1
|
Motter J, Benckendorff CMM, Westarp S, Sunde-Brown P, Neubauer P, Kurreck A, Miller GJ. Purine nucleoside antibiotics: recent synthetic advances harnessing chemistry and biology. Nat Prod Rep 2024; 41:873-884. [PMID: 38197414 PMCID: PMC11188666 DOI: 10.1039/d3np00051f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Indexed: 01/11/2024]
Abstract
Covering: 2019 to 2023Nucleoside analogues represent one of the most important classes of small molecule pharmaceuticals and their therapeutic development is successfully established within oncology and for the treatment of viral infections. However, there are currently no nucleoside analogues in clinical use for the management of bacterial infections. Despite this, a significant number of clinically recognised nucleoside analogues are known to possess some antibiotic activity, thereby establishing a potential source for new therapeutic discovery in this area. Furthermore, given the rise in antibiotic resistance, the discovery of new clinical candidates remains an urgent global priority and natural product-derived nucleoside analogues may also present a rich source of discovery space for new modalities. This Highlight, covering work published from 2019 to 2023, presents a current perspective surrounding the synthesis of natural purine nucleoside antibiotics. By amalgamating recent efforts from synthetic chemistry with advances in biosynthetic understanding and the use of recombinant enzymes, prospects towards different structural classes of purines are detailed.
Collapse
Affiliation(s)
- Jonas Motter
- Chair of Bioprocess Engineering, Institute of Biotechnology, Faculty III Process Sciences, Technische Universität Berlin, Ackerstraße 76, D-13355, Berlin, Germany
| | - Caecilie M M Benckendorff
- School of Chemical and Physical Sciences and Centre for Glycoscience, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| | - Sarah Westarp
- Chair of Bioprocess Engineering, Institute of Biotechnology, Faculty III Process Sciences, Technische Universität Berlin, Ackerstraße 76, D-13355, Berlin, Germany
- BioNukleo GmbH, Ackerstraße 76, 13355 Berlin, Germany.
| | - Peter Sunde-Brown
- School of Chemical and Physical Sciences and Centre for Glycoscience, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| | - Peter Neubauer
- Chair of Bioprocess Engineering, Institute of Biotechnology, Faculty III Process Sciences, Technische Universität Berlin, Ackerstraße 76, D-13355, Berlin, Germany
| | - Anke Kurreck
- Chair of Bioprocess Engineering, Institute of Biotechnology, Faculty III Process Sciences, Technische Universität Berlin, Ackerstraße 76, D-13355, Berlin, Germany
- BioNukleo GmbH, Ackerstraße 76, 13355 Berlin, Germany.
| | - Gavin J Miller
- School of Chemical and Physical Sciences and Centre for Glycoscience, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| |
Collapse
|
2
|
Chen L, Li Y, Han M, Peng Y, Chen X, Xiang S, Gao H, Lu T, Luo SP, Zhou B, Wu H, Yang YF, Liu Y. P/ N-Heteroleptic Cu(I)-Photosensitizer-Catalyzed [3 + 2] Regiospecific Annulation of Aminocyclopropanes and Functionalized Alkynes. J Org Chem 2022; 87:15571-15581. [PMID: 36322051 DOI: 10.1021/acs.joc.2c02138] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We report here a regiospecific [3 + 2] annulation between aminocyclopropanes and various functionalized alkynes enabled by a P/N-heteroleptic Cu(I) photosensitizer under photoredox catalysis conditions. Thus, a divergent construction of 3-aminocyclopentene derivatives including methylsulfonyl-, arylsulfonyl-, chloro-, ester-, and trifluoromethyl-functionalized aminocyclopentenes could be achieved with advantages of high regioselectivity, broad substrate compatibility, and mild and environmentally benign reaction conditions.
Collapse
Affiliation(s)
- Lailin Chen
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Ya Li
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Mingfeng Han
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Yun Peng
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Xiahe Chen
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Siwei Xiang
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Hong Gao
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Tianhao Lu
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Shu-Ping Luo
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Bingwei Zhou
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Huayue Wu
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou 325027, People's Republic of China
| | - Yun-Fang Yang
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Yunkui Liu
- State Key Laboratory Breeding Base of Green Chemistry-Synthesis Technology, College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| |
Collapse
|
3
|
Sun H, Li X, Chen M, Zhong M, Li Y, Wang K, Du Y, Zhen X, Gao R, Wu Y, Shi Y, Yu L, Che Y, Li Y, Jiang JD, Hong B, Si S. Multi-Omics-Guided Discovery of Omicsynins Produced by Streptomyces sp. 1647: Pseudo-Tetrapeptides Active Against Influenza A Viruses and Coronavirus HCoV-229E. ENGINEERING (BEIJING, CHINA) 2022; 16:176-186. [PMID: 35309096 PMCID: PMC8916927 DOI: 10.1016/j.eng.2021.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 05/06/2021] [Accepted: 05/16/2021] [Indexed: 06/14/2023]
Abstract
Many microorganisms have mechanisms that protect cells against attack from viruses. The fermentation components of Streptomyces sp. 1647 exhibit potent anti-influenza A virus (IAV) activity. This strain was isolated from soil in southern China in the 1970s, but the chemical nature of its antiviral substance(s) has remained unknown until now. We used an integrated multi-omics strategy to identify the antiviral agents from this streptomycete. The antibiotics and Secondary Metabolite Analysis Shell (antiSMASH) analysis of its genome sequence revealed 38 biosynthetic gene clusters (BGCs) for secondary metabolites, and the target BGCs possibly responsible for the production of antiviral components were narrowed down to three BGCs by bioactivity-guided comparative transcriptomics analysis. Through bioinformatics analysis and genetic manipulation of the regulators and a biosynthetic gene, cluster 36 was identified as the BGC responsible for the biosynthesis of the antiviral compounds. Bioactivity-based molecular networking analysis of mass spectrometric data from different recombinant strains illustrated that the antiviral compounds were a class of structural analogues. Finally, 18 pseudo-tetrapeptides with an internal ureido linkage, omicsynins A1-A6, B1-B6, and C1-C6, were identified and/or isolated from fermentation broth. Among them, 11 compounds (omicsynins A1, A2, A6, B1-B3, B5, B6, C1, C2, and C6) are new compounds. Omicsynins B1-B4 exhibited potent antiviral activity against IAV with the 50% inhibitory concentration (IC50) of approximately 1 µmol∙L-1 and a selectivity index (SI) ranging from 100 to 300. Omicsynins B1-B4 also showed significant antiviral activity against human coronavirus HCoV-229E. By integrating multi-omics data, we discovered a number of novel antiviral pseudo-tetrapeptides produced by Streptomyces sp. 1647, indicating that the secondary metabolites of microorganisms are a valuable source of novel antivirals.
Collapse
Affiliation(s)
- Hongmin Sun
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xingxing Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Minghua Chen
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ming Zhong
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yihua Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Kun Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yu Du
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xin Zhen
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Rongmei Gao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yexiang Wu
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuanyuan Shi
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Liyan Yu
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yongsheng Che
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuhuan Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jian-Dong Jiang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Bin Hong
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shuyi Si
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
4
|
Alexandrova LA, Khandazhinskaya AL, Matyugina ES, Makarov DA, Kochetkov SN. Analogues of Pyrimidine Nucleosides as Mycobacteria Growth Inhibitors. Microorganisms 2022; 10:microorganisms10071299. [PMID: 35889017 PMCID: PMC9322969 DOI: 10.3390/microorganisms10071299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 01/25/2023] Open
Abstract
Tuberculosis (TB) is the oldest human infection disease. Mortality from TB significantly decreased in the 20th century, because of vaccination and the widespread use of antibiotics. However, about a third of the world’s population is currently infected with Mycobacterium tuberculosis (Mtb) and the death rate from TB is about 1.4–2 million people per year. In the second half of the 20th century, new extensively multidrug-resistant strains of Mtb were identified, which are steadily increasing among TB patients. Therefore, there is an urgent need to develop new anti-TB drugs, which remains one of the priorities of pharmacology and medicinal chemistry. The antimycobacterial activity of nucleoside derivatives and analogues was revealed not so long ago, and a lot of studies on their antibacterial properties have been published. Despite the fact that there are no clinically used drugs based on nucleoside analogues, some progress has been made in this area. This review summarizes current research in the field of the design and study of inhibitors of mycobacteria, primarily Mtb.
