1
|
Zhu Y, Zhang X, Ma C, Ren X, Wang W, Zhang K, Zhang G, Che Q, Zhu T, Li D. Discovery and Characterization of a Fungal N-acetylglucosamine Transferase in the Biosynthesis of Furanone Glycosides. JOURNAL OF NATURAL PRODUCTS 2025; 88:427-432. [PMID: 39881635 DOI: 10.1021/acs.jnatprod.4c01190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Malfilamentosides are a class of fungal secondary metabolites characterized by glycosylated furanone scaffold; however, the enzyme that catalyzes the O-glycosylation of the furanone core with N-acetylglucosamine (GlcNAc) has not yet been identified. In this study, we discovered and identified the biosynthetic gene cluster of the malfilamentosides. In vivo and in vitro investigations revealed that a glycosyltransferase, MftB, catalyzes the O-glycosylation of the furanone scaffold with GlcNAc. Furthermore, MftB exhibits broad promiscuity toward glycosyl donors and acceptors, highlighting its potential in glycoside production.
Collapse
Affiliation(s)
- Yongchun Zhu
- Key Laboratory of Marine Drugs Ministry of Education; School of Medicine and Pharmacy; Sanya Oceanographic Institute, Ocean University of China, Qingdao/Sanya 266100, People's Republic of China
| | - Xianyan Zhang
- Key Laboratory of Marine Drugs Ministry of Education; School of Medicine and Pharmacy; Sanya Oceanographic Institute, Ocean University of China, Qingdao/Sanya 266100, People's Republic of China
| | - Chuanteng Ma
- Key Laboratory of Marine Drugs Ministry of Education; School of Medicine and Pharmacy; Sanya Oceanographic Institute, Ocean University of China, Qingdao/Sanya 266100, People's Republic of China
| | - Xingtao Ren
- Key Laboratory of Marine Drugs Ministry of Education; School of Medicine and Pharmacy; Sanya Oceanographic Institute, Ocean University of China, Qingdao/Sanya 266100, People's Republic of China
| | - Wenxue Wang
- Key Laboratory of Marine Drugs Ministry of Education; School of Medicine and Pharmacy; Sanya Oceanographic Institute, Ocean University of China, Qingdao/Sanya 266100, People's Republic of China
| | - Kaijin Zhang
- Key Laboratory of Marine Drugs Ministry of Education; School of Medicine and Pharmacy; Sanya Oceanographic Institute, Ocean University of China, Qingdao/Sanya 266100, People's Republic of China
| | - Guojian Zhang
- Key Laboratory of Marine Drugs Ministry of Education; School of Medicine and Pharmacy; Sanya Oceanographic Institute, Ocean University of China, Qingdao/Sanya 266100, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, People's Republic of China
| | - Qian Che
- Key Laboratory of Marine Drugs Ministry of Education; School of Medicine and Pharmacy; Sanya Oceanographic Institute, Ocean University of China, Qingdao/Sanya 266100, People's Republic of China
| | - Tianjiao Zhu
- Key Laboratory of Marine Drugs Ministry of Education; School of Medicine and Pharmacy; Sanya Oceanographic Institute, Ocean University of China, Qingdao/Sanya 266100, People's Republic of China
| | - Dehai Li
- Key Laboratory of Marine Drugs Ministry of Education; School of Medicine and Pharmacy; Sanya Oceanographic Institute, Ocean University of China, Qingdao/Sanya 266100, People's Republic of China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, People's Republic of China
| |
Collapse
|
2
|
Jin S, Chen H, Zhang J, Lin Z, Qu X, Jia X, Lei C. Analyzing and engineering of the biosynthetic pathway of mollemycin A for enhancing its production. Synth Syst Biotechnol 2024; 9:445-452. [PMID: 38606205 PMCID: PMC11007384 DOI: 10.1016/j.synbio.2024.03.014] [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: 01/19/2024] [Revised: 02/27/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024] Open
Abstract
Mollemycin A (MOMA) is a unique glyco-hexadepsipeptide-polyketide that was isolated from a Streptomyces sp. derived from the Australian marine environment. MOMA exhibits remarkable inhibitory activity against both drug-sensitive and multidrug-resistant malaria parasites. Optimizing MOMA through structural modifications or product enhancements is necessary for the development of effective analogues. However, modifying MOMA using chemical approaches is challenging, and the production titer of MOMA in the wild-type strain is low. This study identified and characterized the biosynthetic gene cluster of MOMA for the first time, proposed its complex biosynthetic pathway, and achieved an effective two-pronged enhancement of MOMA production. The fermentation medium was optimized to increase the yield of MOMA from 0.9 mg L-1 to 1.3 mg L-1, a 44% boost. Additionally, a synergistic mutant strain was developed by deleting the momB3 gene and overexpressing momB2, resulting in a 2.6-fold increase from 1.3 mg L-1 to 3.4 mg L-1. These findings pave the way for investigating the biosynthetic mechanism of MOMA, creating opportunities to produce a wide range of MOMA analogues, and developing an efficient strain for the sustainable and economical production of MOMA and its analogues.
Collapse
Affiliation(s)
- Shixue Jin
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Huixue Chen
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jun Zhang
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhi Lin
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xudong Qu
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinying Jia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia
- Department of Biochemistry, National University of Singapore, 14 Medical Dr, Singapore, 117599
| | - Chun Lei
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
3
|
Martin ALAR, Pereira RLS, Rocha JE, Farias PAM, Freitas TS, Caldas FRDL, Figueredo FG, Sampaio NFL, Oliveira-Tintino CDDM, Tintino SR, da Hora GCA, Lima MCP, de Menezes IRA, Carvalho DT, Coutinho HDM, Fonteles MMF. Unlocking bacterial defense: Exploring the potent inhibition of NorA efflux pump by coumarin derivatives in Staphylococcus aureus. Microb Pathog 2024; 190:106608. [PMID: 38503396 DOI: 10.1016/j.micpath.2024.106608] [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: 11/24/2023] [Revised: 02/27/2024] [Accepted: 03/03/2024] [Indexed: 03/21/2024]
Abstract
The occurrence of bacterial resistance has been increasing, compromising the treatment of various infections. The high virulence of Staphylococcus aureus allows for the maintenance of the infectious process, causing many deaths and hospitalizations. The MepA and NorA efflux pumps are transporter proteins responsible for expelling antimicrobial agents such as fluoroquinolones from the bacterial cell. Coumarins are phenolic compounds that have been studied for their diverse biological actions, including against bacteria. A pharmacokinetic in silico characterization of compounds C10, C11, C13, and C14 was carried out according to the principles of Lipinski's Rule of Five, in addition to searching for similarity in ChemBL and subsequent search for publications in CAS SciFinder. All compounds were evaluated for their in vitro antibacterial and modulatory activity against standard and multidrug-resistant Gram-positive and Gram-negative strains. The effect of coumarins C9, C10, C11, C13, and C14 as efflux pump inhibitors in Staphylococcus aureus strains was evaluated using the microdilution method (MepA or NorA) and fluorimetry (NorA). The behavior of coumarins regarding the efflux pump was determined from their interaction properties with the membrane and coumarin-protein using molecular docking and molecular dynamics simulations. Only the isolated coumarin compound C13 showed antibacterial activity against standard strains of Staphylococcus aureus and Escherichia coli. However, the other tested coumarins showed modulatory capacity for fluoroquinolone and aminoglycoside antibacterials. Compounds C10, C13, and C14 were effective in reducing the MIC of both antibiotics for both multidrug-resistant strains, while C11 potentiated the effect of norfloxacin and gentamicin for Gram-positive and Gram-negative bacteria and only norfloxacin for Gram-negative. Only coumarin C14 produced synergistic effects when associated with ciprofloxacin in MepA-carrying strains. All tested coumarins have the ability to inhibit the NorA efflux pump present in Staphylococcus aureus, both in reducing the MIC and inducing increased ethidium bromide fluorescence emission in fluorimetry. The findings of this study offer an atomistic perspective on the potential of coumarins as active inhibitors of the NorA pump, highlighting their specific mode of action mainly targeting protein inhibition. In molecular docking, it was observed that coumarins are capable of interacting with various amino acid residues of the NorA pump. The simulation showed that coumarin C10 can cross the bilayer; however, the other coumarins interacted with the membrane but were unable to cross it. Coumarins demonstrated their potentiating role in the effect of norfloxacin through a dual mechanism: efflux pump inhibition through direct interaction with the protein (C9, C10, C11, and C13) and increased interaction with the membrane (C10 and C13). In the context of pharmacokinetic prediction studies, the studied structures have a suitable chemical profile for possible oral use. We suggest that coumarin derivatives may be an interesting alternative in the future for the treatment of resistant bacterial infections, with the possibility of a synergistic effect with other antibacterials, although further studies are needed to characterize their therapeutic effects and toxicity.
