have different targets from the conventional antibiotics. CONCLUSION The inhibitory mechanisms of S. aureus in novel targets and pathways should stimulate further researches to develop AM, BA and BE as therapeutic agents for infections caused by S. aureus. Continued efforts to identify and exploit synergistic combinations by the three compounds and peptidoglycan inhibitors, are also necessary as alternative treatment options for S. aureus infections.
Collapse
Affiliation(s)
- Pooi Yin Chung
- Department of Pathology, School of Medicine, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
17
|
Kronenberger T, de Oliveira Fernades P, Drumond Franco I, Poso A, Gonçalves Maltarollo V. Ligand- and Structure-Based Approaches of Escherichia coli FabI Inhibition by Triclosan Derivatives: From Chemical Similarity to Protein Dynamics Influence. ChemMedChem 2019; 14:1995-2004. [PMID: 31670463 PMCID: PMC6916556 DOI: 10.1002/cmdc.201900415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/03/2019] [Indexed: 12/20/2022]
Abstract
Enoyl-acyl carrier protein reductase (FabI) is the limiting step to complete the elongation cycle in type II fatty acid synthase (FAS) systems and is a relevant target for antibacterial drugs. E. coli FabI has been employed as a model to develop new inhibitors against FAS, especially triclosan and diphenyl ether derivatives. Chemical similarity models (CSM) were used to understand which features were relevant for FabI inhibition. Exhaustive screening of different CSM parameter combinations featured chemical groups, such as the hydroxy group, as relevant to distinguish between active/decoy compounds. Those chemical features can interact with the catalytic Tyr156. Further molecular dynamics simulation of FabI revealed the ionization state as a relevant for ligand stability. Also, our models point the balance between potency and the occupancy of the hydrophobic pocket. This work discusses the strengths and weak points of each technique, highlighting the importance of complementarity among approaches to elucidate EcFabI inhibitor's binding mode and offers insights for future drug discovery.
Collapse
Affiliation(s)
- Thales Kronenberger
- Department of Medical Oncology and Pneumology, Internal Medicine VIIIUniversity Hospital of TübingenOtfried-Müller-Strasse 1472076TübingenGermany
- School of PharmacyUniversity of Eastern Finland Faculty of Health SciencesKuopio70211Finland
| | - Philipe de Oliveira Fernades
- Departamento de Produtos Farmacêuticos, Faculdade de FarmáciaUniversidade Federal de Minas GeraisAv. Antônio Carlos6627Pampulha, Belo Horizonte, MG, 31270-901Brazil
- Departamento de Química, Instituto de Ciências ExatasUniversidade Federal de Minas GeraisAv. Antônio Carlos6627 –Pampulha, Belo Horizonte, MG, 31270-901Brazil
| | - Isabella Drumond Franco
- Departamento de Produtos Farmacêuticos, Faculdade de FarmáciaUniversidade Federal de Minas GeraisAv. Antônio Carlos6627Pampulha, Belo Horizonte, MG, 31270-901Brazil
| | - Antti Poso
- Department of Medical Oncology and Pneumology, Internal Medicine VIIIUniversity Hospital of TübingenOtfried-Müller-Strasse 1472076TübingenGermany
- School of PharmacyUniversity of Eastern Finland Faculty of Health SciencesKuopio70211Finland
| | - Vinícius Gonçalves Maltarollo
- Departamento de Produtos Farmacêuticos, Faculdade de FarmáciaUniversidade Federal de Minas GeraisAv. Antônio Carlos6627Pampulha, Belo Horizonte, MG, 31270-901Brazil
| |
Collapse
|
18
|
Andrews LD, Kane TR, Dozzo P, Haglund CM, Hilderbrandt DJ, Linsell MS, Machajewski T, McEnroe G, Serio AW, Wlasichuk KB, Neau DB, Pakhomova S, Waldrop GL, Sharp M, Pogliano J, Cirz RT, Cohen F. Optimization and Mechanistic Characterization of Pyridopyrimidine Inhibitors of Bacterial Biotin Carboxylase. J Med Chem 2019; 62:7489-7505. [PMID: 31306011 DOI: 10.1021/acs.jmedchem.9b00625] [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
A major challenge for new antibiotic discovery is predicting the physicochemical properties that enable small molecules to permeate Gram-negative bacterial membranes. We have applied physicochemical lessons from previous work to redesign and improve the antibacterial potency of pyridopyrimidine inhibitors of biotin carboxylase (BC) by up to 64-fold and 16-fold against Escherichia coli and Pseudomonas aeruginosa, respectively. Antibacterial and enzyme potency assessments in the presence of an outer membrane-permeabilizing agent or in efflux-compromised strains indicate that penetration and efflux properties of many redesigned BC inhibitors could be improved to various extents. Spontaneous resistance to the improved pyridopyrimidine inhibitors in P. aeruginosa occurs at very low frequencies between 10-8 and 10-9. However, resistant isolates had alarmingly high minimum inhibitory concentration shifts (16- to >128-fold) compared to the parent strain. Whole-genome sequencing of resistant isolates revealed that either BC target mutations or efflux pump overexpression can lead to the development of high-level resistance.
Collapse
Affiliation(s)
- Logan D Andrews
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - Timothy R Kane
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - Paola Dozzo
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - Cat M Haglund
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - Darin J Hilderbrandt
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - Martin S Linsell
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - Timothy Machajewski
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - Glen McEnroe
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - Alisa W Serio
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - Kenneth B Wlasichuk
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - David B Neau
- Northeastern Collaborative Access Team , Argonne National Laboratory , Argonne , Illinois 60439 , United States
| | - Svetlana Pakhomova
- Department of Biological Sciences , Louisiana State University , Baton Rouge , Louisiana 70803 , United States
| | - Grover L Waldrop
- Department of Biological Sciences , Louisiana State University , Baton Rouge , Louisiana 70803 , United States
| | - Marc Sharp
- Linnaeus Bioscience Inc. , 3210 Merryfield Row , San Diego , California 92121 , United States
| | - Joe Pogliano
- Linnaeus Bioscience Inc. , 3210 Merryfield Row , San Diego , California 92121 , United States.,University of California, San Diego , 9500 Gilman Drive , La Jolla, San Diego , California 92093 , United States
| | - Ryan T Cirz
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| | - Frederick Cohen
- Former employee of Achaogen Inc. , 1 Tower Place, Suite 400 , South San Francisco , California 94080 , United States
| |
Collapse
|
19
|
Kumar V, Sharma A, Pratap S, Kumar P. Biochemical and biophysical characterization of 1,4-naphthoquinone as a dual inhibitor of two key enzymes of type II fatty acid biosynthesis from Moraxella catarrhalis. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1866:1131-1142. [PMID: 30282611 DOI: 10.1016/j.bbapap.2018.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 02/03/2023]
Abstract
The fatty acid biosynthesis (FAS II) is a vital process in bacteria and regarded as an attractive pathway for the development of potential antimicrobial agents. In this study, we report 1,4-naphthoquinone (NPQ) as a dual inhibitor of two key enzymes of FAS II pathway, namely FabD (Malonyl-CoA:ACP transacylase) and FabZ (β-hydroxyacyl-ACP dehydratase). Mode of inhibition of NPQ was found to be non-competitive for both enzymes with IC50 of 26.67 μΜ and 23.18 μΜ against McFabZ and McFabD respectively. Conformational changes in secondary and tertiary structures marked by the loss of helical contents were observed in both enzymes upon NPQ binding. The fluorescence quenching was found to be static with a stable ground state complex formation. ITC based studies have shown that NPQ is binding to McFabZ with a stronger affinity (~1.5×) as compared to McFabD. Molecular docking studies have found that NPQ interacts with key residues of both McFabD (Ser209, Arg126, and Leu102) and McFabZ (His74 and Tyr112) enzymes. Both complexes have shown the structural stability during the 20 ns run of molecular dynamics based simulations. Altogether, the present study suggests that NPQ scaffold can be exploited as a multi-targeted inhibitor of FAS II pathway, and these biochemical and biophysical findings will further help in the development of potent antibacterial agents targeting FAS II pathway.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Biotechnology, Indian Institute of Technology Roorkee, India
| | - Anchal Sharma
- Department of Biotechnology, Indian Institute of Technology Roorkee, India
| | - Shivendra Pratap
- Department of Biotechnology, Indian Institute of Technology Roorkee, India
| | - Pravindra Kumar
- Department of Biotechnology, Indian Institute of Technology Roorkee, India.