Collapse
|
5
|
Tsunoda T, Tanoeyadi S, Proteau PJ, Mahmud T. The chemistry and biology of natural ribomimetics and related compounds. RSC Chem Biol 2022; 3:519-538. [PMID: 35656477 PMCID: PMC9092360 DOI: 10.1039/d2cb00019a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/06/2022] [Indexed: 11/21/2022] Open
Abstract
Natural ribomimetics represent an important group of specialized metabolites with significant biological activities. Many of the activities, e.g., inhibition of seryl-tRNA synthetases, glycosidases, or ribosomes, are manifestations of their structural resemblance to ribose or related sugars, which play roles in the structural, physiological, and/or reproductive functions of living organisms. Recent studies on the biosynthesis and biological activities of some natural ribomimetics have expanded our understanding on how they are made in nature and why they have great potential as pharmaceutically relevant products. This review article highlights the discovery, biological activities, biosynthesis, and development of this intriguing class of natural products.
Collapse
Affiliation(s)
- Takeshi Tsunoda
- Department of Pharmaceutical Sciences, Oregon State University Corvallis OR 97331 USA
| | - Samuel Tanoeyadi
- Department of Pharmaceutical Sciences, Oregon State University Corvallis OR 97331 USA
| | - Philip J Proteau
- Department of Pharmaceutical Sciences, Oregon State University Corvallis OR 97331 USA
| | - Taifo Mahmud
- Department of Pharmaceutical Sciences, Oregon State University Corvallis OR 97331 USA
| |
Collapse
|
6
|
Raji M, Le TM, Huynh T, Szekeres A, Nagy V, Zupkó I, Szakonyi Z. Divergent Synthesis, Antiproliferative and Antimicrobial Studies of 1,3-Aminoalcohol and 3-Amino-1,2-Diol Based Diaminopyrimidines. Chem Biodivers 2022; 19:e202200077. [PMID: 35349207 DOI: 10.1002/cbdv.202200077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/18/2022] [Indexed: 12/22/2022]
Abstract
A series of novel diaminopyrimidines containing pinane moieties were synthesized via an efficient methodology starting from pinane-based aminoalcohols, aminodiols and 2,4-dichloropyrimidines. Bioassay tests demonstrated that compound 18a displayed much stronger antiproliferative activities against four human cancer cell lines (HeLa, Siha, MDA-MB-231, MCF-7 and A2780) than positive control cisplatin. In particular, compound 22a was found to be selective in inhibiting HeLa cell proliferation with cancer cell growth inhibition values higher than 95 %. Moreover, the in vitro screening of prepared compounds against different bacterial and fungal strains is reported. The results revealed that 12b and 17a, the most promising compounds, displayed selective inhibition for the Gram-positive bacteria (B. subtilis and S. aureus) with percent inhibition values ranging from 75 to 95 % at 10 μg/mL concentration. Both selective inhibition and the in vitro activity values demonstrated that these compounds have the potential to be developed into clinically important therapeutic choices for the treatment of infections caused by B. subtilis and S. aureus.
Collapse
Affiliation(s)
- Mounir Raji
- Institute of Pharmaceutical Chemistry, University of Szeged, 6720, Szeged, Eötvös u. 6, Hungary
| | - Tam Minh Le
- Institute of Pharmaceutical Chemistry, University of Szeged, 6720, Szeged, Eötvös u. 6, Hungary.,Stereochemistry Research Group of the Hungarian Academy of Sciences, 6720, Szeged, Eötvös u. 6, Hungary
| | - Thu Huynh
- Department of Microbiology, University of Szeged, 6726, Szeged, Közép fasor 52, Hungary
| | - András Szekeres
- Department of Microbiology, University of Szeged, 6726, Szeged, Közép fasor 52, Hungary
| | - Viktória Nagy
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, 6720, Szeged, Eötvös utca 6, Hungary
| | - István Zupkó
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, 6720, Szeged, Eötvös utca 6, Hungary.,Interdisciplinary Center of Natural Products, University of Szeged, 6720, Szeged, Hungary
| | - Zsolt Szakonyi
- Institute of Pharmaceutical Chemistry, University of Szeged, 6720, Szeged, Eötvös u. 6, Hungary.,Interdisciplinary Center of Natural Products, University of Szeged, 6720, Szeged, Hungary
| |
Collapse
|
7
|
Pal S, Chandra G, Patel S, Singh S. Fluorinated Nucleosides: Synthesis, Modulation in Conformation and Therapeutic Application. CHEM REC 2022; 22:e202100335. [PMID: 35253973 DOI: 10.1002/tcr.202100335] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/22/2022] [Indexed: 12/17/2022]
Abstract
Over the last twenty years, fluorination on nucleoside has established itself as the most promising tool to use to get biologically active compounds that could sustain the clinical trial by affecting the pharmacodynamics and pharmacokinetic properties. Due to fluorine's inherent unique properties and its judicious introduction into the molecule, makes the corresponding nucleoside metabolically very stable, lipophilic, and opens a new site of intermolecular binding. Fluorination on various nucleosides has been extensively studied as a result, a series of fluorinated nucleosides come up for different therapeutic uses which are either approved by the FDA or under the advanced stage of the clinical trial. Here in this review, we are summarizing the latest development in the chemistry of fluorination on nucleoside that led to varieties of new analogs like carbocyclic, acyclic, and conformationally biased nucleoside and their biological properties, the influence of fluorine on conformation, oligonucleotide stability, and their use in therapeutics.
Collapse
Affiliation(s)
- Shantanu Pal
- School of Basic Sciences, Indian Institute of Technology, Bhubaneswar Argul, Odisha, India, 752050
| | - Girish Chandra
- Department of Chemistry, School of Physical and Chemical Sciences, Central University of South Bihar, SH-7, Gaya Panchanpur Road, Gaya, Bihar, India, 824236
| | - Samridhi Patel
- Department of Chemistry, School of Physical and Chemical Sciences, Central University of South Bihar, SH-7, Gaya Panchanpur Road, Gaya, Bihar, India, 824236
| | - Sakshi Singh
- School of Basic Sciences, Indian Institute of Technology, Bhubaneswar Argul, Odisha, India, 752050
| |
Collapse
|
8
|
Ojeda-Porras AC, Roy V, Agrofoglio LA. Chemical Approaches to Carbocyclic Nucleosides. CHEM REC 2022; 22:e202100307. [PMID: 35119191 DOI: 10.1002/tcr.202100307] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/22/2022] [Indexed: 02/02/2023]
Abstract
Nucleoside analogues are at the forefront of antiviral therapy for last decades. To circumvent some of their limitations, based on their metabolism, and in order to improve their anti-viral potency and selectivity, several families of nucleoside analogues have been described through structural modifications at the sugar and heterocycles. The replacement of the oxygen of the nucleoside by a methylene has led to the family of carbocyclic (or cyclopentane) nucleoside analogues. Various potent anti-HIV and anti-HBV drugs belong to this family. Main syntheses of carbocyclic analogues of nucleosides used Diels-Alder reactions (in racemic or asymmetric series), but also started from carbohydrates (ribose, glucose), as a source of optically active compounds, which then had to be transformed into carbacycles under various conditions. The growing interest in carbocyclic nucleosides has led several groups, including ours, to develop new analogues and to explore novel routes. This article will review some of the recent chemistry developed on selected five-membered ring carbocyclic nucleosides.