Collapse
Affiliation(s)
- Ana Luíza A R Martin
- Department of Physiology and Pharmacology, Federal University of Ceará - UFC, 60430-160, Fortaleza, Brazil; Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil; School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil
| | | | - Janaína Esmeraldo Rocha
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | - Pablo A M Farias
- School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil; CECAPE College, 63024-015, Juazeiro do Norte, Brazil
| | - Thiago S Freitas
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | | | - Fernando G Figueredo
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil; School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil
| | - Nadghia Figueiredo Leite Sampaio
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil; School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil
| | | | - Saulo Relison Tintino
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | | | | | - Irwin Rose A de Menezes
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | - Diogo T Carvalho
- School of Pharmacy, Federal University of Alfenas - UNIFAL, 37130-001, Alfenas, Brazil
| | - Henrique D M Coutinho
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil.
| | - Marta M F Fonteles
- Department of Physiology and Pharmacology, Federal University of Ceará - UFC, 60430-160, Fortaleza, Brazil
| |
Collapse
|
4
|
Chen D, Song Z, Han J, Liu J, Liu H, Dai J. Targeted Discovery of Glycosylated Natural Products by Tailoring Enzyme-Guided Genome Mining and MS-Based Metabolome Analysis. J Am Chem Soc 2024; 146:9614-9622. [PMID: 38545685 DOI: 10.1021/jacs.3c12895] [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: 04/11/2024]
Abstract
Glycosides make up a biomedically important class of secondary metabolites. Most naturally occurring glycosides were isolated from plants and bacteria; however, the chemical diversity of glycosylated natural products in fungi remains largely unexplored. Herein, we present a paradigm to specifically discover diverse and bioactive glycosylated natural products from fungi by combining tailoring enzyme-guided genome mining with mass spectrometry (MS)-based metabolome analysis. Through in vivo genes deletion and heterologous expression, the first fungal C-glycosyltransferase AuCGT involved in the biosynthesis of stromemycin was identified from Aspergillus ustus. Subsequent homology-based genome mining for fungal glycosyltransferases by using AuCGT as a probe revealed a variety of biosynthetic gene clusters (BGCs) containing its homologues in diverse fungi, of which the glycoside-producing capability was corroborated by high-performance liquid chromatography-mass spectrometry (HPLC-MS) analysis. Consequently, 28 fungal aromatic polyketide C/O-glycosides, including 20 new compounds, were efficiently discovered and isolated from the three selected fungi. Moreover, several novel fungal C/O-glycosyltransferases, especially three novel α-pyrone C-glycosyltransferases, were functionally characterized and verified in the biosynthesis of these glycosides. In addition, a proof of principle for combinatorial biosynthesis was applied to design the production of unnatural glycosides in Aspergillus nidulans. Notably, the newly discovered glycosides exhibited significant antiviral, antibacterial, and antidiabetic activities. Our work demonstrates the promise of tailoring enzyme-guided genome-mining approach for the targeted discovery of fungal glycosides and promotes the exploration of a broader chemical space for natural products with a target structural motif in microbial genomes.
Collapse
Affiliation(s)
- Dawei Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs and NHC Key Laboratory of Biosynthesis of Natural Products, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhijun Song
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs and NHC Key Laboratory of Biosynthesis of Natural Products, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Junjie Han
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jimei Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs and NHC Key Laboratory of Biosynthesis of Natural Products, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hongwei Liu
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jungui Dai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs and NHC Key Laboratory of Biosynthesis of Natural Products, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
5
|
Khanam A, Dubey S, Mandal PK. Mild method for the synthesis of α-glycosyl chlorides: A convenient protocol for quick one-pot glycosylation. Carbohydr Res 2023; 534:108976. [PMID: 37871478 DOI: 10.1016/j.carres.2023.108976] [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: 09/16/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/25/2023]
Abstract
A simple and efficient protocol for the preparation of α-glycosyl chlorides within 15-30 min is described which employs a stable, cheap, and commercially available Trichloroisocyanuric acid (TCCA) as non-toxic chlorinating agent along with PPh3. This process involved a wide range of substrate scope and is well-suited with labile hydroxyl protecting groups such as benzyl, acetyl, benzoyl, isopropylidene, benzylidene, and TBDPS (tert-butyldiphenylsilyl) groups. This process is operationally simple, mild conditions and obtained good yields with excellent α selectivity. Moreover, a multi-catalyst one-pot glycosylation can be carried out to transform the glycosyl hemiacetals directly to a various O-glycosides in high overall yields without the need for separation or purification of the α-glycosyl chloride donors.
Collapse
Affiliation(s)
- Ariza Khanam
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, P.O. Box 173, Lucknow, 226 031, India
| | - Shashiprabha Dubey
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, P.O. Box 173, Lucknow, 226 031, India
| | - Pintu Kumar Mandal
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, P.O. Box 173, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
6
|
Yu S, Zheng J, Zhang Y, Meng D, Wang Y, Xu X, Liang N, Shabiti S, Zhang X, Wang Z, Yang Z, Mi P, Zheng X, Li W, Chen H. The mechanisms of multidrug resistance of breast cancer and research progress on related reversal agents. Bioorg Med Chem 2023; 95:117486. [PMID: 37847948 DOI: 10.1016/j.bmc.2023.117486] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/18/2023] [Accepted: 09/29/2023] [Indexed: 10/19/2023]
Abstract
Chemotherapy is the mainstay in the treatment of breast cancer. However, many drugs that are commonly used in clinical practice have a high incidence of side effects and multidrug resistance (MDR), which is mainly caused by overexpression of drug transporters and related enzymes in breast cancer cells. In recent years, researchers have been working hard to find newer and safer drugs to overcome MDR in breast cancer. In this review, we provide the molecule mechanism of MDR in breast cancer, categorize potential lead compounds that inhibit single or multiple drug transporter proteins, as well as related enzymes. Additionally, we have summarized the structure-activity relationship (SAR) based on potential breast cancer MDR modulators with lower side effects. The development of novel approaches to suppress MDR is also addressed. These lead compounds hold great promise for exploring effective chemotherapy agents to overcome MDR, providing opportunities for curing breast cancer in the future.
Collapse
Affiliation(s)
- Shiwen Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China; Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Jinling Zheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China; Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Yan Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China
| | - Dandan Meng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China; Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Yujue Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China
| | - Xiaoyu Xu
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Na Liang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Shayibai Shabiti
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Xu Zhang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Zixi Wang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Zehua Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China
| | - Pengbing Mi
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China
| | - Xing Zheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China; Department of Pharmacy, Hunan Vocational College of Science and Technology, Third Zhongyi Shan Road, Changsha, Hunan Province 425101, PR China.
| | - Wenjun Li
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nano formulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Hongfei Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, China Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research [Hunan Provincial Science and Technology Department document (Approval number: 2019-56)], School of Pharmaceutical Science, Hengyang Medical School, University of South China, No.28 Changshengxi Road, Hengyang 421001, PR China.
| |
Collapse
|
7
|
Křen V, Bojarová P. Rutinosidase and other diglycosidases: Rising stars in biotechnology. Biotechnol Adv 2023; 68:108217. [PMID: 37481095 DOI: 10.1016/j.biotechadv.2023.108217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/09/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Diglycosidases are a special class of glycosidases (EC 3.2.1) that catalyze the separation of intact disaccharide moieties from the aglycone part. The main diglycosidase representatives comprise rutinosidases that cleave rutinose (α-l-Rha-(1-6)-β-d-Glc) from rutin or other rutinosides, and (iso)primeverosidases processing (iso)primeverosides (d-Xyl-(1-6)-β-d-Glc), but other activities are known. Notably, some diglycosidases may be ranked as monoglucosidases with enlarged substrate specificity. Diglycosidases are found in various microorganisms and plants. Diglycosidases are used in the food industry for aroma enhancement and flavor modification. Besides their hydrolytic activity, they also possess pronounced synthetic (transglycosylating) capabilities. Recently, they have been demonstrated to glycosylate various substrates in a high yield, including peculiar species like inorganic azide or carboxylic acids, which is a unique feature in biocatalysis. Rhamnose-containing compounds such as rutinose are currently receiving increased attention due to their proven activity in anti-cancer and dermatological experimental studies. This review demonstrates the vast and yet underrated biotechnological potential of diglycosidases from various sources (plant, microbial), and reveals perspectives on the use of these catalysts as well as of their products in biotechnology.