| |
Collapse
|
20
|
Ren J, Mistry TL, Su PC, Mehboob S, Demissie R, Fung LWM, Ghosh AK, Johnson ME. Determination of absolute configuration and binding efficacy of benzimidazole-based FabI inhibitors through the support of electronic circular dichroism and MM-GBSA techniques. Bioorg Med Chem Lett 2018; 28:2074-2079. [PMID: 29730028 DOI: 10.1016/j.bmcl.2018.04.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 01/12/2023]
Abstract
We have previously reported benzimidazole-based compounds to be potent inhibitors of FabI for Francisella tularensis (FtFabI), making them promising antimicrobial hits. Optically active enantiomers exhibit markedly differing affinities toward FtFabI. The IC50 of benzimidazole (-)-1 is ∼100× lower than the (+)-enantiomer, with similar results for the 2 enantiomers. Determining the absolute configuration for these optical compounds and elucidating their binding modes is important for further design. Electronic circular dichroism (ECD) quantum calculations have become important in determining absolute configurations of optical compounds. We determined the absolute configuration of (-)/(+)-1 and (-)/(+)-2 by comparing experimental spectra and theoretical density functional theory (DFT) simulations of ECD spectra at the B3LYP/6-311+G(2d, p) level using Gaussian09. Comparison of experimental and calculated ECD spectra indicates that the S configuration corresponds to the (-)-rotation for both compounds 1 and 2, while the R configuration corresponds to the (+)-rotation. Further, molecular dynamics simulations and MM-GBSA binding energy calculations for these two pairs of enantiomers with FtFabI show much tighter binding MM-GBSA free energies for S-1 and S-2 than for their enantiomers, R-1 and R-2, consistent with the S configuration being the more active one, and with the ECD determination of the S configuration corresponding to (-) and the R configuration corresponding to (+). Thus, our computational studies allow us to assign (-) to (S)- and (+) to (R)- for compounds 1 and 2, and to further evaluate structural changes to improve efficacy.
Collapse
Affiliation(s)
- Jinhong Ren
- Center for Biomolecular Sciences, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL 60607, USA
| | - Tina L Mistry
- Center for Biomolecular Sciences, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL 60607, USA
| | - Pin-Chih Su
- Center for Biomolecular Sciences, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL 60607, USA
| | - Shahila Mehboob
- Novalex Therapeutics, Inc., 2242 W Harrison, Chicago, IL 60612, USA
| | - Robel Demissie
- Department of Chemistry, University of Illinois at Chicago, 845 W. Taylor St, Chicago, IL 60607, USA
| | - Leslie Wo-Mei Fung
- Department of Chemistry, University of Illinois at Chicago, 845 W. Taylor St, Chicago, IL 60607, USA
| | - Arun K Ghosh
- Department of Chemistry and Department of Medicinal Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
| | - Michael E Johnson
- Center for Biomolecular Sciences, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL 60607, USA; Novalex Therapeutics, Inc., 2242 W Harrison, Chicago, IL 60612, USA.
| |
Collapse
|
21
|
Wang J, Guo H, Cao C, Zhao W, Kwok LY, Zhang H, Zhang W. Characterization of the Adaptive Amoxicillin Resistance of Lactobacillus casei Zhang by Proteomic Analysis. Front Microbiol 2018; 9:292. [PMID: 29515561 PMCID: PMC5826216 DOI: 10.3389/fmicb.2018.00292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 02/07/2018] [Indexed: 12/15/2022] Open
Abstract
Amoxicillin is one of the most commonly prescribed antibiotics for bacterial infections and gastrointestinal disorders. To investigate the adaptation of Lactobacillus (L.) casei Zhang to amoxicillin stress, an iTRAQ-based comparative proteomic analysis was performed to compare the protein profiles between the parental L. casei Zhang and its amoxicillin-resistant descendent strains. Our results revealed a significant increase in the relative expression of 38 proteins (>2.0-folds, P < 0.05), while the relative expression of 34 proteins significantly decreased (<-2.0-folds, P < 0.05). The amoxicillin-resistant descendent strain exhibited marked alterations in carbohydrate and amino acid metabolism. Moreover, certain components involving in membrane metabolism were activated. The differences in the proteomic profiles between the two strains might explain the enhanced stress resistance of the adapted bacteria.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wenyi Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
22
|
Modification of membrane properties and fatty acids biosynthesis-related genes in Escherichia coli and Staphylococcus aureus: Implications for the antibacterial mechanism of naringenin. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:481-490. [PMID: 29138066 DOI: 10.1016/j.bbamem.2017.11.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 10/11/2017] [Accepted: 11/10/2017] [Indexed: 11/24/2022]
Abstract
In this work, modifications of cell membrane fluidity, fatty acid composition and fatty acid biosynthesis-associated genes of Escherichia coli ATCC 25922 (E. coli) and Staphylococcus aureus ATCC 6538 (S. aureus), during growth in the presence of naringenin (NAR), one of the natural antibacterial components in citrus plants, was investigated. Compared to E. coli, the growth of S. aureus was significantly inhibited by NAR in low concentrations. Combination of gas chromatography-mass spectrometry with fluorescence polarization analysis revealed that E. coli and S. aureus cells increased membrane fluidity by altering the composition of membrane fatty acids after exposure to NAR. For example, E. coli cells produced more unsaturated fatty acids (from 18.5% to 43.3%) at the expense of both cyclopropane and saturated fatty acids after growth in the concentrations of NAR from 0 to 2.20mM. For S. aureus grown with NAR at 0 to 1.47mM, the relative proportions of anteiso-branched chain fatty acids increased from 37.2% to 54.4%, whereas iso-branched and straight chain fatty acids decreased from 30.0% and 33.1% to 21.6% and 23.7%, respectively. Real time q-PCR analysis showed that NAR at higher concentrations induced a significant down-regulation of fatty acid biosynthesis-associated genes in the bacteria, with the exception of an increased expression of fabA gene. The minimum inhibitory concentration (MIC) of NAR against these two bacteria was determined, and both of bacteria underwent morphological changes after exposure to 1.0 and 2.0 MIC.