Collapse
Affiliation(s)
- Andrea C Ojeda-Porras
- ICOA, Univ. Orléans, CNRS UMR 7311, Université d'Orléans, Rue de Chartres, 45067, Orléans Cedex 2, France
| | - Vincent Roy
- ICOA, Univ. Orléans, CNRS UMR 7311, Université d'Orléans, Rue de Chartres, 45067, Orléans Cedex 2, France
| | - Luigi A Agrofoglio
- ICOA, Univ. Orléans, CNRS UMR 7311, Université d'Orléans, Rue de Chartres, 45067, Orléans Cedex 2, France
| |
Collapse
|
9
|
Barik R, Halder J, Jana P, Nanda S. Stereoselective synthesis of novel carbocyclic and heterocyclic scaffolds of medicinal importance from biocatalytically derived enantiopure α-substituted-β-hydroxy esters. Tetrahedron 2021. [DOI: 10.1016/j.tet.2021.132356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
10
|
Shiomi K. Antiparasitic antibiotics from Japan. Parasitol Int 2021; 82:102298. [PMID: 33548522 DOI: 10.1016/j.parint.2021.102298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/19/2020] [Accepted: 01/27/2021] [Indexed: 11/29/2022]
Abstract
Antibiotics are microbial secondary metabolites and they are important for the treatment of infectious diseases. Japanese researchers have made a large contribution to studies of antibiotics, and they have also been important in the discovery of antiparasitic antibiotics. Satoshi Ōmura received the Nobel Prize in 2015 for the "discoveries concerning a novel therapy against infections caused by roundworm parasites", which means discovery of a new nematocidal antibiotic, avermectin. Here, I review the many antiparasitic antibiotics and their lead compounds that have been discovered for use in human and veterinary medicine.
Collapse
Affiliation(s)
- Kazuro Shiomi
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan.
| |
Collapse
|
11
|
Pang L, Nautiyal M, De Graef S, Gadakh B, Zorzini V, Economou A, Strelkov SV, Van Aerschot A, Weeks SD. Structural Insights into the Binding of Natural Pyrimidine-Based Inhibitors of Class II Aminoacyl-tRNA Synthetases. ACS Chem Biol 2020; 15:407-415. [PMID: 31869198 DOI: 10.1021/acschembio.9b00887] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The pyrimidine-containing Trojan horse antibiotics albomycin and a recently discovered cytidine-containing microcin C analog target the class II seryl- and aspartyl-tRNA synthetases (serRS and aspRS), respectively. The active components of these compounds are competitive inhibitors that mimic the aminoacyl-adenylate intermediate. How they effectively substitute for the interactions mediated by the canonical purine group is unknown. Employing nonhydrolyzable aminoacyl-sulfamoyl nucleosides substituting the base with cytosine, uracil, and N3-methyluracil the structure-activity relationship of the natural compounds was evaluated. In vitro using E. coli serRS and aspRS, the best compounds demonstrated IC50 values in the low nanomolar range, with a clear preference for cytosine or N3-methyluracil over uracil. X-ray crystallographic structures of K. pneumoniae serRS and T. thermophilus aspRS in complex with the compounds showed the contribution of structured waters and residues in the conserved motif-2 loop in defining base preference. Utilizing the N3-methyluracil bound serRS structure, MD simulations of the fully modified albomycin base were performed to identify the interacting network that drives stable association. This analysis pointed to key interactions with a methionine in the motif-2 loop. Interestingly, this residue is mutated to a glycine in a second serRS (serRS2) found in albomycin-producing actinobacteria possessing self-immunity to this antibiotic. A comparative study demonstrated that serRS2 is poorly inhibited by the pyrimidine-containing intermediate analogs, and an equivalent mutation in E. coli serRS significantly decreased the affinity of the cytosine congener. These findings highlight the crucial role of dynamics and solvation of the motif-2 loop in modulating the binding of the natural antibiotics.
Collapse
Affiliation(s)
- Luping Pang
- Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49 Box 822, B-3000 Leuven, Belgium
- Medicinal Chemistry, Rega Institute for Medical Research, Herestraat 49 Box 1041, B-3000 Leuven, Belgium
| | - Manesh Nautiyal
- Medicinal Chemistry, Rega Institute for Medical Research, Herestraat 49 Box 1041, B-3000 Leuven, Belgium
| | - Steff De Graef
- Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49 Box 822, B-3000 Leuven, Belgium
| | - Bharat Gadakh
- Medicinal Chemistry, Rega Institute for Medical Research, Herestraat 49 Box 1041, B-3000 Leuven, Belgium
| | - Valentina Zorzini
- Laboratory for Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Herestraat 49, Gasthuisberg Campus, B-3000 Leuven, Belgium
| | - Anastassios Economou
- Laboratory for Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Herestraat 49, Gasthuisberg Campus, B-3000 Leuven, Belgium
| | - Sergei V. Strelkov
- Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49 Box 822, B-3000 Leuven, Belgium
| | - Arthur Van Aerschot
- Medicinal Chemistry, Rega Institute for Medical Research, Herestraat 49 Box 1041, B-3000 Leuven, Belgium
| | - Stephen D. Weeks
- Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49 Box 822, B-3000 Leuven, Belgium
| |
Collapse
|
12
|
Chen SJ, Chen GS, Zhang JW, Li ZD, Zhao YL, Liu YL. Phosphine-catalyzed [3 + 2] cycloadditions of trifluoromethyl enynes/enediynes with allenoates: access to cyclopentenes containing a CF3-substituted quaternary carbon center. Org Chem Front 2020. [DOI: 10.1039/d0qo00807a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The first use of trifluoromethyl-substituted enynes/enediynes as non-classical electron-deficient olefins for phosphine-catalyzed [3 + 2] cycloaddition with allenoates is presented.
Collapse
Affiliation(s)
- Shu-Jie Chen
- School of Chemistry and Chemical Engineering
- Guangzhou University
- Guangzhou
- China
| | - Guo-Shu Chen
- School of Chemistry and Chemical Engineering
- Guangzhou University
- Guangzhou
- China
| | - Jia-Wei Zhang
- College of Materials Science and Engineering
- Hunan University
- Changsha
- China
| | - Zhao-Dong Li
- Department of Applied Chemistry
- College of Materials and Energy
- South China Agricultural University
- Guangzhou
- China
| | - Yu-Lei Zhao
- School of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu
- China
| | - Yun-Lin Liu
- School of Chemistry and Chemical Engineering
- Guangzhou University
- Guangzhou
- China
| |
Collapse
|
13
|
Kudo F, Tsunoda T, Yamaguchi K, Miyanaga A, Eguchi T. Stereochemistry in the Reaction of the myo-Inositol Phosphate Synthase Ortholog Ari2 during Aristeromycin Biosynthesis. Biochemistry 2019; 58:5112-5116. [PMID: 31825604 DOI: 10.1021/acs.biochem.9b00981] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The myo-inositol-1-phosphate synthase (MIPS) ortholog Ari2, which is encoded in the aristeromycin biosynthetic gene cluster, catalyzes the formation of five-membered cyclitol phosphate using d-fructose 6-phosphate (F6P) as a substrate. To understand the stereochemistry during the Ari2 reaction in vivo, we carried out feeding experiments with (6S)-d-[6-2H1]- and (6R)-d-[6-2H1]glucose in the aristeromycin-producing strain Streptomyces citricolor. We observed retention of the 2H atom of (6S)-d-[6-2H1]glucose and no incorporation of the 2H atom from (6R)-d-[6-2H1]glucose in aristeromycin. This indicates that Ari2 abstracts the pro-R proton at C6 of F6P after oxidation of C5-OH by nicotinamide adenine dinucleotide (NAD+) to generate the enolate intermediate, which then attacks the C2 ketone to form the C-C bond via aldol-type condensation. The reaction of Ari2 with (6S)-d-[6-2H1]- and (6R)-d-[6-2H1]F6P in vitro exhibited identical stereochemistry compared with that observed during the feeding experiments. Furthermore, analysis of the crystal structure of Ari2, including NAD+ as a ligand, revealed the active site of Ari2 to be similar to that of MIPS of Mycobacterium tuberculosis, supporting the similarity of the reaction mechanisms of Ari2 and MIPS.