Collapse
Affiliation(s)
- Vladimír Křen
- Institute of Microbiology of the Czech Academy of Sciences, Laboratory of Biotransformation, Vídeňská 1083, CZ 14200 Prague 4, Czech Republic.
| | - Pavla Bojarová
- Institute of Microbiology of the Czech Academy of Sciences, Laboratory of Biotransformation, Vídeňská 1083, CZ 14200 Prague 4, Czech Republic.
| |
Collapse
|
8
|
Zargar IA, Rasool B, Sakander N, Mukherjee D. Switchable reactivity of 2-benzoyl glycals towards stereoselective access of 1-3 and 1-1 S/ O linked disaccharides. Chem Commun (Camb) 2023; 59:10448-10451. [PMID: 37555476 DOI: 10.1039/d3cc02870d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
We have developed a synthesis of 1-3 and 1-1 disaccharides from 2-benzoyl glycal and anomeric thiol and/or hydroxy sugar acceptors under mild conditions at room temperature. The regio and stereo-selectivity of the newly formed inter-glycosidic linkages are dependent on the nature of the glycal donor (D or L) and anomeric acceptor.
Collapse
Affiliation(s)
- Irshad Ahmad Zargar
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (IIIM), Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Bisma Rasool
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (IIIM), Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Norein Sakander
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (IIIM), Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debaraj Mukherjee
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Department of Chemical Sciences, Bose Institute Kolkata, EN 80, Sector V, Bidhan Nagar, Kolkata-700091, WB, India
| |
Collapse
|
9
|
Huynh NO, Hodík T, Krische MJ. Enantioselective Transfer Hydrogenative Cycloaddition Unlocks the Total Synthesis of SF2446 B3: An Aglycone of Arenimycin and SF2446 Type II Polyketide Antibiotics. J Am Chem Soc 2023; 145:17461-17467. [PMID: 37494281 PMCID: PMC10443208 DOI: 10.1021/jacs.3c06225] [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] [Indexed: 07/28/2023]
Abstract
The first total synthesis and structure validation of an arenimycin/SF2446 type II polyketide is described, as represented by de novo construction of SF2446 B3, the aglycone shared by this family of type II polyketides. Ruthenium-catalyzed α-ketol-benzocyclobutenone [4 + 2] cycloaddition, which occurs via successive stereoablation-stereoregeneration, affects a double dynamic kinetic asymmetric transformation wherein two racemic starting materials combine to form the congested angucycline bay region with control of regio-, diastereo-, and enantioselectivity. This work represents the first application of transfer hydrogenative cycloaddition and enantioselective intermolecular metal-catalyzed C-C bond activation in target-oriented synthesis.
Collapse
Affiliation(s)
- Nancy O Huynh
- Department of Chemistry, University of Texas at Austin, 105 E 24th Street, Austin, Texas 78712, United States
| | - Tomáš Hodík
- Department of Chemistry, University of Texas at Austin, 105 E 24th Street, Austin, Texas 78712, United States
| | - Michael J Krische
- Department of Chemistry, University of Texas at Austin, 105 E 24th Street, Austin, Texas 78712, United States
| |
Collapse
|
10
|
Ren J, Barton CD, Zhan J. Engineered production of bioactive polyphenolic O-glycosides. Biotechnol Adv 2023; 65:108146. [PMID: 37028465 DOI: 10.1016/j.biotechadv.2023.108146] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/04/2023] [Accepted: 04/02/2023] [Indexed: 04/09/2023]
Abstract
Polyphenolic compounds (such as quercetin and resveratrol) possess potential medicinal values due to their various bioactivities, but poor water solubility hinders their health benefits to humankind. Glycosylation is a well-known post-modification method to biosynthesize natural product glycosides with improved hydrophilicity. Glycosylation has profound effects on decreasing toxicity, increasing bioavailability and stability, together with changing bioactivity of polyphenolic compounds. Therefore, polyphenolic glycosides can be used as food additives, therapeutics, and nutraceuticals. Engineered biosynthesis provides an environmentally friendly and cost-effective approach to generate polyphenolic glycosides through the use of various glycosyltransferases (GTs) and sugar biosynthetic enzymes. GTs transfer the sugar moieties from nucleotide-activated diphosphate sugar (NDP-sugar) donors to sugar acceptors such as polyphenolic compounds. In this review, we systematically review and summarize the representative polyphenolic O-glycosides with various bioactivities and their engineered biosynthesis in microbes with different biotechnological strategies. We also review the major routes towards NDP-sugar formation in microbes, which is significant for producing unusual or novel glycosides. Finally, we discuss the trends in NDP-sugar based glycosylation research to promote the development of prodrugs that positively impact human health and wellness.
Collapse
Affiliation(s)
- Jie Ren
- Department of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, UT 84322-4105, USA
| | - Caleb Don Barton
- Department of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, UT 84322-4105, USA
| | - Jixun Zhan
- Department of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, UT 84322-4105, USA.
| |
Collapse
|
11
|
Ni J, Yu L, Li F, Li Y, Zhang M, Deng Y, Liu X. Macrolactin R from Bacillus siamensis and its antifungal activity against Botrytis cinerea. World J Microbiol Biotechnol 2023; 39:117. [PMID: 36918502 DOI: 10.1007/s11274-023-03563-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/02/2023] [Indexed: 03/16/2023]
Abstract
Botrytis cinerea is listed among the most important fungal pathogens infecting strawberries. The use of biological control agents, such as Bacillus species, offers an alternative and effective way to reduce airborne pathogens. The aim of this research was to select the macrolactin R produced by Bacillus siamensis with potential for using as biological agents against the pathogenetic fungi (Botrytis cinerea) of strawberries, and to assess the mechanisms involved. Macrolactin R had significant inhibitory effects on spore germination, germ tube elongation, and mycelial growth of Botrytis cinerea. The MICs of macrolactin R inhibitions in vitro was 12.5 mg/L and The EC50 value of NJ08-3 to Botrytis cinerea spores and mycelial was 1.93 and 2.88 mg/L, respectively. Macrolactin R impacted the membrane structure of Botrytis cinerea, resulting in changes in membrane permeability and leakage of proteins and nucleic acids, then cell death. The application of the macrolactin R of Bacillus siamensis reduced the disease severity index of gray mold on strawberries. This study demonstrated that the production of macrolactin R produced by Bacillus siamensis are involved in the antifungal activity against Botrytis cinerea.
Collapse
Affiliation(s)
- Jie Ni
- Department of Food Science and Engineering, College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, Guangxi, P.R. China
| | - Lian Yu
- Department of Food Science and Engineering, College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, Guangxi, P.R. China.
| | - Fei Li
- Guangxi Key Laboratory of Marine Environmental Science, Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning, 530007, P.R. China.
| | - Yulin Li
- Department of Food Science and Engineering, College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, Guangxi, P.R. China
| | - Mengfei Zhang
- Department of Food Science and Engineering, College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, Guangxi, P.R. China
| | - Yuping Deng
- Department of Food Science and Engineering, College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, Guangxi, P.R. China
| | - Xiaoling Liu
- Department of Food Science and Engineering, College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, Guangxi, P.R. China
| |
Collapse
|
12
|
Zheng S, Zhao H, Yuan Z, Si X, Li Z, Song J, Zhu Y, Wu H. The Analysis of the Glycosyltransferase Gene Function From a Novel Granaticin Producer, Streptomyces Vilmorinianum. YP1. Curr Microbiol 2023; 80:103. [PMID: 36781498 DOI: 10.1007/s00284-023-03192-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 12/15/2022] [Indexed: 02/15/2023]
Abstract
Glycosylation is common among the synthesis of natural product and imparts the bioactivity for natural product. As for granaticin, a natural product with great bioactivity, glycosylation is an unusual sugar attachment and remains enigmatic. Orf14 in the gra cluster is the predicted glycosyltransferase but without being identified. Recently, we isolated and identified a novel granaticin producer Streptomyces vilmorinianum YP1. Orf14 gene in gra cluster of YP1 is knocked out and complemented. The instrumental analysis of the blue product synthesized by orf14-deficient mutant exhibits the none-granaticin detection and deglycosylated intermediates accumulation. The bioactivity and stability test suggests the weaker or none antibacterial activity and cytotoxicity of this blue product with greater ultraviolet stability and thermostability than granaticin and derivatives produced by YP1. All the result indicates that orf14 encodes glycosyltransferase and glycosylation played an important role in the bioactivity of granaticin. Meanwhile, the blue pigment, deglycosylated intermediates, has favorable processing characteristics. Our finding supplies the function of orf14 and glycosylation, but also indicates a promising candidate of edible blue pigment applicated in food industry.