Collapse
|
23
|
Tahtaci H, Karacık H, Ece A, Er M, Şeker MG. Design, Synthesis, SAR and Molecular Modeling Studies of Novel Imidazo[2,1-b][1,3,4]Thiadiazole Derivatives as Highly Potent Antimicrobial Agents. Mol Inform 2017; 37. [DOI: 10.1002/minf.201700083] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/19/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Hakan Tahtaci
- Department of Polymer Engineering; Faculty of Technology, Karabuk University; 78050 Karabuk, Turkey
| | - Hatice Karacık
- Department of Polymer Engineering; Faculty of Technology, Karabuk University; 78050 Karabuk, Turkey
| | - Abdulilah Ece
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy; Biruni University; 34010 Istanbul Turkey
| | - Mustafa Er
- Department of Chemical Engineering, Faculty of Engineering; Karabuk University; 78050 Karabuk Turkey
| | - Mine Gül Şeker
- Department of Molecular Biology and Genetics, Faculty of Science; Gebze Technical University; 41400 Gebze Turkey
| |
Collapse
|
24
|
Yao J, Rock CO. Exogenous fatty acid metabolism in bacteria. Biochimie 2017; 141:30-39. [PMID: 28668270 DOI: 10.1016/j.biochi.2017.06.015] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/26/2017] [Indexed: 10/19/2022]
Abstract
Bacterial type II fatty acid synthesis (FASII) is a target for novel antibiotic development. All bacteria encode for mechanisms to incorporate exogenous fatty acids, and some bacteria can use exogenous fatty acids to bypass FASII inhibition. Bacteria encode three different mechanisms for activating exogenous fatty acids for incorporation into phospholipid synthesis. Exogenous fatty acids are converted into acyl-CoA in Gammaproteobacteria such as E. coli. Acyl-CoA molecules constitute a separate pool from endogenously synthesized acyl-ACP. Acyl-CoA can be used for phospholipid synthesis or broken down by β-oxidation, but cannot be used for lipopolysaccharide synthesis. Exogenous fatty acids are converted into acyl-ACP in some Gram-negative bacteria. The resulting acyl-ACP undergoes the same fates as endogenously synthesized acyl-ACP. Exogenous fatty acids are converted into acyl-phosphates in Gram-positive bacteria, and can be used for phospholipid synthesis or become acyl-ACP. Only the order Lactobacillales can use exogenous fatty acids to bypass FASII inhibition. FASII shuts down completely in presence of exogenous fatty acids in Lactobacillales, allowing Lactobacillales to synthesize phospholipids entirely from exogenous fatty acids. Inhibition of FASII cannot be bypassed in other bacteria because FASII is only partially down-regulated in presence of exogenous fatty acid or FASII is required to synthesize essential metabolites such as β-hydroxyacyl-ACP. Certain selective pressures such as FASII inhibition or growth in biofilms can select for naturally occurring one step mutations that attenuate endogenous fatty acid synthesis. Although attempts have been made to estimate the natural prevalence of these mutants, culture-independent metagenomic methods would provide a better estimate.
Collapse
Affiliation(s)
- Jiangwei Yao
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Charles O Rock
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
25
|
Studies of Staphylococcus aureus FabI inhibitors: fragment-based approach based on holographic structure-activity relationship analyses. Future Med Chem 2017; 9:135-151. [PMID: 28128979 DOI: 10.4155/fmc-2016-0179] [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] [Indexed: 12/16/2022] Open
Abstract
AIM FabI is a key enzyme in the fatty acid metabolism of Gram-positive bacteria such as Staphylococcus aureus and is an established drug target for known antibiotics such as triclosan. However, due to increasing antibacterial resistance, there is an urgent demand for new drug discovery. Recently, aminopyridine derivatives have been proposed as promising competitive inhibitors of FabI. METHODS In the present study, holographic structure-activity relationship (HQSAR) analyses were employed for determining structural contributions of a series containing 105 FabI inhibitors. RESULTS & CONCLUSION The final HQSAR model was robust and predictive according to statistical validation (q2 and r2pred equal to 0.696 and 0.854, respectively) and could be further employed to generate fragment contribution maps. Then, final HQSAR model together with FabI active site information can be useful for designing novel bioactive ligands.
Collapse
|
26
|
Docking and molecular dynamics studies on triclosan derivatives binding to FabI. J Mol Model 2017; 23:25. [DOI: 10.1007/s00894-016-3192-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 12/15/2016] [Indexed: 10/20/2022]
|
27
|
|
28
|
Zhou Y, Yang YS, Song XD, Lu L, Zhu HL. Study of Schiff-Base-Derived with Dioxygenated Rings and Nitrogen Heterocycle as Potential β-Ketoacyl-acyl Carrier Protein Synthase III (FabH) Inhibitors. Chem Pharm Bull (Tokyo) 2017; 65:178-185. [DOI: 10.1248/cpb.c16-00772] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yang Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University
| | - Yu-Shun Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University
| | - Xiao-Da Song
- School of Life Science and Technology, China Pharmaceutical University
| | - Liang Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University
| |
Collapse
|
29
|
Scheie AA, Petersen FC. The Biofilm Concept: Consequences for Future Prophylaxis of Oral Diseases? ACTA ACUST UNITED AC 2016; 15:4-12. [PMID: 14761896 DOI: 10.1177/154411130401500102] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Biofilm control is fundamental to oral health. Existing oral prophylactic measures, however, are insufficient. The main reason is probably because the micro-organisms involved organize into complex biofilm communities with features that differ from those of planktonic cells. Micro-organisms have traditionally been studied in the planktonic state. Conclusions drawn from many of these studies, therefore, need to be revalidated. Recent global approaches to the study of microbial gene expression and regulation in non-oral micro-organisms have shed light on two-component and quorum-sensing systems for the transduction of stimuli that allow for coordinated gene expression. We suggest interference with two-component and quorum-sensing systems as potential novel strategies for the prevention of oral diseases through control of oral biofilms. Information is still lacking, however, on the genetic regulation of oral biofilm formation. A better understanding of these processes is of considerable importance.