Collapse
Affiliation(s)
- Fumitaka Kudo
- Department of Chemistry , Tokyo Institute of Technology , 2-12-1, O-okayama , Meguro-ku, Tokyo 152-8551 , Japan
| | - Takeshi Tsunoda
- Department of Chemistry , Tokyo Institute of Technology , 2-12-1, O-okayama , Meguro-ku, Tokyo 152-8551 , Japan
| | - Kaito Yamaguchi
- Department of Chemistry , Tokyo Institute of Technology , 2-12-1, O-okayama , Meguro-ku, Tokyo 152-8551 , Japan
| | - Akimasa Miyanaga
- Department of Chemistry , Tokyo Institute of Technology , 2-12-1, O-okayama , Meguro-ku, Tokyo 152-8551 , Japan
| | - Tadashi Eguchi
- Department of Chemistry , Tokyo Institute of Technology , 2-12-1, O-okayama , Meguro-ku, Tokyo 152-8551 , Japan
| |
Collapse
|
14
|
Shin YS, Jarhad DB, Jang MH, Kovacikova K, Kim G, Yoon JS, Kim HR, Hyun YE, Tipnis AS, Chang TS, van Hemert MJ, Jeong LS. Identification of 6'-β-fluoro-homoaristeromycin as a potent inhibitor of chikungunya virus replication. Eur J Med Chem 2019; 187:111956. [PMID: 31841728 PMCID: PMC7115507 DOI: 10.1016/j.ejmech.2019.111956] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/06/2019] [Indexed: 11/27/2022]
Abstract
We have reported on aristeromycin (1) and 6′-fluorinated-aristeromycin analogues (2), which are active against RNA viruses such as Middle East respiratory syndrome coronavirus (MERS-CoV), severe acute respiratory syndrome coronavirus (SARS-CoV), Zika virus (ZIKV), and Chikungunya virus (CHIKV). However, these exhibit substantial cytotoxicity. As this cytotoxicity may be attributed to 5′-phosphorylation, we designed and synthesized one-carbon homologated 6′-fluorinated-aristeromycin analogues. This modification prevents 5′-phosphorlyation by cellular kinases, whereas the inhibitory activity towards S-adenosyl-l-homocysteine (SAH) hydrolase will be retained. The enantiomerically pure 6′-fluorinated-5′-homoaristeromycin analogues 3a-e were synthesized via the electrophilic fluorination of the silyl enol ether with Selectfluor, using a base-build up approach as the key steps. All synthesized compounds exhibited potent inhibitory activity towards SAH hydrolase, among which 6′-β-fluoroadenosine analogue 3a was the most potent (IC50 = 0.36 μM). Among the compounds tested, 6′-β-fluoro-homoaristeromycin 3a showed potent antiviral activity (EC50 = 0.12 μM) against the CHIKV, without noticeable cytotoxicity up to 250 μM. Only 3a displayed anti-CHIKV activity, whereas both3a and 3b inhibited SAH hydrolase with similar IC50 values (0.36 and 0.37 μM, respectively), which suggested that 3a’s antiviral activity did not merely depend on the inhibition of SAH hydrolase. This is further supported by the fact that the antiviral effect was specific for CHIKV and some other alphaviruses and none of the homologated analogues inhibited other RNA viruses, such as SARS-CoV, MERS-CoV, and ZIKV. The potent inhibition and high selectivity index make 6′-β-fluoro-homoaristeromycin (3a) a promising new template for the development of antivirals against CHIKV, a serious re-emerging pathogen that has infected millions of people over the past 15 years.
Collapse
Affiliation(s)
- Young Sup Shin
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Dnyandev B Jarhad
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Min Hwan Jang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Kristina Kovacikova
- Department of Medical Microbiology, Leiden University Medical Center, Albinusdreef 2, 2333ZA, Leiden, the Netherlands
| | - Gyudong Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Ji-Seong Yoon
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Hong-Rae Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Young Eum Hyun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Amol S Tipnis
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Tong-Shin Chang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Martijn J van Hemert
- Department of Medical Microbiology, Leiden University Medical Center, Albinusdreef 2, 2333ZA, Leiden, the Netherlands
| | - Lak Shin Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
15
|
Yoon JS, Kim G, Jarhad DB, Kim HR, Shin YS, Qu S, Sahu PK, Kim HO, Lee HW, Wang SB, Kong YJ, Chang TS, Ogando NS, Kovacikova K, Snijder EJ, Posthuma CC, van Hemert MJ, Jeong LS. Design, Synthesis, and Anti-RNA Virus Activity of 6'-Fluorinated-Aristeromycin Analogues. J Med Chem 2019; 62:6346-6362. [PMID: 31244113 PMCID: PMC7075649 DOI: 10.1021/acs.jmedchem.9b00781] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The 6'-fluorinated aristeromycins were designed as dual-target antiviral compounds aimed at inhibiting both the viral RNA-dependent RNA polymerase (RdRp) and the host cell S-adenosyl-l-homocysteine (SAH) hydrolase, which would indirectly target capping of viral RNA. The introduction of a fluorine at the 6'-position enhanced the inhibition of SAH hydrolase and the activity against RNA viruses. The adenosine and N6-methyladenosine analogues 2a-e showed potent inhibition against SAH hydrolase, while only the adenosine derivatives 2a-c exhibited potent antiviral activity against all tested RNA viruses such as Middle East respiratory syndrome-coronavirus (MERS-CoV), severe acute respiratory syndrome-coronavirus, chikungunya virus, and/or Zika virus. 6',6'-Difluoroaristeromycin (2c) showed the strongest antiviral effect for MERS-CoV, with a ∼2.5 log reduction in infectious progeny titer in viral load reduction assay. The phosphoramidate prodrug 3a also demonstrated potent broad-spectrum antiviral activity, possibly by inhibiting the viral RdRp. This study shows that 6'-fluorinated aristeromycins can serve as starting points for the development of broad-spectrum antiviral agents that target RNA viruses.
Collapse
Affiliation(s)
- Ji-Seong Yoon
- Research Institute of Pharmaceutical Sciences, College of Pharmacy , Seoul National University , Seoul 151-742 , Korea
| | - Gyudong Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy , Seoul National University , Seoul 151-742 , Korea.,College of Pharmacy and Research Institute of Drug Development , Chonnam National University , Gwangju 500-757 , Korea
| | - Dnyandev B Jarhad
- Research Institute of Pharmaceutical Sciences, College of Pharmacy , Seoul National University , Seoul 151-742 , Korea
| | - Hong-Rae Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy , Seoul National University , Seoul 151-742 , Korea
| | - Young-Sup Shin
- Research Institute of Pharmaceutical Sciences, College of Pharmacy , Seoul National University , Seoul 151-742 , Korea
| | - Shuhao Qu
- Research Institute of Pharmaceutical Sciences, College of Pharmacy , Seoul National University , Seoul 151-742 , Korea.,College of Pharmaceutical Engineering , Henan University of Animal Husbandry and Economy , Zhengzhou , 450046 , China
| | | | - Hea Ok Kim
- Future Medicine Co., Ltd. , Seoul 06665 , Korea
| | | | - Su Bin Wang
- College of Pharmacy , Ewha Womans University , Seoul 120-750 , Korea
| | - Yun Jeong Kong
- College of Pharmacy , Ewha Womans University , Seoul 120-750 , Korea
| | - Tong-Shin Chang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy , Seoul National University , Seoul 151-742 , Korea.,College of Pharmacy , Ewha Womans University , Seoul 120-750 , Korea
| | - Natacha S Ogando
- Department of Medical Microbiology , Leiden University Medical Center , Albinusdreef 2 , 2333ZA Leiden , The Netherlands
| | - Kristina Kovacikova
- Department of Medical Microbiology , Leiden University Medical Center , Albinusdreef 2 , 2333ZA Leiden , The Netherlands
| | - Eric J Snijder
- Department of Medical Microbiology , Leiden University Medical Center , Albinusdreef 2 , 2333ZA Leiden , The Netherlands
| | - Clara C Posthuma
- Department of Medical Microbiology , Leiden University Medical Center , Albinusdreef 2 , 2333ZA Leiden , The Netherlands
| | - Martijn J van Hemert
- Department of Medical Microbiology , Leiden University Medical Center , Albinusdreef 2 , 2333ZA Leiden , The Netherlands
| | - Lak Shin Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy , Seoul National University , Seoul 151-742 , Korea
| |
Collapse
|
16
|
Dynamic kinetic resolution of Vince lactam catalyzed by γ-lactamases: a mini-review. ACTA ACUST UNITED AC 2018; 45:1017-1031. [DOI: 10.1007/s10295-018-2093-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 10/16/2018] [Indexed: 10/28/2022]
Abstract
Abstract
γ-Lactamases are versatile enzymes used for enzymatic kinetic resolution of racemic Vince lactam (2-azabicyclo[2.2.1]hept-5-en-3-one) in the industry. Optically pure enantiomers and their hydrolytic products are widely employed as key chemical intermediates for developing a wide range of carbocyclic nucleoside medicines, including US FDA-approved drugs peramivir and abacavir. Owing to the broad applications in the healthcare industry, the resolution process of Vince lactam has witnessed tremendous progress during the past decades. Some of the most important advances are the enzymatic strategies involving γ-lactamases. The strong industrial demand drives the progress in various strategies for discovering novel biocatalysts. In the past few years, several new scientific breakthroughs, including the genome-mining strategy and elucidation of several crystal structures, boosted the research on γ-lactamases. So far, several families of γ-lactamases for resolution of Vince lactam have been discovered, and their number is continuously increasing. The purpose of this mini-review is to describe the discovery strategy and classification of these intriguing enzymes and to cover our current knowledge on their potential biological functions. Moreover, structural properties are described in addition to their possible catalytic mechanisms. Additionally, recent advances in the newest approaches, such as immobilization to increase stability, and other engineering efforts are introduced.