Collapse
Affiliation(s)
- Shenglan Zheng
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, 100048, China
| | - Hongling Zhao
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, 100048, China
| | - Zuoyun Yuan
- Department of Science Technology and Innovation, Future Science and Technology Park South, COFCO Nutrition and Health Research Institute, BeiQiJia, ChangPing, BeiJing, 102209, China
| | - Xuechen Si
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, 100048, China
| | - Zongxian Li
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, 100048, China
| | - Jingyi Song
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, No.33, Fucheng Road, Beijing, 100048, China
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing, 100048, China
| | - Yunping Zhu
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, 100048, China.
- College of Food and Health, Beijing Technology and Business University, No. 33, Fucheng Road, Haidian District, Beijing, 100048, China.
| | - Hua Wu
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, No.33, Fucheng Road, Beijing, 100048, China
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing, 100048, China
| |
Collapse
|
13
|
Choknud S, Prawisut A, Gorantla JN, Cairns JRK. Expression, purification, characterization and glycoside production potential of rice β-d-glucan glucohydrolase I (OsExoI). Process Biochem 2023. [DOI: 10.1016/j.procbio.2023.02.015] [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: 02/17/2023]
|
14
|
Pasternak ARO, Balunas MJ, Zechel DL. Discovery of 3'- O-β-Glucosyltubercidin and the Nucleoside Specific Glycosyltransferase AvpGT through Genome Mining. ACS Chem Biol 2022; 17:3507-3514. [PMID: 36356213 DOI: 10.1021/acschembio.2c00707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A genome mining approach was used to identify a hybrid tubercidin-nucleocidin biosynthetic gene cluster (BGC) in Streptomyces sp. AVP053U2. Analysis of culture extracts by liquid chromatography-mass spectrometry revealed the presence of a glucosylated tubercidin derivative. A gene, avpGT, was identified within the hybrid cluster that has homology to the glucosyltransferase that is responsible for 3'-O-β-glucosylation of the fluorinated natural product nucleocidin. AvpGT was heterologously expressed and purified from Escherichia coli for in vitro characterization. AvpGT is active toward UDP-glucose and UDP-galactose as glycosyl donors and several nucleosides as acceptors. Kinetic analysis revealed that AvpGT is most specific for UDP-glucose [kcat/KMapp = (1.1 ± 0.3) × 105 M-1·s-1] as the glycosyl donor and tubercidin [kcat/KMapp = (5.3 ± 1.8) × 104 M-1·s-1] as the glycosyl acceptor. NMR spectroscopic analysis revealed the product of this reaction to be 3'-O-β-glucopyranosyl tubercidin. A sequence analysis of AvpGT reveals a family of nucleoside-specific GTs, which may be used as markers of BGCs that produce glycosylated nucleosides.
Collapse
Affiliation(s)
- A R Ola Pasternak
- Department of Chemistry, Queen's University, Kingston, K7L 3N6 Ontario, Canada
| | - Marcy J Balunas
- Departments of Microbiology and Immunology and Medicinal Chemistry, University of Michigan, Ann Arbor, 48109 Michigan, United States
| | - David L Zechel
- Department of Chemistry, Queen's University, Kingston, K7L 3N6 Ontario, Canada
| |
Collapse
|
15
|
Seara FAC, Kasai-Brunswick TH, Nascimento JHM, Campos-de-Carvalho AC. Anthracycline-induced cardiotoxicity and cell senescence: new therapeutic option? Cell Mol Life Sci 2022; 79:568. [PMID: 36287277 PMCID: PMC11803035 DOI: 10.1007/s00018-022-04605-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/30/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022]
Abstract
Anthracyclines are chemotherapeutic drugs widely used in the frontline of cancer treatment. The therapeutic mechanisms involve the stabilization of topoisomerase IIα, DNA, and the anthracycline molecule in a ternary complex that is recognized as DNA damage. Redox imbalance is another vital source of oxidative DNA damage. Together, these mechanisms lead to cytotoxic effects in neoplastic cells. However, anthracycline treatment can elicit cardiotoxicity and heart failure despite the therapeutic benefits. Topoisomerase IIβ and oxidative damage in cardiac cells have been the most reported pathophysiological mechanisms. Alternatively, cardiac cells can undergo stress-induced senescence when exposed to anthracyclines, a state primarily characterized by cell cycle arrest, organelle dysfunction, and a shift to senescence-associated secretory phenotype (SASP). The SASP can propagate senescence to neighboring cells in an ongoing process that leads to the accumulation of senescent cells, promoting cellular dysfunction and extracellular matrix remodeling. Therefore, the accumulation of senescent cardiac cells is an emerging pathophysiological mechanism associated with anthracycline-induced cardiotoxicity. This paradigm also raises the potential for therapeutic approaches to clear senescent cells in treating anthracycline-induced cardiotoxicity (i,e, senolytic therapies).
Collapse
Affiliation(s)
- Fernando A C Seara
- Departament of Physiological Sciences, Institute of Health and Biological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
- Multicenter Graduate Program of Physiological Sciences, Brazilian Society of Physiology, Rio de Janeiro, Brazil
| | - Tais H Kasai-Brunswick
- National Centre of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Science and Technology Institute in Regenerative Medicine, Rio de Janeiro, Brazil
| | - Jose H M Nascimento
- Laboratory of Cellular and Molecular Cardiology, Health Sciences Building, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Room G2-053, 373 Carlos Chagas Filho Avenue, Rio de Janeiro, RJ, 21941-590, Brazil
- National Science and Technology Institute in Regenerative Medicine, Rio de Janeiro, Brazil
| | - Antonio C Campos-de-Carvalho
- National Centre of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Laboratory of Cellular and Molecular Cardiology, Health Sciences Building, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Room G2-053, 373 Carlos Chagas Filho Avenue, Rio de Janeiro, RJ, 21941-590, Brazil.
- National Science and Technology Institute in Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Potent Antibiotic Lemonomycin: A Glimpse of Its Discovery, Origin, and Chemical Synthesis. Molecules 2022; 27:molecules27134324. [PMID: 35807568 PMCID: PMC9268379 DOI: 10.3390/molecules27134324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/01/2022] [Accepted: 07/01/2022] [Indexed: 02/05/2023] Open
Abstract
Lemonomycin (1) was first isolated from the fermentation broth of Streptomyces candidus in 1964. The complete chemical structure was not elucidated until 2000 with extensive spectroscopic analysis. Lemonomycin is currently known as the only glycosylated tetrahydroisoquinoline antibiotic. Its potent antibacterial activity against Staphylococcus aureus and Bacillus subtilis and complex architecture make it an ideal target for total synthesis. In this short review, we summarize the research status of lemonomycin for biological activity, biosynthesis, and chemical synthesis. The unique deoxy aminosugar-lemonose was proposed to play a crucial role in biological activity, as shown in other antibiotics, such as arimetamycin A, nocathiacin I, glycothiohexide α, and thiazamycins. Given the self-resistance of the original bacterial host, the integration of biosynthesis and chemical synthesis to pursue efficient synthesis and further derivatization is in high demand for the development of novel antibiotics to combat antibiotic-resistant infections.