Collapse
Affiliation(s)
- Anne Aamdal Scheie
- Dept. of Oral Biology, Faculty of Dentistry, University of Oslo, PB 1052 Blindern, 0316 Oslo, Norway
| | | |
Collapse
|
30
|
Bacterial fatty acid metabolism in modern antibiotic discovery. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:1300-1309. [PMID: 27668701 DOI: 10.1016/j.bbalip.2016.09.014] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/12/2016] [Accepted: 09/19/2016] [Indexed: 12/28/2022]
Abstract
Bacterial fatty acid synthesis is essential for many pathogens and different from the mammalian counterpart. These features make bacterial fatty acid synthesis a desirable target for antibiotic discovery. The structural divergence of the conserved enzymes and the presence of different isozymes catalyzing the same reactions in the pathway make bacterial fatty acid synthesis a narrow spectrum target rather than the traditional broad spectrum target. Furthermore, bacterial fatty acid synthesis inhibitors are single-targeting, rather than multi-targeting like traditional monotherapeutic, broad-spectrum antibiotics. The single-targeting nature of bacterial fatty acid synthesis inhibitors makes overcoming fast-developing, target-based resistance a necessary consideration for antibiotic development. Target-based resistance can be overcome through multi-targeting inhibitors, a cocktail of single-targeting inhibitors, or by making the single targeting inhibitor sufficiently high affinity through a pathogen selective approach such that target-based mutants are still susceptible to therapeutic concentrations of drug. Many of the pathogens requiring new antibiotic treatment options encode for essential bacterial fatty acid synthesis enzymes. This review will evaluate the most promising targets in bacterial fatty acid metabolism for antibiotic therapeutics development and review the potential and challenges in advancing each of these targets to the clinic and circumventing target-based resistance. This article is part of a Special Issue entitled: Bacterial Lipids edited by Russell E. Bishop.
Collapse
|
31
|
Pompilio A, Riviello A, Crocetta V, Di Giuseppe F, Pomponio S, Sulpizio M, Di Ilio C, Angelucci S, Barone L, Di Giulio A, Di Bonaventura G. Evaluation of antibacterial and antibiofilm mechanisms by usnic acid against methicillin-resistant Staphylococcus aureus. Future Microbiol 2016; 11:1315-1338. [PMID: 27633726 DOI: 10.2217/fmb-2016-0049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To evaluate the antibacterial and antibiofilm mechanisms of usnic acid (USN) against methicillin-resistant Staphylococcus aureus from cystic fibrosis patients. MATERIALS & METHODS The effects exerted by USN at subinhibitory concentrations on S. aureus Sa3 strain was evaluated by proteomic, real-time PCR and electron microscopy analyses. RESULTS & CONCLUSION Proteomic analysis showed that USN caused damage in peptidoglycan synthesis, as confirmed by microscopy. Real-time PCR analysis showed that antibiofilm activity of USN is mainly due to impaired adhesion to the host matrix binding proteins, and decreasing lipase and thermonuclease expression. Our data show that USN exerts anti-staphylococcal effects through multitarget inhibitory effects, thus confirming the rationale for considering it 'lead compound' for the treatment of cystic fibrosis infections.
Collapse
Affiliation(s)
- Arianna Pompilio
- Department of Medical, Oral & Biotechnological Sciences, 'G d'Annunzio' University of Chieti-Pescara, Via Vestini 31, Chieti, Italy.,Aging Research Center and Translational Medicine, 'G d'Annunzio' University of Chieti-Pescara, Via L Polacchi 13, Chieti, Italy
| | - Antonella Riviello
- Department of Medical, Oral & Biotechnological Sciences, 'G d'Annunzio' University of Chieti-Pescara, Via Vestini 31, Chieti, Italy.,Aging Research Center and Translational Medicine, 'G d'Annunzio' University of Chieti-Pescara, Via L Polacchi 13, Chieti, Italy.,Stem TeCh Group, Via L Polacchi 13, Chieti, Italy
| | - Valentina Crocetta
- Department of Medical, Oral & Biotechnological Sciences, 'G d'Annunzio' University of Chieti-Pescara, Via Vestini 31, Chieti, Italy.,Aging Research Center and Translational Medicine, 'G d'Annunzio' University of Chieti-Pescara, Via L Polacchi 13, Chieti, Italy
| | - Fabrizio Di Giuseppe
- Department of Medical, Oral & Biotechnological Sciences, 'G d'Annunzio' University of Chieti-Pescara, Via Vestini 31, Chieti, Italy.,Aging Research Center and Translational Medicine, 'G d'Annunzio' University of Chieti-Pescara, Via L Polacchi 13, Chieti, Italy.,Stem TeCh Group, Via L Polacchi 13, Chieti, Italy
| | - Stefano Pomponio
- Department of Medical, Oral & Biotechnological Sciences, 'G d'Annunzio' University of Chieti-Pescara, Via Vestini 31, Chieti, Italy.,Aging Research Center and Translational Medicine, 'G d'Annunzio' University of Chieti-Pescara, Via L Polacchi 13, Chieti, Italy
| | - Marilisa Sulpizio
- Department of Medical, Oral & Biotechnological Sciences, 'G d'Annunzio' University of Chieti-Pescara, Via Vestini 31, Chieti, Italy.,Aging Research Center and Translational Medicine, 'G d'Annunzio' University of Chieti-Pescara, Via L Polacchi 13, Chieti, Italy.,Stem TeCh Group, Via L Polacchi 13, Chieti, Italy
| | - Carmine Di Ilio
- Department of Medical, Oral & Biotechnological Sciences, 'G d'Annunzio' University of Chieti-Pescara, Via Vestini 31, Chieti, Italy.,Aging Research Center and Translational Medicine, 'G d'Annunzio' University of Chieti-Pescara, Via L Polacchi 13, Chieti, Italy.,Stem TeCh Group, Via L Polacchi 13, Chieti, Italy
| | - Stefania Angelucci
- Department of Medical, Oral & Biotechnological Sciences, 'G d'Annunzio' University of Chieti-Pescara, Via Vestini 31, Chieti, Italy.,Aging Research Center and Translational Medicine, 'G d'Annunzio' University of Chieti-Pescara, Via L Polacchi 13, Chieti, Italy.,Stem TeCh Group, Via L Polacchi 13, Chieti, Italy
| | - Luana Barone
- Department of Science, LIME, University Roma Tre, Viale G Marconi 446, Rome, Italy
| | - Andrea Di Giulio
- Department of Science, LIME, University Roma Tre, Viale G Marconi 446, Rome, Italy
| | - Giovanni Di Bonaventura
- Department of Medical, Oral & Biotechnological Sciences, 'G d'Annunzio' University of Chieti-Pescara, Via Vestini 31, Chieti, Italy.,Aging Research Center and Translational Medicine, 'G d'Annunzio' University of Chieti-Pescara, Via L Polacchi 13, Chieti, Italy
| |
Collapse
|
32
|
Kar SS, Bhat G V, Rao PPN, Shenoy VP, Bairy I, Shenoy GG. Rational design and synthesis of novel diphenyl ether derivatives as antitubercular agents. Drug Des Devel Ther 2016; 10:2299-310. [PMID: 27486307 PMCID: PMC4958353 DOI: 10.2147/dddt.s104037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A series of triclosan mimic diphenyl ether derivatives have been synthesized and evaluated for their in vitro antitubercular activity against Mycobacterium tuberculosis H37Rv. The binding mode of the compounds at the active site of enoyl-acyl carrier protein reductase of M. tuberculosis has been explored. Among them, compound 10b was found to possess antitubercular activity (minimum inhibitory concentration =12.5 µg/mL) comparable to triclosan. All the synthesized compounds exhibited low levels of cytotoxicity against Vero and HepG2 cell lines, and three compounds 10a, 10b, and 10c had a selectivity index more than 10. Compound 10b was also evaluated for log P, pKa, human liver microsomal stability, and % protein binding, in order to probe its druglikeness. Based on the antitubercular activity and druglikeness profile, it may be concluded that compound 10b could be a lead for future development of antitubercular drugs.