Collapse
|
17
|
Khandazhinskaya AL, Alexandrova LA, Matyugina ES, Solyev PN, Efremenkova OV, Buckheit KW, Wilkinson M, Buckheit RW, Chernousova LN, Smirnova TG, Andreevskaya SN, Leonova OG, Popenko VI, Kochetkov SN, Seley-Radtke KL. Novel 5'-Norcarbocyclic Pyrimidine Derivatives as Antibacterial Agents. Molecules 2018; 23:E3069. [PMID: 30477147 PMCID: PMC6321083 DOI: 10.3390/molecules23123069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 11/16/2022] Open
Abstract
A series of novel 5'-norcarbocyclic derivatives of 5-alkoxymethyl or 5-alkyltriazolyl-methyl uracil were synthesized and the activity of the compounds evaluated against both Gram-positive and Gram-negative bacteria. The growth of Mycobacterium smegmatis was completely inhibited by the most active compounds at a MIC99 of 67 μg/mL (mc²155) and a MIC99 of 6.7⁻67 μg/mL (VKPM Ac 1339). Several compounds also showed the ability to inhibit the growth of attenuated strains of Mycobacterium tuberculosis ATCC 25177 (MIC99 28⁻61 μg/mL) and Mycobacterium bovis ATCC 35737 (MIC99 50⁻60 μg/mL), as well as two virulent strains of M. tuberculosis; a laboratory strain H37Rv (MIC99 20⁻50 μg/mL) and a clinical strain with multiple drug resistance MS-115 (MIC99 20⁻50 μg/mL). Transmission electron microscopy (TEM) evaluation of M. tuberculosis H37Rv bacterial cells treated with one of the compounds demonstrated destruction of the bacterial cell wall, suggesting that the mechanism of action for these compounds may be related to their interactions with bacteria cell walls.
Collapse
Affiliation(s)
- Anastasia L Khandazhinskaya
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., Moscow 119991, Russia.
| | - Liudmila A Alexandrova
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., Moscow 119991, Russia.
| | - Elena S Matyugina
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., Moscow 119991, Russia.
| | - Pavel N Solyev
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., Moscow 119991, Russia.
| | - Olga V Efremenkova
- Gause Institute of New Antibiotics, 11 Bol'shaya Pirogovskaya St., Moscow 119021, Russia.
| | - Karen W Buckheit
- ImQuest BioSciences, 7340 Executive Way Suite R, Frederick, MD 21704, USA.
| | - Maggie Wilkinson
- ImQuest BioSciences, 7340 Executive Way Suite R, Frederick, MD 21704, USA.
| | - Robert W Buckheit
- ImQuest BioSciences, 7340 Executive Way Suite R, Frederick, MD 21704, USA.
| | - Larisa N Chernousova
- Central Tuberculosis Research Institute, 2 Yauzskaya Alley, Moscow 107564, Russia.
| | - Tatiana G Smirnova
- Central Tuberculosis Research Institute, 2 Yauzskaya Alley, Moscow 107564, Russia.
| | - Sofya N Andreevskaya
- Central Tuberculosis Research Institute, 2 Yauzskaya Alley, Moscow 107564, Russia.
| | - Olga G Leonova
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., Moscow 119991, Russia.
| | - Vladimir I Popenko
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., Moscow 119991, Russia.
| | - Sergey N Kochetkov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 32 Vavilov St., Moscow 119991, Russia.
| | - Katherine L Seley-Radtke
- Department of Chemistry & Biochemistry, University of Maryland, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
18
|
Coordinated Biosynthesis of the Purine Nucleoside Antibiotics Aristeromycin and Coformycin in Actinomycetes. Appl Environ Microbiol 2018; 84:AEM.01860-18. [PMID: 30217843 DOI: 10.1128/aem.01860-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 09/04/2018] [Indexed: 02/05/2023] Open
Abstract
Purine nucleoside antibiotic pairs, concomitantly produced by a single strain, are an important group of microbial natural products. Here, we report a target-directed genome mining approach to elucidate the biosynthesis of the purine nucleoside antibiotic pair aristeromycin (ARM) and coformycin (COF) in Micromonospora haikouensis DSM 45626 (a new producer for ARM and COF) and Streptomyces citricolor NBRC 13005 (a new COF producer). We also provide biochemical data that MacI and MacT function as unusual phosphorylases, catalyzing an irreversible reaction for the tailoring assembly of neplanocin A (NEP-A) and ARM. Moreover, we demonstrate that MacQ is shown to be an adenosine-specific deaminase, likely relieving the potential "excess adenosine" for producing cells. Finally, we report that MacR, an annotated IMP dehydrogenase, is actually an NADPH-dependent GMP reductase, which potentially plays a salvage role for the efficient supply of the precursor pool. Hence, these findings illustrate a fine-tuned pathway for the biosynthesis of ARM and also open the way for the rational search for purine antibiotic pairs.IMPORTANCE ARM and COF are well known for their prominent biological activities and unusual chemical structures; however, the logic of their biosynthesis has long been poorly understood. Actually, the new insights into the ARM and COF pathway will not only enrich the biochemical repertoire for interesting enzymatic reactions but may also lay a solid foundation for the combinatorial biosynthesis of this group of antibiotics via a target-directed genome mining strategy.
Collapse
|
19
|
Kim G, Yoon JS, Jarhad DB, Shin YS, Majik MS, Mulamoottil VA, Hou X, Qu S, Park J, Baik MH, Jeong LS. Asymmetric Synthesis of (-)-6'-β-Fluoro-aristeromycin via Stereoselective Electrophilic Fluorination. Org Lett 2018; 19:5732-5735. [PMID: 29028350 DOI: 10.1021/acs.orglett.7b02470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
(-)-6'-β-Fluoro-aristeromycin (2), a potent inhibitor of S-adenosylhomocysteine (AdoHcy) hydrolase, has been synthesized via stereoselective electrophilic fluorination followed by a purine base build-up approach. Interestingly, purine base condensation using a cyclic sulfate resulted in a synthesis of (+)-5'-β-fluoro-isoaristeromycin (2a). Computational analysis indicates that the fluorine atom controlled the regioselectivity of the purine base substitution.