Collapse
|
17
|
Adel M. Kamal El-Dean, Geies AA, Hassanien R, Abdel-Wadood FK, El-Naeem EEA. Novel Synthesis, Reactions, and Biological Study of New Morpholino-Thieno[2,3-c][2,7]Naphthyridines as Anti-Cancer and Anti-Microbial Agents. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022040021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Singh K, Kulkarni SS. Small Carbohydrate Derivatives as Potent Antibiofilm Agents. J Med Chem 2022; 65:8525-8549. [PMID: 35777073 DOI: 10.1021/acs.jmedchem.1c01039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Biofilm formation by most pathogenic bacteria is considered as one of the key mechanisms associated with virulence and antibiotic resistance. Biofilm-forming bacteria adhere to the surfaces of biological or implant medical devices and create communities within their self-produced extracellular matrix that are difficult to treat by existing antibiotics. There is an urgent need to synthesize and screen structurally diverse molecules for their antibiofilm activity that can remove or minimize the bacterial biofilm. The development of carbohydrate-based small molecules as antibiofilm agents holds a great promise in addressing the problem of the eradication of biofilm-related infections. Owing to their structural diversity and specificity, the sugar scaffolds are valuable entities for developing antibiofilm agents. In this perspective, we discuss the literature pertaining to carbohydrate-based natural antibiofilm agents and provide an overview of the design, activity, and mode of action of potent synthetic carbohydrate-based molecules.
Collapse
Affiliation(s)
- Kartikey Singh
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Suvarn S Kulkarni
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, India 400076
| |
Collapse
|
19
|
Reintjens NR, Yakovlieva L, Marinus N, Hekelaar J, Nuti F, Papini AM, Witte MD, Minnaard AJ, Walvoort M. Palladium‐Catalyzed Oxidation of Glucose in Glycopeptides. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Niels R.M. Reintjens
- University of Groningen: Rijksuniversiteit Groningen Stratingh Institute for Chemistry NETHERLANDS
| | - Liubov Yakovlieva
- University of Groningen: Rijksuniversiteit Groningen Stratingh Institute for Chemistry NETHERLANDS
| | - Nittert Marinus
- University of Groningen: Rijksuniversiteit Groningen Stratingh Institute for Chemistry NETHERLANDS
| | - Johan Hekelaar
- University of Groningen: Rijksuniversiteit Groningen Stratingh Institute for Chemistry NETHERLANDS
| | - Francesca Nuti
- University of Florence: Universita degli Studi di Firenze Department of Chemistry “Ugo Schiff” ITALY
| | - Anna Maria Papini
- University of Florence: Universita degli Studi di Firenze Department of Chemistry “Ugo Schiff” ITALY
| | - Martin D. Witte
- University of Groningen: Rijksuniversiteit Groningen Stratingh Institute for Chemistry NETHERLANDS
| | - Adriaan J. Minnaard
- University of Groningen: Rijksuniversiteit Groningen Stratingh Institute for Chemistry NETHERLANDS
| | - Marthe Walvoort
- University of Groningen: Rijksuniversiteit Groningen Stratingh Institute for Chemistry Nijenborgh 7 9747 AG Groningen NETHERLANDS
| |
Collapse
|
20
|
Lv WX, Chen H, Zhang X, Ho CC, Liu Y, Wu S, Wang H, Jin Z, Chi YR. Programmable selective acylation of saccharides mediated by carbene and boronic acid. Chem 2022. [DOI: 10.1016/j.chempr.2022.04.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
21
|
Bhayani J, Iglesias MJ, Minen RI, Cereijo AE, Ballicora MA, Iglesias AA, Asencion Diez MD. Carbohydrate Metabolism in Bacteria: Alternative Specificities in ADP-Glucose Pyrophosphorylases Open Novel Metabolic Scenarios and Biotechnological Tools. Front Microbiol 2022; 13:867384. [PMID: 35572620 PMCID: PMC9093745 DOI: 10.3389/fmicb.2022.867384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
We explored the ability of ADP-glucose pyrophosphorylase (ADP-Glc PPase) from different bacteria to use glucosamine (GlcN) metabolites as a substrate or allosteric effectors. The enzyme from the actinobacteria Kocuria rhizophila exhibited marked and distinctive sensitivity to allosteric activation by GlcN-6P when producing ADP-Glc from glucose-1-phosphate (Glc-1P) and ATP. This behavior is also seen in the enzyme from Rhodococcus spp., the only one known so far to portray this activation. GlcN-6P had a more modest effect on the enzyme from other Actinobacteria (Streptomyces coelicolor), Firmicutes (Ruminococcus albus), and Proteobacteria (Agrobacterium tumefaciens) groups. In addition, we studied the catalytic capacity of ADP-Glc PPases from the different sources using GlcN-1P as a substrate when assayed in the presence of their respective allosteric activators. In all cases, the catalytic efficiency of Glc-1P was 1-2 orders of magnitude higher than GlcN-1P, except for the unregulated heterotetrameric protein (GlgC/GgD) from Geobacillus stearothermophilus. The Glc-1P substrate preference is explained using a model of ADP-Glc PPase from A. tumefaciens based on the crystallographic structure of the enzyme from potato tuber. The substrate-binding domain localizes near the N-terminal of an α-helix, which has a partial positive charge, thus favoring the interaction with a hydroxyl rather than a charged primary amine group. Results support the scenario where the ability of ADP-Glc PPases to use GlcN-1P as an alternative occurred during evolution despite the enzyme being selected to use Glc-1P and ATP for α-glucans synthesis. As an associated consequence in such a process, certain bacteria could have improved their ability to metabolize GlcN. The work also provides insights in designing molecular tools for producing oligo and polysaccharides with amino moieties.
Collapse
Affiliation(s)
- Jaina Bhayani
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, IL, United States
| | - Maria Josefina Iglesias
- Facultad de Bioquímica y Ciencias Biológicas, Instituto de Agrobiotecnología del Litoral, Universidad Nacional del Litoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Romina I. Minen
- Facultad de Bioquímica y Ciencias Biológicas, Instituto de Agrobiotecnología del Litoral, Universidad Nacional del Litoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Antonela E. Cereijo
- Facultad de Bioquímica y Ciencias Biológicas, Instituto de Agrobiotecnología del Litoral, Universidad Nacional del Litoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Miguel A. Ballicora
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, IL, United States
| | - Alberto A. Iglesias
- Facultad de Bioquímica y Ciencias Biológicas, Instituto de Agrobiotecnología del Litoral, Universidad Nacional del Litoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Matias D. Asencion Diez
- Facultad de Bioquímica y Ciencias Biológicas, Instituto de Agrobiotecnología del Litoral, Universidad Nacional del Litoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| |
Collapse
|
22
|
Adeniyi O, Baptista R, Bhowmick S, Cookson A, Nash RJ, Winters A, Shen J, Mur LAJ. Isolation and Characterisation of Quercitrin as a Potent Anti-Sickle Cell Anaemia Agent from Alchornea cordifolia. J Clin Med 2022; 11:jcm11082177. [PMID: 35456270 PMCID: PMC9024604 DOI: 10.3390/jcm11082177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 02/02/2023] Open
Abstract
Alchornea cordifolia Müll. Arg. (commonly known as Christmas Bush) has been used traditionally in Africa to treat sickle cell anaemia (a recessive disease, arising from the S haemoglobin (Hb) allele), but the active compounds are yet to be identified. Herein, we describe the use of sequential fractionation coupled with in vitro anti-sickling assays to purify the active component. Sickling was induced in HbSS genotype blood samples using sodium metabisulphite (Na2S2O5) or through incubation in 100% N2. Methanol extracts of A. cordifolia leaves and its sub-fractions showed >70% suppression of HbSS erythrocyte sickling. The purified compound demonstrated a 87.2 ± 2.39% significant anti-sickling activity and 93.1 ± 2.69% erythrocyte sickling-inhibition at 0.4 mg/mL. Nuclear magnetic resonance (NMR) spectra and high-resolution mass spectroscopy identified it as quercitrin (quercetin 3-rhamnoside). Purified quercitrin also inhibited the polymerisation of isolated HbS and stabilized sickle erythrocytes membranes. Metabolomic comparisons of blood samples using flow-infusion electrospray-high resolution mass spectrometry indicated that quercitrin could convert HbSS erythrocyte metabolomes to be like HbAA. Sickling was associated with changes in antioxidants, anaerobic bioenergy, and arachidonic acid metabolism, all of which were reversed by quercitrin. The findings described could inform efforts directed to the development of an anti-sickling drug or quality control assessments of A. cordifolia preparations.