Collapse
Affiliation(s)
- Sidhartha S Kar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, India
| | - Varadaraj Bhat G
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, India
| | - Praveen PN Rao
- School of Pharmacy, Health Sciences Campus, University of Waterloo, Waterloo, ON, Canada
| | - Vishnu P Shenoy
- Department of Microbiology, Kasturba Medical College, Manipal
| | - Indira Bairy
- Melaka-Manipal Medical College, Manipal University, Manipal, India
| | - G Gautham Shenoy
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, India
| |
Collapse
|
33
|
Furi L, Haigh R, Al Jabri ZJH, Morrissey I, Ou HY, León-Sampedro R, Martinez JL, Coque TM, Oggioni MR. Dissemination of Novel Antimicrobial Resistance Mechanisms through the Insertion Sequence Mediated Spread of Metabolic Genes. Front Microbiol 2016; 7:1008. [PMID: 27446047 PMCID: PMC4923244 DOI: 10.3389/fmicb.2016.01008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 06/13/2016] [Indexed: 12/14/2022] Open
Abstract
The widely used biocide triclosan selectively targets FabI, the NADH-dependent trans-2-enoyl-acyl carrier protein (ACP) reductase, which is also an important target for the development of narrow spectrum antibiotics. The analysis of triclosan resistant Staphylococcus aureus isolates had previously shown that in about half of the strains, the mechanism of triclosan resistance consists on the heterologous duplication of the triclosan target gene due to the acquisition of an additional fabI allele derived from Staphylococcus haemolyticus (sh-fabI). In the current work, the genomic sequencing of 10 of these strains allowed the characterization of two novel composite transposons TnSha1 and TnSha2 involved in the spread of sh-fabI. TnSha1 harbors one copy of IS1272, whereas TnSha2 is a 11.7 kb plasmid carrying TnSha1 present either as plasmid or in an integrated form generally flanked by two IS1272 elements. The target and mechanism of integration for IS1272 and TnSha1 are novel and include targeting of DNA secondary structures, generation of blunt-end deletions of the stem-loop and absence of target duplication. Database analyses showed widespread occurrence of these two elements in chromosomes and plasmids, with TnSha1 mainly in S. aureus and with TnSha2 mainly in S. haemolyticus and S. epidermidis. The acquisition of resistance by means of an insertion sequence-based mobilization and consequent duplication of drug-target metabolic genes, as observed here for sh-fabI, is highly reminiscent of the situation with the ileS2 gene conferring mupirocin resistance, and the dfrA and dfrG genes conferring trimethoprim resistance both of which are mobilized by IS257. These three examples, which show similar mechanisms and levels of spread of metabolic genes linked to IS elements, highlight the importance of this genetic strategy for recruitment and rapid distribution of novel resistance mechanisms in staphylococci.
Collapse
Affiliation(s)
- Leonardo Furi
- Department of Genetics, University of LeicesterLeicester, UK; Dipartimento di Biotecnologie Mediche, Universita di SienaSiena, Italy
| | - Richard Haigh
- Department of Genetics, University of Leicester Leicester, UK
| | | | | | - Hong-Yu Ou
- State Key Laboratory for Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiaotong University Shanghai, China
| | - Ricardo León-Sampedro
- Departamento de Microbiología, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y CajalMadrid, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Spain
| | - Jose L Martinez
- Departamento de Biotecnología Microbiana, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones CientíficasMadrid, Spain; Unidad de Resistencia a Antibióticos y Virulencia Bacteriana (RYC-Consejo Superior de Investigaciones Científicas)Madrid, Spain
| | - Teresa M Coque
- Departamento de Microbiología, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y CajalMadrid, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)Spain; Unidad de Resistencia a Antibióticos y Virulencia Bacteriana (RYC-Consejo Superior de Investigaciones Científicas)Madrid, Spain
| | - Marco R Oggioni
- Department of Genetics, University of LeicesterLeicester, UK; Dipartimento di Biotecnologie Mediche, Universita di SienaSiena, Italy
| |
Collapse
|
34
|
Zhou Y, Luo Y, Yang YS, Lu L, Zhu HL. Study of acylhydrazone derivatives with deoxygenated seven-membered rings as potential β-ketoacyl-acyl carrier protein synthase III (FabH) inhibitors. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00263c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fatty acid biosynthesis is essential for bacterial survival.
Collapse
Affiliation(s)
- Yang Zhou
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210023
- People's Republic of China
| | - Yin Luo
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210023
- People's Republic of China
| | - Yu-Shun Yang
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210023
- People's Republic of China
| | - Liang Lu
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210023
- People's Republic of China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210023
- People's Republic of China
| |
Collapse
|
35
|
Sim WC, Kim DG, Lee KJ, Choi YJ, Choi YJ, Shin KJ, Jun DW, Park SJ, Park HJ, Kim J, Oh WK, Lee BH. Cinnamamides, Novel Liver X Receptor Antagonists that Inhibit Ligand-Induced Lipogenesis and Fatty Liver. J Pharmacol Exp Ther 2015; 355:362-9. [PMID: 26384859 DOI: 10.1124/jpet.115.226738] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/17/2015] [Indexed: 08/30/2023] Open
Abstract
Liver X receptor (LXR) is a member of the nuclear receptor superfamily, and it regulates various biologic processes, including de novo lipogenesis, cholesterol metabolism, and inflammation. Selective inhibition of LXR may aid the treatment of nonalcoholic fatty liver diseases. In the present study, we evaluated the effects of three cinnamamide derivatives on ligand-induced LXRα activation and explored whether these derivatives could attenuate steatosis in mice. N-(4-trifluoromethylphenyl) 3,4-dimethoxycinnamamide (TFCA) decreased the luciferase activity in LXRE-tk-Luc-transfected cells and also suppressed ligand-induced lipid accumulation and expression of the lipogenic genes in murine hepatocytes. Furthermore, it significantly attenuated hepatic neutral lipid accumulation in a ligand-induced fatty liver mouse system. Modeling study indicated that TFCA inhibited activation of the LXRα ligand-binding domain by hydrogen bonding to Arg305 in the H5 region of that domain. It regulated the transcriptional control exerted by LXRα by influencing coregulator exchange; this process involves dissociation of the thyroid hormone receptor-associated proteins (TRAP)/DRIP coactivator and recruitment of the nuclear receptor corepressor. These results show that TFCA has the potential to attenuate ligand-induced lipogenesis and fatty liver by selectively inhibiting LXRα in the liver.