Collapse
Affiliation(s)
- Gyudong Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, 151-742, Korea
| | - Ji-Seong Yoon
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, 151-742, Korea
| | - Dnyandev B Jarhad
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, 151-742, Korea
| | - Young Sup Shin
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, 151-742, Korea
| | - Mahesh S Majik
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, 151-742, Korea
| | - Varughese A Mulamoottil
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, 151-742, Korea
| | - Xiyan Hou
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, 151-742, Korea
| | - Shuhao Qu
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, 151-742, Korea
| | - Jiyong Park
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS) , Daejeon, 34141, Korea
| | - Mu-Hyun Baik
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS) , Daejeon, 34141, Korea
| | - Lak Shin Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, 151-742, Korea
| |
Collapse
|
20
|
Chen Q, Liu C, Bowlin TL, Schneller SW. 5'-Hydroxy-5'-homoaristeromycin: Synthesis and antiviral properties. Bioorg Med Chem Lett 2018; 28:1456-1458. [PMID: 29628328 PMCID: PMC7126772 DOI: 10.1016/j.bmcl.2018.03.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 03/30/2018] [Indexed: 11/29/2022]
Abstract
Synthetically combining the C-4′ side-chain structural features of the antiviral candidates 5′-methylaristeromycin and 5′-homoaristeromycin into a diastereomeric pair of C-4′ side-chain dihydroxylated aristeromycins (6 and 7) is reported. Broad antiviral analyses of the both targets found promising effects towards HBV (6, 6.7 μM and 7, 7.74 μM) and HCMV (only 7, 0.72 μM). No other activity was found. Neither of the diastereomers was cytotoxic in the assays performed.
Collapse
Affiliation(s)
- Qi Chen
- Molette Laboratory for Drug Discovery, Department of Chemistry and Biochemistry, Auburn University, Auburn, AL 36849-5312, United States; Department of Chemistry, Slippery Rock University, Slippery Rock, PA 16057, United States
| | - Chong Liu
- Molette Laboratory for Drug Discovery, Department of Chemistry and Biochemistry, Auburn University, Auburn, AL 36849-5312, United States
| | - Terry L Bowlin
- Microbiotix, Inc., One Innovation Drive, Worcester, MA 01605, United States
| | - Stewart W Schneller
- Molette Laboratory for Drug Discovery, Department of Chemistry and Biochemistry, Auburn University, Auburn, AL 36849-5312, United States.
| |
Collapse
|
21
|
Li H, Zhu S, Zheng G. Promiscuous (+)-γ-lactamase activity of an amidase from nitrile hydratase pathway for efficient synthesis of carbocyclic nucleosides intermediate. Bioorg Med Chem Lett 2018; 28:1071-1076. [DOI: 10.1016/j.bmcl.2018.02.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/04/2018] [Accepted: 02/09/2018] [Indexed: 11/17/2022]
|
22
|
Abstract
Objective To synthesize 3,7-dideazaneplanocin and evaluate its antiviral potential. Methods The target 3,7-dideazaneplanocin has been prepared in five steps from a readily available cyclopentenol. A thorough in vitro antiviral analysis was conducted versus both DNA and RNA viruses. Results A rational synthesis of 3,7-dideazaneplanocin was conceived and successfully pursued in such a way that it can be adapted to various analogs of 3,7-dideazaneplanocin. Using standard antiviral assays, no activity for 3,7-dideazaneplanocn was found. Conclusion Two structural features are necessary for adenine-based carbocyclic nucleosides (like neplanocin) for potential antiviral properties: (i) inhibition of S-adenosylhomocysteine hydrolase and/or (ii) C-5′ activation via the mono-nucleotide. These two requisite adenine structural features to fit these criteria are not present in in the target 3,7-dideazaneplanocin: (i) an N-7 is necessary for inhibition of the hydrolase and the N-3 is claimed to be essential for phosphorylation at C-5′. Thus, it is not surprising that 3,7-dideazaneplaoncin lacked antiviral properties.
Collapse
Affiliation(s)
- Xue-Qiang Yin
- Department of Chemistry and Biochemistry, Molette Laboratory for Drug Discovery, Auburn University, Auburn, USA
| | - Stewart W Schneller
- Department of Chemistry and Biochemistry, Molette Laboratory for Drug Discovery, Auburn University, Auburn, USA
| |
Collapse
|
23
|
Evaluation of the antiprotozoan properties of 5′-norcarbocyclic pyrimidine nucleosides. Bioorg Med Chem Lett 2017; 27:3081-3086. [DOI: 10.1016/j.bmcl.2017.05.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 12/11/2022]
|
24
|
Kudo F, Tsunoda T, Takashima M, Eguchi T. Five-Membered Cyclitol Phosphate Formation by a myo-Inositol Phosphate Synthase Orthologue in the Biosynthesis of the Carbocyclic Nucleoside Antibiotic Aristeromycin. Chembiochem 2016; 17:2143-2148. [PMID: 27577857 DOI: 10.1002/cbic.201600348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Indexed: 12/21/2022]
Abstract
Aristeromycin is a unique carbocyclic nucleoside antibiotic produced by Streptomyces citricolor. In order to elucidate its intriguing carbocyclic formation, we used a genome-mining approach to identify the responsible enzyme. In silico screening with known cyclitol synthases involved in primary metabolism, such as myo-inositol-1-phosphate synthase (MIPS) and dehydroqunate synthase (DHQS), identified a unique MIPS orthologue (Ari2) encoded in the genome of S. citricolor. Heterologous expression of the gene cluster containing ari2 with a cosmid vector in Streptomyces albus resulted in the production of aristeromycin, thus indicating that the cloned DNA region (37.5 kb) with 33 open reading frames contains its biosynthetic gene cluster. We verified that Ari2 catalyzes the formation of a novel five-membered cyclitol phosphate from d-fructose 6-phosphate (F6P) with NAD+ as a cofactor. This provides insight into cyclitol phosphate synthase as a member of the MIPS family of enzymes. A biosynthetic pathway to aristeromycin is proposed based on bioinformatics analysis of the gene cluster.
Collapse
Affiliation(s)
- Fumitaka Kudo
- Department of Chemistry, Tokyo Institute of Technology, 2-12-1 O-Okayama, Meguro-ku, Tokyo, 152-8551 (, Japan)
| | - Takeshi Tsunoda
- Department of Chemistry, Tokyo Institute of Technology, 2-12-1 O-Okayama, Meguro-ku, Tokyo, 152-8551 (, Japan)
| | - Makoto Takashima
- Department of Chemistry, Tokyo Institute of Technology, 2-12-1 O-Okayama, Meguro-ku, Tokyo, 152-8551 (, Japan)
| | - Tadashi Eguchi
- Department of Chemistry, Tokyo Institute of Technology, 2-12-1 O-Okayama, Meguro-ku, Tokyo, 152-8551 (, Japan)
| |
Collapse
|
25
|
Serpi M, Ferrari V, Pertusati F. Nucleoside Derived Antibiotics to Fight Microbial Drug Resistance: New Utilities for an Established Class of Drugs? J Med Chem 2016; 59:10343-10382. [PMID: 27607900 DOI: 10.1021/acs.jmedchem.6b00325] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Novel antibiotics are urgently needed to combat the rise of infections due to drug-resistant microorganisms. Numerous natural nucleosides and their synthetically modified analogues have been reported to have moderate to good antibiotic activity against different bacterial and fungal strains. Nucleoside-based compounds target several crucial processes of bacterial and fungal cells such as nucleoside metabolism and cell wall, nucleic acid, and protein biosynthesis. Nucleoside analogues have also been shown to target many other bacterial and fungal cellular processes although these are not well characterized and may therefore represent opportunities to discover new drugs with unique mechanisms of action. In this Perspective, we demonstrate that nucleoside analogues, cornerstones of anticancer and antiviral treatments, also have great potential to be repurposed as antibiotics so that an old drug can learn new tricks.