Collapse
Affiliation(s)
- Olayemi Adeniyi
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
- Biochemistry Unit, Department of Science Technology, The Federal Polytechnic, Ado-Ekiti 360231, Nigeria
| | - Rafael Baptista
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
| | - Sumana Bhowmick
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
| | - Alan Cookson
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
| | - Robert J. Nash
- PhytoQuest Ltd., Plas Gogerddan, Aberystwyth SY23 3EB, UK;
| | - Ana Winters
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
| | - Jianying Shen
- Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Correspondence: (J.S.); (L.A.J.M.)
| | - Luis A. J. Mur
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (O.A.); (R.B.); (S.B.); (A.C.); (A.W.)
- Correspondence: (J.S.); (L.A.J.M.)
| |
Collapse
|
23
|
Antibacterial Activity of a Novel Oligosaccharide from Streptomyces californics against Erwinia carotovora subsp. Carotovora. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27082384. [PMID: 35458585 PMCID: PMC9032947 DOI: 10.3390/molecules27082384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/25/2022] [Accepted: 04/03/2022] [Indexed: 11/26/2022]
Abstract
The present study aims to characterize and predict models for antibacterial activity of a novel oligosaccharide from Streptomyces californics against Erwinia carotovora subsp. carotovora using an adaptive neuro-fuzzy inference system and an artificial neural network. The mathematical predication models were used to determine the optimal conditions to produce oligosaccharide and determine the relationship between the factors (pH, temperature, and time). The characteristics of the purified antibacterial agent were determined using ultraviolet spectroscopy (UV/Vis), infrared spectroscopy (FT-IR), nuclear magnetic resonance spectroscopy (1H- and 13C-NMR), and mass spectrometry (MS). The best performances for the model were 39.45 and 35.16 recorded at epoch 1 for E. carotovora Erw5 and E. carotovora EMCC 1687, respectively. The coefficient (R2) of the training was more than 0.90. The highest antimicrobial production was recorded after 9 days at 25 °C and a pH of 6.2, at which more than 17 mm of the inhibition zone was obtained. The mass spectrum of antimicrobial agent (peak at R.T. = 3.433 of fraction 6) recorded two molecular ion peaks at m/z = 703.70 and m/z = 338.30, corresponding to molecular weights of 703.70 and 338.30 g/mol, respectively. The two molecular ion peaks matched well with the molecular formulas C29H53NO18 and C14H26O9, respectively, which were obtained from the elemental analysis result. A novel oligosaccharide from Streptomyces californics with potential activity against E. carotovora EMCC 1687 and E. carotovora Erw5 was successfully isolated, purified, and characterized.
Collapse
|
24
|
Hutchins M, Bovill RA, Stephens PJ, Brazier JA, Osborn HMI. Glycosides of Nadifloxacin-Synthesis and Antibacterial Activities against Methicillin-Resistant Staphylococcus aureus. Molecules 2022; 27:1504. [PMID: 35268604 PMCID: PMC8912027 DOI: 10.3390/molecules27051504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 11/16/2022] Open
Abstract
The increase in the number of bacteria that are resistant to multiple antibiotics poses a serious clinical problem that threatens the health of humans worldwide. Nadifloxacin (1) is a highly potent antibacterial agent with broad-spectrum activity. However, its poor aqueous solubility has limited its use to topical applications. To increase its solubility, it was glycosylated herein to form a range of trans-linked (3a-e) and cis-linked (7a,b) glycosides, each of which was prepared and purified to afford single anomers. The seven glycoside derivatives (3a-e, 7a,b) were examined for potency against eight strains of S. aureus, four of which were methicillin-resistant. Although less potent than free nadifloxacin (1), the α-L-arabinofuransoside (3a) was effective against all strains that were tested (minimum inhibitory concentrations of 1-8 μg/mL compared to 0.1-0.25 μg/mL for nadifloxacin), demonstrating the potential of this glycoside as an antibacterial agent. Estimation of Log P as well as observations made during preparation of these compounds reveal that the solubilities of the glycosides were greatly improved compared with nadifloxacin (1), raising the prospect of its use in oral applications.
Collapse
Affiliation(s)
- Mark Hutchins
- ThermoFisher Scientific, Wade Road, Basingstoke RG24 8PW, Hampshire, UK
| | - Richard A. Bovill
- ThermoFisher Scientific, Wade Road, Basingstoke RG24 8PW, Hampshire, UK
| | - Peter J. Stephens
- ThermoFisher Scientific, Wade Road, Basingstoke RG24 8PW, Hampshire, UK
| | - John A. Brazier
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD, Berkshire, UK
| | - Helen M. I. Osborn
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD, Berkshire, UK
| |
Collapse
|
25
|
Li X, Wu J, Tang W. General Strategy for the Synthesis of Rare Sugars via Ru(II)-Catalyzed and Boron-Mediated Selective Epimerization of 1,2- trans-Diols to 1,2- cis-Diols. J Am Chem Soc 2022; 144:3727-3736. [PMID: 35168319 DOI: 10.1021/jacs.1c13399] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Human glycans are primarily composed of nine common sugar building blocks. On the other hand, several hundred monosaccharides have been discovered in bacteria and most of them are not readily available. The ability to access these rare sugars and the corresponding glycoconjugates can facilitate the studies of various fundamentally important biological processes in bacteria, including interactions between microbiota and the human host. Many rare sugars also exist in a variety of natural products and pharmaceutical reagents with significant biological activities. Although several methods have been developed for the synthesis of rare monosaccharides, most of them involve lengthy steps. Herein, we report an efficient and general strategy that can provide access to rare sugars from commercially available common monosaccharides via a one-step Ru(II)-catalyzed and boron-mediated selective epimerization of 1,2-trans-diols to 1,2-cis-diols. The formation of boronate esters drives the equilibrium toward 1,2-cis-diol products, which can be immediately used for further selective functionalization and glycosylation. The utility of this strategy was demonstrated by the efficient construction of glycoside skeletons in natural products or bioactive compounds.
Collapse
Affiliation(s)
- Xiaolei Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jicheng Wu
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Weiping Tang
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
26
|
Weiz G, Molejon MI, Malvicini M, Sukowati CHC, Tiribelli C, Mazzolini G, Breccia JD. Glycosylated 4-methylumbelliferone as a targeted therapy for hepatocellular carcinoma. Liver Int 2022; 42:444-457. [PMID: 34800352 DOI: 10.1111/liv.15084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/22/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Reaching efficacious drug delivery to target cells/tissues represents a major obstacle in the current treatment of solid malignancies including hepatocellular carcinoma (HCC). In this study, we developed a pipeline to selective add complex-sugars to the aglycone 4-methylumbelliferone (4MU) to help their bioavailability and tumour cell intake. METHODS The therapeutic efficacy of sugar-modified rutinosyl-4-methylumbelliferone (4MUR) and 4MU were compared in vitro and in an orthotopic HCC model established in fibrotic livers. The mechanistic bases of its selective target to liver tumour cells were evaluated by the interaction with asialoglycoprotein receptor (ASGPR), the mRNA expression of hyaluronan synthases (HAS2 or HAS3) and hyaluronan deposition. RESULTS 4MUR showed a significant antiproliferative effect on liver tumoural cells as compared to non-tumoural cells in a dose-dependent manner. Further analysis showed that 4MUR is incorporated mostly into HCC cells by interaction with ASGPR, a receptor commonly overexpressed in HCC cells. 4MUR-treatment decreased the levels of HAS2 and HAS3 and the cytoplasmic deposition of hyaluronan. Moreover, 4MUR reduced CFSC-2G activation, hence reducing the fibrosis. In vivo efficacy showed that 4MUR treatment displayed a greater tumour growth inhibition and increased survival in comparison to 4MU. 4MUR administration was associated with a significant reduction of liver fibrosis without any signs of tissue damage. Further, 60% of 4MUR treated mice did not present macroscopically tumour mass post-treatment. CONCLUSION Our results provide evidence that 4MUR may be used as an effective HCC therapy, without damaging non-tumoural cells or other organs, most probably due to the specific targeting.