Collapse
Affiliation(s)
- Woo-Cheol Sim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - Dong Gwang Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - Kyeong Jin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - You-Jin Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - Yeon Jae Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - Kye Jung Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - Dae Won Jun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - So-Jung Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - Hyun-Ju Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - Jiwon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - Won Keun Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| | - Byung-Hoon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea (W.C.S., D.G.K., K.J.L., You-Jin C., J.W.K., W.K.O., B.H.L.); College of Pharmacy, The Catholic University, Republic of Korea (Yeon-Jae C., K.J.S.); Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea (D.W.J.); School of Pharmacy, Sungkyunkwan University, Jangan-gu, Republic of Korea (S.J.P., H.J.P.)
| |
Collapse
|
36
|
Shang R, Liang J, Yi Y, Liu Y, Wang J. Review of Platensimycin and Platencin: Inhibitors of β-Ketoacyl-acyl Carrier Protein (ACP) Synthase III (FabH). Molecules 2015; 20:16127-41. [PMID: 26404223 PMCID: PMC6332302 DOI: 10.3390/molecules200916127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 08/24/2015] [Accepted: 08/28/2015] [Indexed: 11/29/2022] Open
Abstract
Platensimycin and platencin were successively discovered from the strain Streptomyces platensis through systematic screening. These natural products have been defined as promising agents for fighting multidrug resistance in bacteria by targeting type II fatty acid synthesis with slightly different mechanisms. Bioactivity studies have shown that platensimycin and platencin offer great potential to inhibit many resistant bacteria with no cross-resistance or toxicity observed in vivo. This review summarizes the general information on platensimycin and platencin, including antibacterial and self-resistant mechanisms. Furthermore, the total synthesis pathways of platensimycin and platencin and their analogues from recent studies are presented.
Collapse
Affiliation(s)
- Ruofeng Shang
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China.
| | - Jianping Liang
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China.
| | - Yunpeng Yi
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China.
| | - Yu Liu
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China.
| | - Jiatu Wang
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China.
| |
Collapse
|
37
|
Kondakova T, D'Heygère F, Feuilloley MJ, Orange N, Heipieper HJ, Duclairoir Poc C. Glycerophospholipid synthesis and functions in Pseudomonas. Chem Phys Lipids 2015; 190:27-42. [PMID: 26148574 DOI: 10.1016/j.chemphyslip.2015.06.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/29/2015] [Accepted: 06/30/2015] [Indexed: 11/25/2022]
Abstract
The genus Pseudomonas is one of the most heterogeneous groups of eubacteria, presents in all major natural environments and in wide range of associations with plants and animals. The wide distribution of these bacteria is due to the use of specific mechanisms to adapt to environmental modifications. Generally, bacterial adaptation is only considered under the aspect of genes and protein expression, but lipids also play a pivotal role in bacterial functioning and homeostasis. This review resumes the mechanisms and regulations of pseudomonal glycerophospholipid synthesis, and the roles of glycerophospholipids in bacterial metabolism and homeostasis. Recently discovered specific pathways of P. aeruginosa lipid synthesis indicate the lineage dependent mechanisms of fatty acids homeostasis. Pseudomonas glycerophospholipids ensure structure functions and play important roles in bacterial adaptation to environmental modifications. The lipidome of Pseudomonas contains a typical eukaryotic glycerophospholipid--phosphatidylcholine -, which is involved in bacteria-host interactions. The ability of Pseudomonas to exploit eukaryotic lipids shows specific and original strategies developed by these microorganisms to succeed in their infectious process. All compiled data provide the demonstration of the importance of studying the Pseudomonas lipidome to inhibit the infectious potential of these highly versatile germs.
Collapse
Affiliation(s)
- Tatiana Kondakova
- Normandie University of Rouen, Laboratory of Microbiology Signals and Microenvironment (LMSM), EA 4312, 55 rue St. Germain, 27000 Evreux, France
| | - François D'Heygère
- Centre de Biophysique Moléculaire, CNRS, UPR4301, rue Charles Sadron, 45071 Orléans, France
| | - Marc J Feuilloley
- Normandie University of Rouen, Laboratory of Microbiology Signals and Microenvironment (LMSM), EA 4312, 55 rue St. Germain, 27000 Evreux, France
| | - Nicole Orange
- Normandie University of Rouen, Laboratory of Microbiology Signals and Microenvironment (LMSM), EA 4312, 55 rue St. Germain, 27000 Evreux, France
| | - Hermann J Heipieper
- Department of Environmental Biotechnology, UFZ Helmholtz Centre for Environmental Research, Permoserstr. 15, 04318 Leipzig, Germany
| | - Cécile Duclairoir Poc
- Normandie University of Rouen, Laboratory of Microbiology Signals and Microenvironment (LMSM), EA 4312, 55 rue St. Germain, 27000 Evreux, France.
| |
Collapse
|
38
|
Target-based molecular modeling strategies for schistosomiasis drug discovery. Future Med Chem 2015; 7:753-64. [DOI: 10.4155/fmc.15.21] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Schistosomiasis, a neglected tropical disease caused by worms from the class Trematoda (genus Schistosoma), is a serious chronic condition that has been reported in approximately 80 countries. Nearly 250 million people are affected worldwide, mostly in the sub-Saharan Africa. Praziquantel, the mainstay of treatment, has been used for 30 years, and cases of resistance have been reported. The purpose of this perspective is to discuss current target-based molecular modeling strategies in schistosomiasis drug discovery. Advances in the field and the role played by the integration between computational modeling and experimental validation are also discussed. Finally, recent cases of the contribution of modern approaches in computational medicinal chemistry to the field are explored.
Collapse
|
39
|
Mobegi FM, van Hijum SAFT, Burghout P, Bootsma HJ, de Vries SPW, van der Gaast-de Jongh CE, Simonetti E, Langereis JD, Hermans PWM, de Jonge MI, Zomer A. From microbial gene essentiality to novel antimicrobial drug targets. BMC Genomics 2014; 15:958. [PMID: 25373505 PMCID: PMC4233050 DOI: 10.1186/1471-2164-15-958] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 10/23/2014] [Indexed: 01/12/2023] Open
Abstract
Background Bacterial respiratory tract infections, mainly caused by Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis are among the leading causes of global mortality and morbidity. Increased resistance of these pathogens to existing antibiotics necessitates the search for novel targets to develop potent antimicrobials. Result Here, we report a proof of concept study for the reliable identification of potential drug targets in these human respiratory pathogens by combining high-density transposon mutagenesis, high-throughput sequencing, and integrative genomics. Approximately 20% of all genes in these three species were essential for growth and viability, including 128 essential and conserved genes, part of 47 metabolic pathways. By comparing these essential genes to the human genome, and a database of genes from commensal human gut microbiota, we identified and excluded potential drug targets in respiratory tract pathogens that will have off-target effects in the host, or disrupt the natural host microbiota. We propose 249 potential drug targets, 67 of which are targets for 75 FDA-approved antimicrobials and 35 other researched small molecule inhibitors. Two out of four selected novel targets were experimentally validated, proofing the concept. Conclusion Here we have pioneered an attempt in systematically combining the power of high-density transposon mutagenesis, high-throughput sequencing, and integrative genomics to discover potential drug targets at genome-scale. By circumventing the time-consuming and expensive laboratory screens traditionally used to select potential drug targets, our approach provides an attractive alternative that could accelerate the much needed discovery of novel antimicrobials. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-958) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Sacha A F T van Hijum
- Radboud Institute for Molecular Life Sciences, Laboratory of Paediatric Infectious Diseases, Radboud University Medical Centre, Nijmegen 6500 HB, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang SF, Yin Y, Wu X, Qiao F, Sha S, Lv PC, Zhao J, Zhu HL. Synthesis, molecular docking and biological evaluation of coumarin derivatives containing piperazine skeleton as potential antibacterial agents. Bioorg Med Chem 2014; 22:5727-37. [PMID: 25306465 DOI: 10.1016/j.bmc.2014.09.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 09/06/2014] [Accepted: 09/23/2014] [Indexed: 11/24/2022]
Abstract
A series of 4-hydroxycoumarin derivatives were designed and synthesized in order to find some more potent antibacterial drugs. Their antibacterial activities against Escherichia coli, Pseudomonas aeruginosa, Bacillus subtilis and Staphylococcus aureus were tested. These compounds showed good antibacterial activities against Gram-positive strains. Compound 4 g represented the most potent antibacterial activity against Bacillus subtilis and S. aureus with MIC of 0.236, 0.355 μg/mL, respectively. What's more, it showed the most potent activity against SaFabI with IC50 of 0.57 μM. Molecular docking of 4 g into S. aureus Enoyl-ACP-reductase active site were performed to determine the probable binding mode, while the QSAR model was built to check the previous work as well as to introduce new directions.