Collapse
Affiliation(s)
- Michaela Serpi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University , Redwood Building, King Edward VII Avenue, CF10 3NB Cardiff, United Kingdom
| | - Valentina Ferrari
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University , Redwood Building, King Edward VII Avenue, CF10 3NB Cardiff, United Kingdom
| | - Fabrizio Pertusati
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University , Redwood Building, King Edward VII Avenue, CF10 3NB Cardiff, United Kingdom
| |
Collapse
|
26
|
Kasula M, Toyama M, Samunuri R, Rozy F, Yadav M, Bal C, Jha AK, Baba M, Sharon A. Synthesis and anti-HBV activity of α-stereoisomer of aristeromycin based analogs. Bioorg Med Chem Lett 2016; 26:3945-9. [PMID: 27426303 DOI: 10.1016/j.bmcl.2016.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 06/30/2016] [Accepted: 07/02/2016] [Indexed: 10/21/2022]
Abstract
The potential antiviral activity of aristeromycin type of derivatives (I) is limited by associated toxicity due to its possible 5'-O-phosphorylation and S-adenosyl-l-homocysteine hydrolase (SAHase) inhibitory activity. Aristeromycin structure has major pharmacophoric motif as 5'-OH and adenosine base, which may have significant role in enzyme binding followed by activity and or toxicity. Thus, the structural optimization to alter this major motif by replacing with its bioisostere and changing the 5'-O conformation through stereochemistry reversal was of interest. Thus, the inverted stereochemistry at 4'-position coupled with bioisostere of adenosine base in the target compounds (6-7) to access antiviral potential. The stereoselective formation of a key stereoisomer (2a) was achieved exclusively from neplanocin sugar (1a) by reduction in a single step. The novel target molecules (6-7) were synthesized in 4 steps with 55-62% yield. Compound 6 was analyzed by single crystal X-ray diffraction, which confirms the stereoselective formation of α-analogs with highly puckered cyclopentane ring and 2'-endo conformation. The compound 6 shown significant anti-hepatitis B virus activity of 6.5μM with CC50>100μM and yielded a promising lead with novel structural feature.
Collapse
Affiliation(s)
- Mohan Kasula
- Department of Chemistry, Birla Institute of Technology, Mesra, Ranchi 835215, India
| | - Masaaki Toyama
- Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8544, Japan
| | | | - Farhana Rozy
- Department of Chemistry, Birla Institute of Technology, Mesra, Ranchi 835215, India
| | - Monika Yadav
- Department of Chemistry, Birla Institute of Technology, Mesra, Ranchi 835215, India
| | - Chandralata Bal
- Department of Chemistry, Birla Institute of Technology, Mesra, Ranchi 835215, India
| | - Ashok Kumar Jha
- Chemistry Services, GVK Biosciences Pvt, Ltd, IDA Uppal, Hyderabad 500039, India
| | - Masanori Baba
- Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8544, Japan
| | - Ashoke Sharon
- Department of Chemistry, Birla Institute of Technology, Mesra, Ranchi 835215, India
| |
Collapse
|
27
|
1′-Homonucleosides and their structural analogues: A review. Eur J Med Chem 2016; 118:121-42. [DOI: 10.1016/j.ejmech.2016.04.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 04/12/2016] [Accepted: 04/12/2016] [Indexed: 11/18/2022]
|
28
|
You C, Wei B, Li X, Yang Y, Liu Y, Lv H, Zhang X. Rhodium-Catalyzed Desymmetrization by Hydroformylation of Cyclopentenes: Synthesis of Chiral Carbocyclic Nucleosides. Angew Chem Int Ed Engl 2016; 55:6511-4. [PMID: 27086543 DOI: 10.1002/anie.201601478] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Cai You
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Biao Wei
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Xiuxiu Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Yusheng Yang
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Yue Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Hui Lv
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Xumu Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| |
Collapse
|
29
|
You C, Wei B, Li X, Yang Y, Liu Y, Lv H, Zhang X. Rhodium-Catalyzed Desymmetrization by Hydroformylation of Cyclopentenes: Synthesis of Chiral Carbocyclic Nucleosides. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201601478] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Cai You
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Biao Wei
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Xiuxiu Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Yusheng Yang
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Yue Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Hui Lv
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| | - Xumu Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & College of Chemistry and Molecular Sciences; Wuhan University; Wuhan Hubei 430072 China
| |
Collapse
|
30
|
Kasula M, Samunuri R, Chakravarty H, Bal C, Baba M, Jha AK, Sharon A. Regioselective Synthesis of Pyrazolo[3,4-D]Pyrimidine Based Carbocyclic Nucleosides as Possible Antiviral Agent. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2016; 35:43-52. [PMID: 26810255 DOI: 10.1080/15257770.2015.1114126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Carbocyclic nucleosides are considered as nucleoside mimetic having high therapeutic potentials, however diverse exploration is still limited due to their synthetic difficulties. The major challenges are associated with the preparation of new base and carbocyclic sugar key intermediates. The modified base may provide conformational advantage to achieve better nucleoside mimetics and may also help in increasing the drug-like properties. In this manuscript, we report the use of acetamidine hydrochloride to synthesize 6-methyl-4-amino-pyrazolo[3,4-d]pyrimidine base and regioselective synthesis of six new carbocyclic nucleosides (6a-f) for antiviral evaluation. Theoretical investigations were carried out on the basis of thermodynamic and kinetic stability using MM based energy optimizations and QM based transition state search for the significant regioselectivity, which was further experimentally analyzed by NOE and UV spectroscopy.
Collapse
Affiliation(s)
- Mohan Kasula
- a Department of Chemistry , Birla Institute of Technology , Mesra , Ranchi -835215 , India
| | | | - Harapriya Chakravarty
- a Department of Chemistry , Birla Institute of Technology , Mesra , Ranchi -835215 , India
| | - Chandralata Bal
- a Department of Chemistry , Birla Institute of Technology , Mesra , Ranchi -835215 , India
| | - Masanori Baba
- c Division of Antiviral Chemotherapy , Center for Chronic Viral Diseases, Kagoshima University , Kagoshima , Japan
| | - Ashok Kumar Jha
- b Chemistry Services, GVK Biosciences Pvt. Ltd., IDA Uppal , Hyderabad 500039 , India
| | - Ashoke Sharon
- a Department of Chemistry , Birla Institute of Technology , Mesra , Ranchi -835215 , India
| |
Collapse
|
31
|
[3+2] Redox-Neutral Cycloaddition of Nitrocyclopropanes with Styrenes by Visible-Light Photocatalysis. Chemistry 2015; 21:9676-80. [DOI: 10.1002/chem.201500873] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Indexed: 12/22/2022]
|
32
|
|
33
|
|
34
|
Xie MS, Wang Y, Li JP, Du C, Zhang YY, Hao EJ, Zhang YM, Qu GR, Guo HM. A straightforward entry to chiral carbocyclic nucleoside analogues via the enantioselective [3+2] cycloaddition of α-nucleobase substituted acrylates. Chem Commun (Camb) 2015; 51:12451-4. [DOI: 10.1039/c5cc04832j] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A straightforward entry to chiral carbocyclic nucleoside analogues is achievedviathe enantioselective [3+2] cycloaddition of α-nucleobase substituted acrylates to vinyl cyclopropanes.