Collapse
Affiliation(s)
- Gisela Weiz
- Facultad de Ciencias Exactas y Naturales, Instituto de Ciencias de la Tierra y Ambientales de La Pampa (INCITAP), Universidad Nacional de La Pampa - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLPam-CONICET), Santa Rosa, Argentina
| | - Maria I Molejon
- Facultad de Ciencias Exactas y Naturales, Instituto de Ciencias de la Tierra y Ambientales de La Pampa (INCITAP), Universidad Nacional de La Pampa - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLPam-CONICET), Santa Rosa, Argentina
| | - Mariana Malvicini
- Laboratorio de Inmunobiología del Cáncer, Instituto de Investigaciones en Medicina Traslacional (IIMT) Facultad de Ciencias Biomédicas, CONICET, Universidad Austral, Derqui-Pilar, Argentina
| | | | - Claudio Tiribelli
- Fondazione Italiana Fegato, AREA Science Park Basovizza, Trieste, Italy
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET, Universidad Austral, Derqui-Pilar, Argentina
- Liver Unit, Hospital Universitario Austral, Universidad Austral, Derqui-Pilar, Argentina
| | - Javier D Breccia
- Facultad de Ciencias Exactas y Naturales, Instituto de Ciencias de la Tierra y Ambientales de La Pampa (INCITAP), Universidad Nacional de La Pampa - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLPam-CONICET), Santa Rosa, Argentina
| |
Collapse
|
27
|
Kumar M, Gurawa A, Kumar N, Kashyap S. Bismuth-Catalyzed Stereoselective 2-Deoxyglycosylation of Disarmed/Armed Glycal Donors. Org Lett 2022; 24:575-580. [PMID: 34995079 DOI: 10.1021/acs.orglett.1c04008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bi(OTf)3 promoted direct and highly stereoselective glycosylation of "disarmed" and "armed" glycals to synthesize 2-deoxyglycosides has been reported. The tunable and solvent-controlled chemoselective activation of deactivated glycal donors distinguishing the competitive Ferrier and 1,2-addition pathways was discovered to improve substrate scope. The practical versatility of the method has been amply demonstrated with the oligosaccharide syntheses and 2-deoxyglycosylation of high-value natural products and drugs.
Collapse
Affiliation(s)
- Manoj Kumar
- Carbohydrate Chemistry Research Laboratory (CCRL), Department of Chemistry, MNIT, Jaipur 302017, India
| | - Aakanksha Gurawa
- Carbohydrate Chemistry Research Laboratory (CCRL), Department of Chemistry, MNIT, Jaipur 302017, India
| | - Nitin Kumar
- Carbohydrate Chemistry Research Laboratory (CCRL), Department of Chemistry, MNIT, Jaipur 302017, India
| | - Sudhir Kashyap
- Carbohydrate Chemistry Research Laboratory (CCRL), Department of Chemistry, MNIT, Jaipur 302017, India
| |
Collapse
|
28
|
Beerens K, Gevaert O, Desmet T. GDP-Mannose 3,5-Epimerase: A View on Structure, Mechanism, and Industrial Potential. Front Mol Biosci 2022; 8:784142. [PMID: 35087867 PMCID: PMC8787198 DOI: 10.3389/fmolb.2021.784142] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
GDP-mannose 3,5-epimerase (GM35E, GME) belongs to the short-chain dehydrogenase/reductase (SDR) protein superfamily and catalyses the conversion of GDP-d-mannose towards GDP-l-galactose. Although the overall reaction seems relatively simple (a double epimerization), the enzyme needs to orchestrate a complex set of chemical reactions, with no less than 6 catalysis steps (oxidation, 2x deprotonation, 2x protonation and reduction), to perform the double epimerization of GDP-mannose to GDP-l-galactose. The enzyme is involved in the biosynthesis of vitamin C in plants and lipopolysaccharide synthesis in bacteria. In this review, we provide a clear overview of these interesting epimerases, including the latest findings such as the recently characterized bacterial and thermostable GM35E representative and its mechanism revision but also focus on their industrial potential in rare sugar synthesis and glycorandomization.
Collapse
Affiliation(s)
| | | | - Tom Desmet
- *Correspondence: Koen Beerens, ; Tom Desmet,
| |
Collapse
|
29
|
Mendoza F, Jaña GA. The inverting mechanism of the metal ion-independent LanGT2: the first step to understand the glycosylation of natural product antibiotic precursors through QM/MM simulations. Org Biomol Chem 2021; 19:5888-5898. [PMID: 34132308 DOI: 10.1039/d1ob00544h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Glycosyltransferases (GTs) from the GT1 family are responsible for the glycosylation of various important organic structures such as terpenes, steroids and peptide antibiotics, making it one of the most intensely studied families of GTs. The target of our study, LanGT2, is a member of the GT1 family that uses an inverting mechanism for transferring olivose from TDP-olivose, the donor substrate, to the natural product tetrangulol (Tet), the precursor of the antibiotic landomycin A. X-ray crystallography in conjunction with mutagenesis experiments has revealed the catalytic significance of 3 amino acids (Ser10, Ser219 and Asp137), suggesting Asp137 as the base catalyst. In the absence of X-ray structures that include the acceptor substrate Tet, in silico experiments and MD simulations that have modeled ternary complexes propose that Asp137 could recruit a water molecule to facilitate the nucleophilic activation of Tet, since the distance between Asp137 and the nucleophile is too long to directly deprotonate the nucleophilic moiety. So far, there is no computational evidence regarding the precise mechanism by which LanGT2 catalyzes the transfer of olivose, which raises questions such as: is a water-assisted mechanism possible? and how does this metal ion-independent GT stabilize the growing negative charge of the diphosphate leaving group? In this work, the QM/MM approach was used to unravel the catalytic mechanism of LanGT2, and to identify the role of crucial catalytic amino acids at a molecular level. Our calculations show that the minimum energy path (MEP) describes an SN2-like mechanism, identifying an oxocarbenium ion-like TS in which the olivosyl moiety adopts a 4H3 conformation. Interactions established between the diphosphate group of TDP and Ser10, Ser219, Arg220 and His283 are key to stabilize the development of charge on the leaving group. Our work also suggests that a water-mediated proton transfer mechanism is feasible, in which the water molecule is key to stabilize the phenolate ion-like nucleophile in the TS. This is the first computational insight into the inverting mechanism of an antibiotic natural product GT, and its implications may serve to guide the design of new biocatalysts for natural product glycodiversification.
Collapse
Affiliation(s)
- Fernanda Mendoza
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Autopista Concepción-Talcahuano 7100, Talcahuano, Chile.
| | - Gonzalo A Jaña
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Autopista Concepción-Talcahuano 7100, Talcahuano, Chile.
| |
Collapse
|
30
|
Novel fidaxomicin antibiotics through site-selective catalysis. Commun Chem 2021; 4:59. [PMID: 36697765 PMCID: PMC9814943 DOI: 10.1038/s42004-021-00501-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/31/2021] [Indexed: 01/28/2023] Open
Abstract
Fidaxomicin (FDX) is a marketed antibiotic for the treatment of Clostridioides difficile infections (CDI). Fidaxomicin displays antibacterial properties against many Gram-positive bacteria, yet the application of this antibiotic is currently limited to treatment of CDI. Semisynthetic modifications present a promising strategy to improve its pharmacokinetic properties and also circumvent resistance development by broadening the structural diversity of the derivatives. Here, based on a rational design using cryo-EM structural analysis, we implement two strategic site-selective catalytic reactions with a special emphasis to study the role of the carbohydrate units. Site-selective introduction of various ester moieties on the noviose as well as a Tsuji-Trost type rhamnose cleavage allow the synthesis of novel fidaxomicin analogs with promising antibacterial activities against C. difficile and Mycobacterium tuberculosis.
Collapse
|
31
|
Meng S, Li X, Zhu J. Recent advances in direct synthesis of 2-deoxy glycosides and thioglycosides. Tetrahedron 2021. [DOI: 10.1016/j.tet.2021.132140] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
32
|
Cristófalo AE, Uhrig ML. Synthetic Studies on the Incorporation of N-Acetylallosamine in Hyaluronic Acid-Inspired Thiodisaccharides. Molecules 2021; 26:E180. [PMID: 33401465 PMCID: PMC7796257 DOI: 10.3390/molecules26010180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
Two approaches for the synthesis of the thiodisaccharide β-S-GlcA(1→3)β-S-AllNAc are described here. The target disaccharide was a C-3 epimer and thio-analogue of the hyaluronic acid repetitive unit, tuned with a thiopropargyl anomeric group for further click conjugation. Thus, we analysed and tested two convenient sequences, combining the two key steps required to introduce the thioglycosidic bonds and consequently reach the target molecule: the SN2 substitution of a good leaving group (triflate) present at C-3 of a GlcNAc derivative and the introduction of the anomeric thiopropargyl substituent. The use of a 2-azido precursor showed to be a convenient substrate for the SN2 step. Nevertheless, further protecting group manipulation and the introduction of the thiopropargyl anomeric residue were then required. This approach showed to provide access to a variety of thiodisaccharide derivatives as interesting building blocks for the construction of neoglycoconjugates.