Collapse
Affiliation(s)
- She-Feng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Yong Yin
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Xun Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Fang Qiao
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Shao Sha
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Peng-Cheng Lv
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Jing Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China.
| |
Collapse
|
41
|
Joshi SD, Dixit SR, More UA, Raju KVSN, Narayan R, Aminabhavi TM, Kulkarni VH. 3D-QSAR and molecular docking studies of 1,3,4-oxadiazoles containing substituted phenoxy fragment as inhibitors of enoyl-acyl carrier protein reductase from Escherichia coli. Med Chem Res 2014. [DOI: 10.1007/s00044-014-1013-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
42
|
Falzone M, Martens E, Tynan H, Maggio C, Golden S, Nayda V, Crespo E, Inamine G, Gelber M, Lemence R, Chiappini N, Friedman E, Shen B, Gullo V, Demain AL. Development of a chemically defined medium for the production of the antibiotic platensimycin by Streptomyces platensis. Appl Microbiol Biotechnol 2014; 97:9535-9. [PMID: 24022611 DOI: 10.1007/s00253-013-5201-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 06/14/2013] [Accepted: 08/14/2013] [Indexed: 11/28/2022]
Abstract
The actinomycete Streptomyces platensis produces two compounds that display antibacterial activity: platensimycin and platencin. These compounds were discovered by the Merck Research Laboratories, and a complex insoluble production medium was reported. We have used this medium as our starting point in our studies. In a previous study, we developed a semi-defined production medium, i.e., PM5. In the present studies, by varying the concentration of the components of PM5, we were able to develop a superior semi-defined medium, i.e., PM6, which contains a higher concentration of lactose. Versions of PM6, containing lower concentrations of all components, were also found to be superior to PM5. The new semi-defined production media contain dextrin, lactose, MOPS buffer, and ammonium sulfate in different concentrations. We determined antibiotic production capabilities using agar diffusion assays and chemical assays via thin-layer silica chromatography and high-performance liquid chromatography. We reduced crude nutrient carryover from the seed medium by washing the cells with distilled water. Using these semi-defined media, we determined that addition of the semi-defined component soluble starch stimulated antibiotic production and that it and dextrin could both be replaced with glucose, resulting in the chemically defined medium, PM7.
Collapse
|
43
|
Patel KN, Telvekar VN. Design, synthesis and antitubercular evaluation of novel series of N-[4-(piperazin-1-yl)phenyl]cinnamamide derivatives. Eur J Med Chem 2014; 75:43-56. [DOI: 10.1016/j.ejmech.2014.01.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 01/13/2014] [Accepted: 01/18/2014] [Indexed: 11/30/2022]
|
44
|
Shirude PS, Madhavapeddi P, Naik M, Murugan K, Shinde V, Nandishaiah R, Bhat J, Kumar A, Hameed S, Holdgate G, Davies G, McMiken H, Hegde N, Ambady A, Venkatraman J, Panda M, Bandodkar B, Sambandamurthy VK, Read JA. Methyl-thiazoles: a novel mode of inhibition with the potential to develop novel inhibitors targeting InhA in Mycobacterium tuberculosis. J Med Chem 2013; 56:8533-42. [PMID: 24107081 DOI: 10.1021/jm4012033] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
InhA is a well validated Mycobacterium tuberculosis (Mtb) target as evidenced by the clinical success of isoniazid. Translating enzyme inhibition to bacterial cidality by targeting the fatty acid substrate site of InhA remains a daunting challenge. The recent disclosure of a methyl-thiazole series demonstrates that bacterial cidality can be achieved with potent enzyme inhibition and appropriate physicochemical properties. In this study, we report the molecular mode of action of a lead methyl-thiazole, along with analogues with improved CYP inhibition profile. We have identified a novel mechanism of InhA inhibition characterized by a hitherto unreported "Y158-out" inhibitor-bound conformation of the protein that accommodates a neutrally charged "warhead". An additional novel hydrophilic interaction with protein residue M98 allows the incorporation of favorable physicochemical properties for cellular activity. Notably, the methyl-thiazole prefers the NADH-bound form of the enzyme with a Kd of ~13.7 nM, as against the NAD(+)-bound form of the enzyme.
Collapse
Affiliation(s)
- Pravin S Shirude
- Department of Medicinal Chemistry, ‡Department of Biosciences, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore-560024, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Liu J, Dyer D, Wang J, Wang S, Du X, Xu B, Zhang H, Wang X, Hu W. 3-oxoacyl-ACP reductase from Schistosoma japonicum: integrated in silico-in vitro strategy for discovering antischistosomal lead compounds. PLoS One 2013; 8:e64984. [PMID: 23762275 PMCID: PMC3676400 DOI: 10.1371/journal.pone.0064984] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 04/18/2013] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Schistosomiasis is a disease caused by parasitic worms and more than 200 million people are infected worldwide. The emergence of resistance to the most commonly used drug, praziquantel (PZQ), makes the development of novel drugs an urgent task. 3-oxoacyl-ACP reductase (OAR), a key enzyme involved in the fatty acid synthesis pathway, has been identified as a potential drug target against many pathogenic organisms. However, no research on Schistosoma japonicum OAR (SjOAR) has been reported. The characterization of the SjOAR protein will provide new strategies for screening antischistosomal drugs that target SjOAR. METHODOLOGY/PRINCIPAL FINDINGS After cloning the SjOAR gene, recombinant SjOAR protein was purified and assayed for enzymatic activity. The tertiary structure of SjOAR was obtained by homology modeling and 27 inhibitor candidates were identified from 14,400 compounds through molecular docking based on the structure. All of these compounds were confirmed to be able to bind to the SjOAR protein by BIAcore analysis. Two compounds exhibited strong antischistosomal activity and inhibitory effects on the enzymatic activity of SjOAR. In contrast, these two compounds showed relatively low toxicity towards host cells. CONCLUSIONS/SIGNIFICANCE The work presented here shows the feasibility of isolation of new antischistosomal compounds using a combination of virtual screening and experimental validation. Based on this strategy, we successfully identified 2 compounds that target SjOAR with strong antischistosomal activity but relatively low cytotoxicity to host cells.