Collapse
Affiliation(s)
- Ming-Sheng Xie
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| | - Yong Wang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| | - Jian-Ping Li
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| | - Cong Du
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| | - Yan-Yan Zhang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| | - Er-Jun Hao
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| | - Yi-Ming Zhang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| | - Gui-Rong Qu
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| | - Hai-Ming Guo
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| |
Collapse
|
35
|
Kaneko C, Katagiri N, Nomura M, Sato H. A New Method for the Stereoselective Synthesis of Nucleosides by Means of Sodium Borohydride Mediated Reductive C-C or C-N Bond Cleavage Reaction. Isr J Chem 2013. [DOI: 10.1002/ijch.199100029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
36
|
Katane M, Matsuda S, Saitoh Y, Sekine M, Furuchi T, Koyama N, Nakagome I, Tomoda H, Hirono S, Homma H. The Antiviral Drug Acyclovir Is a Slow-Binding Inhibitor of d-Amino Acid Oxidase. Biochemistry 2013; 52:5665-74. [DOI: 10.1021/bi400478a] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Masumi Katane
- Laboratory of Biomolecular Science,
Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641,
Japan
| | - Satsuki Matsuda
- Laboratory of Biomolecular Science,
Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641,
Japan
| | - Yasuaki Saitoh
- Laboratory of Biomolecular Science,
Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641,
Japan
| | - Masae Sekine
- Laboratory of Biomolecular Science,
Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641,
Japan
| | - Takemitsu Furuchi
- Laboratory of Biomolecular Science,
Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641,
Japan
| | - Nobuhiro Koyama
- Laboratory of Microbial
Chemistry,
Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641,
Japan
| | - Izumi Nakagome
- Laboratory of Physical
Chemistry
for Drug Design, Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo
108-8641, Japan
| | - Hiroshi Tomoda
- Laboratory of Microbial
Chemistry,
Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641,
Japan
| | - Shuichi Hirono
- Laboratory of Physical
Chemistry
for Drug Design, Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo
108-8641, Japan
| | - Hiroshi Homma
- Laboratory of Biomolecular Science,
Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641,
Japan
| |
Collapse
|
37
|
Cho JH, Amblard F, Coats SJ, Schinazi RF. Synthesis of cyclopentanyl carbocyclic 5-fluorocytosine ((-)-5-fluorocarbodine) using a facially selective hydrogenation approach. J Org Chem 2013; 78:723-7. [PMID: 23231055 DOI: 10.1021/jo302038d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An efficient synthetic route to biologically relevant (-)-5-fluorocarbodine 6 was developed. Direct coupling of N(6)-protected 5-fluorouracil 15 with cyclopentenyl intermediate 13, followed by formation of a macrocycle between the base and the carbocyclic sugar moiety, via ring-closing metathesis, allowed for a facial selective hydrogenation of the sugar double bond to give, exclusively, the desired 4'-β stereoisomer.
Collapse
Affiliation(s)
- Jong Hyun Cho
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Veterans Affairs Medical Center, Decatur, Georgia 30033, USA
| | | | | | | |
Collapse
|
38
|
Liu C, Chen Q, Yang M, Schneller SW. C-3 halo and 3-methyl substituted 5′-nor-3-deazaaristeromycins: Synthesis and antiviral properties. Bioorg Med Chem 2013. [DOI: 10.1016/j.bmc.2012.09.051] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
39
|
Georgieva MK, Duarte FJS, Queirós MVB, Santos AG. Catalytic Asymmetric 5-enolexo Aldolizations. A Computational Study. J Org Chem 2012; 77:5569-76. [DOI: 10.1021/jo300633y] [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]
Affiliation(s)
- Miglena K. Georgieva
- REQUIMTE, CQFB, Departamento de Química, Faculdade
de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Filipe J. S. Duarte
- REQUIMTE, CQFB, Departamento de Química, Faculdade
de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Margarida V. B. Queirós
- REQUIMTE, CQFB, Departamento de Química, Faculdade
de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - A. Gil Santos
- REQUIMTE, CQFB, Departamento de Química, Faculdade
de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| |
Collapse
|
40
|
Singh R, Vince R. 2-Azabicyclo[2.2.1]hept-5-en-3-one: Chemical Profile of a Versatile Synthetic Building Block and its Impact on the Development of Therapeutics. Chem Rev 2012; 112:4642-86. [DOI: 10.1021/cr2004822] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Rohit Singh
- Center for Drug Design, Academic Health Center, University of Minnesota, 516 Delaware Street Southeast,
Minneapolis, MN 55455, United States
| | - Robert Vince
- Center for Drug Design, Academic Health Center, University of Minnesota, 516 Delaware Street Southeast,
Minneapolis, MN 55455, United States
| |
Collapse
|
41
|
W. Schneller S, Chen C, Ye W. A CONVENIENT SYNTHESIS OF THE L-LIKE ENANTIOMER OF 4'-METHYL-3-dEAZAARISTEROMYCIN. HETEROCYCLES 2012. [DOI: 10.3987/com-12-s(n)20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
42
|
Rawal RK, Singh US, Gadthula S, Chu CK. Synthesis of Entecavir and Its Novel Class of Analogs. ACTA ACUST UNITED AC 2011; Chapter 14:Unit 14.7.1-17. [DOI: 10.1002/0471142700.nc1407s47] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
| | | | | | - Chung K. Chu
- The University of Georgia College of Pharmacy Athens Georgia
| |
Collapse
|
43
|
Catalytic [3+2] Annulation of Aminocyclopropanes for the Enantiospecific Synthesis of Cyclopentylamines. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201106255] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
44
|
de Nanteuil F, Waser J. Catalytic [3+2] Annulation of Aminocyclopropanes for the Enantiospecific Synthesis of Cyclopentylamines. Angew Chem Int Ed Engl 2011; 50:12075-9. [DOI: 10.1002/anie.201106255] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2011] [Indexed: 11/08/2022]
|
45
|
Kulkarni MG, Borhade AS, Shaikh YB, Dhondge AP, Birhade DR, Dhatrak NR. A stereodivergent approach to carbahexofuranoses: synthesis of carba-α-d-glucofuranose, carba-β-d-altrofuranose, carba-α-d-allofuranose, carba-β-d-idofuranose, carba-α-d-galactofuranose and carba-β-d-talofuranose. Tetrahedron Lett 2011. [DOI: 10.1016/j.tetlet.2011.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Nakano C, Kudo F, Eguchi T, Ohnishi Y. Genome Mining Reveals Two Novel Bacterial Sesquiterpene Cyclases: (−)-Germacradien-4-ol and (−)-epi-α-Bisabolol Synthases from Streptomyces citricolor. Chembiochem 2011; 12:2271-5. [DOI: 10.1002/cbic.201100418] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
47
|
Chattopadhyay A, Tripathy S. Stereodivergent Route to the Carbocyclic Core of 2′,3′-Olefinic Carbanucleosides: Toward the Synthesis of (l)-(+)- and (d)-(−)-Carbovir. J Org Chem 2011; 76:5856-61. [DOI: 10.1021/jo200670v] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
48
|
Leung LMH, Gibson V, Linclau B. A linchpin carbacyclization approach for the synthesis of carbanucleosides. J Org Chem 2010; 73:9197-206. [PMID: 18975910 DOI: 10.1021/jo801848h] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A convenient synthesis of carbanucleosides, with both enantiomers equally accessible, is reported. The key step is a tandem linchpin cyclization process to give access to substituted carbafuranose derivatives having the correct relative stereochemistry for subsequent nucleobase introduction with inversion of configuration at C1. This was illustrated by the synthesis of 2',3'-dideoxycarbathymidine via a convergent nucleobase introduction and of 2',3'-dideoxy-6'-hydroxycarbauridine via a linear nucleobase introduction. Both methods relied on Mitsunobu chemistry, and the first example of the Mukaiyama modification of the Mitsunobu reaction involving nucleobases as nucleophiles is reported.
Collapse
Affiliation(s)
- Leo M H Leung
- School of Chemistry, University of Southampton, Highfield, Southampton SO17 1BJ, UK
| | | | | |
Collapse
|
49
|
Szakonyi Z, Fülöp F. Carbocyclic nucleosides from enantiomeric, α-pinane-based aminodiols. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/j.tetasy.2010.04.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
50
|
Kurteva VB, Afonso CAM. Synthesis of Cyclopentitols by Ring-Closing Approaches. Chem Rev 2009; 109:6809-57. [DOI: 10.1021/cr900169j] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Vanya B. Kurteva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev str., bl. 9, 1113 Sofia, Bulgaria, and CQFM, Centro de Química-Física Molecular, IN - Institute of Nanosciences and Nanotechnology, Instituto Superior Técnico, 1049-001 Lisboa, Portugal
| | - Carlos A. M. Afonso
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev str., bl. 9, 1113 Sofia, Bulgaria, and CQFM, Centro de Química-Física Molecular, IN - Institute of Nanosciences and Nanotechnology, Instituto Superior Técnico, 1049-001 Lisboa, Portugal
| |
Collapse
|