Collapse
Affiliation(s)
- Jian Liu
- State Key Laboratory of Genetic Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Key Laboratory of Parasite and Vector Biology of Ministry of Public Health, Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Dave Dyer
- Department of Entomology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jipeng Wang
- State Key Laboratory of Genetic Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Shuqi Wang
- State Key Laboratory of Genetic Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaofeng Du
- State Key Laboratory of Genetic Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Bin Xu
- Key Laboratory of Parasite and Vector Biology of Ministry of Public Health, Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Haobing Zhang
- Key Laboratory of Parasite and Vector Biology of Ministry of Public Health, Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Xiaoning Wang
- State Key Laboratory of Genetic Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei Hu
- State Key Laboratory of Genetic Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Key Laboratory of Parasite and Vector Biology of Ministry of Public Health, Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| |
Collapse
|
46
|
Aluotto S, Tynan H, Maggio C, Falzone M, Mukherjee A, Gullo V, Demain AL. Development of a semi-defined medium supporting production of platensimycin and platencin by Streptomyces platensis. J Antibiot (Tokyo) 2012. [PMID: 23188381 DOI: 10.1038/ja.2012.97] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Platensimycin and platencin are compounds that were discovered at Merck Research Laboratories and have shown promising antibacterial activity. They are both produced in fermentation by the actinomycete Streptomyces platensis. Merck reported a crude, insoluble production medium to produce the antibiotics. To test the possible effects of different primary metabolites and inorganic compounds on the production of these antibiotics, a chemically-defined medium is needed. The effects that these compounds have on production could provide information about the precursors and biosynthetic pathway of the antibiotics. We have tested and developed a number of media with varying degrees of chemical definition and solubility using the Merck medium as our starting point. Our latest production medium, PM5, is soluble and semi-defined. It yields suitable production of the compounds, as shown by agar diffusion assays, bioautography and HPLC. The antibiotics were located in the extracellular broths and not in the mycelia.
Collapse
Affiliation(s)
- Sabrina Aluotto
- Charles A Dana Research Institute for Scientists Emerti, Drew University, Madison, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Mode of action, in vitro activity, and in vivo efficacy of AFN-1252, a selective antistaphylococcal FabI inhibitor. Antimicrob Agents Chemother 2012; 56:5865-74. [PMID: 22948878 PMCID: PMC3486558 DOI: 10.1128/aac.01411-12] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The mechanism of action of AFN-1252, a selective inhibitor of Staphylococcus aureus enoyl-acyl carrier protein reductase (FabI), which is involved in fatty acid biosynthesis, was confirmed by using biochemistry, macromolecular synthesis, genetics, and cocrystallization of an AFN-1252-FabI complex. AFN-1252 demonstrated a low propensity for spontaneous resistance development and a time-dependent reduction of the viability of both methicillin-susceptible and methicillin-resistant S. aureus, achieving a ≥2-log(10) reduction in S. aureus counts over 24 h, and was extremely potent against clinical isolates of S. aureus (MIC(90), 0.015 μg/ml) and coagulase-negative staphylococci (MIC(90), 0.12 μg/ml), regardless of their drug resistance, hospital- or community-associated origin, or other clinical subgroup. AFN-1252 was orally available in mouse pharmacokinetic studies, and a single oral dose of 1 mg/kg AFN-1252 was efficacious in a mouse model of septicemia, providing 100% protection from an otherwise lethal peritoneal infection of S. aureus Smith. A median effective dose of 0.15 mg/kg indicated that AFN-1252 was 12 to 24 times more potent than linezolid in the model. These studies, demonstrating a selective mode of action, potent in vitro activity, and in vivo efficacy, support the continued investigation of AFN-1252 as a targeted therapeutic for staphylococcal infections.
Collapse
|
48
|
Gerusz V, Denis A, Faivre F, Bonvin Y, Oxoby M, Briet S, LeFralliec G, Oliveira C, Desroy N, Raymond C, Peltier L, Moreau F, Escaich S, Vongsouthi V, Floquet S, Drocourt E, Walton A, Prouvensier L, Saccomani M, Durant L, Genevard JM, Sam-Sambo V, Soulama-Mouze C. From triclosan toward the clinic: discovery of nonbiocidal, potent FabI inhibitors for the treatment of resistant bacteria. J Med Chem 2012; 55:9914-28. [PMID: 23092194 DOI: 10.1021/jm301113w] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In this paper, we present some elements of our optimization program to decouple triclosan's specific FabI effect from its nonspecific cytotoxic component. The implementation of this strategy delivered highly specific, potent, and nonbiocidal new FabI inhibitors. We also disclose some preclinical data of one of their representatives, 83, a novel antibacterial compound active against resistant staphylococci and some clinically relevant Gram negative bacteria that is currently undergoing clinical trials.
Collapse
Affiliation(s)
- Vincent Gerusz
- Medicinal Chemistry and ‡Biology, Mutabilis, 102 Avenue Gaston Roussel, 93230 Romainville, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Schiebel J, Chang A, Lu H, Baxter MV, Tonge PJ, Kisker C. Staphylococcus aureus FabI: inhibition, substrate recognition, and potential implications for in vivo essentiality. Structure 2012; 20:802-13. [PMID: 22579249 DOI: 10.1016/j.str.2012.03.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 03/01/2012] [Accepted: 03/03/2012] [Indexed: 12/11/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections constitute a serious health threat worldwide, and novel antibiotics are therefore urgently needed. The enoyl-ACP reductase (saFabI) is essential for the S. aureus fatty acid biosynthesis and, hence, serves as an attractive drug target. We have obtained a series of snapshots of this enzyme that provide a mechanistic picture of ligand and inhibitor binding, including a dimer-tetramer transition combined with extensive conformational changes. Significantly, our results reveal key differences in ligand binding and recognition compared to orthologous proteins. The remarkable observed protein flexibility rationalizes our finding that saFabI is capable of efficiently reducing branched-chain fatty acid precursors. Importantly, branched-chain fatty acids represent a major fraction of the S. aureus cell membrane and are crucial for its in vivo fitness. Our discovery thus addresses a long-standing controversy regarding the essentiality of the fatty acid biosynthesis pathway in S. aureus rationalizing saFabI as a drug target.
Collapse
Affiliation(s)
- Johannes Schiebel
- Rudolf Virchow Center for Experimental Biomedicine, Institute for Structural Biology, University of Würzburg, D-97080 Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
The emergence of bacteria that are multiply resistant to commonly used antibiotics has created the medical need for novel classes of antibacterial agents. The unique challenges to the discovery of new antibacterial drugs include the following: spectrum, selectivity, low emergence of new resistance, and high potency. With the emergence of genomic information, dozens of antibacterial targets have been pursued over the last 2 decades often using SBDD. This chapter reviews the application of structure-based drug design approaches on a selected group of antibacterial targets (DHFR, DHNA, PDF, and FabI) where significant progress has been made. We compare and contrast the different approaches and evaluate the results in terms of the biological profiles of the leads produced. Several common themes have emerged from this survey, resulting in a set of recommendations.
Collapse
Affiliation(s)
- John Finn
- Trius Therapeutics, San Diego, CA, USA.
| |
Collapse
|