Collapse
Affiliation(s)
- Alejandro E. Cristófalo
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, Buenos Aires C1428EGA, Argentina;
- Centro de Investigaciones en Hidratos de Carbono (CIHIDECAR), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - María Laura Uhrig
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, Buenos Aires C1428EGA, Argentina;
- Centro de Investigaciones en Hidratos de Carbono (CIHIDECAR), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| |
Collapse
|
33
|
Sridharan AP, Sugitha T, Karthikeyan G, Nakkeeran S, Sivakumar U. Metabolites of Trichoderma longibrachiatum EF5 inhibits soil borne pathogen, Macrophomina phaseolina by triggering amino sugar metabolism. Microb Pathog 2020; 150:104714. [PMID: 33383148 DOI: 10.1016/j.micpath.2020.104714] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 01/07/2023]
Abstract
An endophytic fungal antagonist Trichoderma longibrachiatum EF5 exhibited biocontrol activity against a soil-borne fungal pathogen Macrophomina phaseolina. Under dual co-culture, T. longibrachiatum EF5 showed 58% inhibition against M. phaseolina. Crude soluble metabolites (SMs) extracted from EF5 exhibited biocontrol activity (61%), which is more significant than the cell-free extract. Dual culture of both T. longibrachiatum EF5 and M. phaseolina displayed entangled mycelial structures and retarded hyphal growth. The metabolites responsible for antibiosis and pathogenic activity profiled through GC-MS revealed a total of 131 SMs from axenic culture and upon the interaction of T. longibrachiatum EF5 and M. phaseolina. Interestingly, potential plant-growth-promoting and antimicrobial compounds such as 1- pentanol, 1-hexanol, myristonyl pantothenate, bisabolol, d-Alanine, and diethyl trisulphide were unique with T. longibrachiatum EF5. Few compounds that were not observed or produced minimally under axenic culture were increased during their interaction (e.g., 1,6-anhydro-á-d-Glucopyranose and 5-heptyl dihydro-2(3H)-Furanone), suggesting antimicrobial action against the pathogen. This study also unraveled the induction of amino sugar metabolism when T. longibrachiatum EF5 interacts with M. phaseolina, which is responsible for colonization and counterfeiting the pathogen. Hence T. longibrachiatum EF5 could be a potential biocontrol agent employed for defense priming and plant growth promotion.
Collapse
Affiliation(s)
- A P Sridharan
- Department of Plant Pathology, Tamil Nadu Agricultural University, Coimbatore, 641003, India
| | - Thangappan Sugitha
- Biocatalysts Laboratory, Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, 641003, India
| | - G Karthikeyan
- Department of Plant Pathology, Tamil Nadu Agricultural University, Coimbatore, 641003, India
| | - S Nakkeeran
- Department of Plant Pathology, Tamil Nadu Agricultural University, Coimbatore, 641003, India
| | - Uthandi Sivakumar
- Biocatalysts Laboratory, Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, 641003, India.
| |
Collapse
|
34
|
Liu Z, Zhao S, Sun X, Mao X. Biological synthesis and anti-HeLa cells effect of glycosylated bafilomycins. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.08.025] [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: 11/26/2022]
|
35
|
Cao J, Tamura M, Hosaka R, Nakayama A, Hasegawa JY, Nakagawa Y, Tomishige K. Mechanistic Study on Deoxydehydration and Hydrogenation of Methyl Glycosides to Dideoxy Sugars over a ReO x–Pd/CeO 2 Catalyst. ACS Catal 2020. [DOI: 10.1021/acscatal.0c02309] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Ji Cao
- Department of Applied Chemistry, School of Engineering, Tohoku University, 6-6-07, Aoba, Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | - Masazumi Tamura
- Research Center for Artificial Photosynthesis, Advanced Research Institute for Natural Science and Technology, Osaka City University, Osaka 558-8585, Japan
| | - Ryu Hosaka
- Institute for Catalysis, Hokkaido University, Sapporo 001-0021, Japan
| | - Akira Nakayama
- Institute for Catalysis, Hokkaido University, Sapporo 001-0021, Japan
- Department of Chemical System Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Jun-ya Hasegawa
- Institute for Catalysis, Hokkaido University, Sapporo 001-0021, Japan
| | - Yoshinao Nakagawa
- Department of Applied Chemistry, School of Engineering, Tohoku University, 6-6-07, Aoba, Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | - Keiichi Tomishige
- Department of Applied Chemistry, School of Engineering, Tohoku University, 6-6-07, Aoba, Aramaki, Aoba-ku, Sendai 980-8579, Japan
| |
Collapse
|
36
|
Cereijo AE, Kuhn ML, Hernández MA, Ballicora MA, Iglesias AA, Alvarez HM, Asencion Diez MD. Study of duplicated galU genes in Rhodococcus jostii and a putative new metabolic node for glucosamine-1P in rhodococci. Biochim Biophys Acta Gen Subj 2020; 1865:129727. [PMID: 32890704 DOI: 10.1016/j.bbagen.2020.129727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/11/2020] [Accepted: 08/30/2020] [Indexed: 01/10/2023]
Abstract
BACKGOUND Studying enzymes that determine glucose-1P fate in carbohydrate metabolism is important to better understand microorganisms as biotechnological tools. One example ripe for discovery is the UDP-glucose pyrophosphorylase enzyme from Rhodococcus spp. In the R. jostii genome, this gene is duplicated, whereas R. fascians contains only one copy. METHODS We report the molecular cloning of galU genes from R. jostii and R. fascians to produce recombinant proteins RjoGalU1, RjoGalU2, and RfaGalU. Substrate saturation curves were conducted, kinetic parameters were obtained and the catalytic efficiency (kcat/Km) was used to analyze enzyme promiscuity. We also investigated the response of R. jostii GlmU pyrophosphorylase activity with different sugar-1Ps, which may compete for substrates with RjoGalU2. RESULTS All enzymes were active as pyrophosphorylases and exhibited substrate promiscuity toward sugar-1Ps. Remarkably, RjoGalU2 exhibited one order of magnitude higher activity with glucosamine-1P than glucose-1P, the canonical substrate. Glucosamine-1P activity was also significant in RfaGalU. The efficient use of the phospho-amino-sugar suggests the feasibility of the reaction to occur in vivo. Also, RjoGalU2 and RfaGalU represent enzymatic tools for the production of (amino)glucosyl precursors for the putative synthesis of novel molecules. CONCLUSIONS Results support the hypothesis that partitioning of glucosamine-1P includes an uncharacterized metabolic node in Rhodococcus spp., which could be important for producing diverse alternatives for carbohydrate metabolism in biotechnological applications. GENERAL SIGNIFICANCE Results presented here provide a model to study evolutionary enzyme promiscuity, which could be used as a tool to expand an organism's metabolic repertoire by incorporating non-canonical substrates into novel metabolic pathways.
Collapse
Affiliation(s)
- A E Cereijo
- Instituto de Agrobiotecnología del Litoral (UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, CCT-Santa Fe, Colectora Ruta Nac 168 km 0, 3000 Santa Fe, Argentina
| | - M L Kuhn
- Department of Chemistry and Biochemistry, San Francisco State University, 1600 Holloway Ave., San Francisco, CA, United States
| | - M A Hernández
- Instituto de Biociencias de la Patagonia (INBIOP), Universidad Nacional de la Patagonia San Juan Bosco y CONICET, Km 4-Ciudad Universitaria 9000, Comodoro Rivadavia, Chubut, Argentina
| | - M A Ballicora
- Department of Chemistry and Biochemistry, Loyola University Chicago, 1068 W. Sheridan Rd., Chicago, IL 60660, United States
| | - A A Iglesias
- Instituto de Agrobiotecnología del Litoral (UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, CCT-Santa Fe, Colectora Ruta Nac 168 km 0, 3000 Santa Fe, Argentina
| | - H M Alvarez
- Instituto de Biociencias de la Patagonia (INBIOP), Universidad Nacional de la Patagonia San Juan Bosco y CONICET, Km 4-Ciudad Universitaria 9000, Comodoro Rivadavia, Chubut, Argentina.
| | - M D Asencion Diez
- Instituto de Agrobiotecnología del Litoral (UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, CCT-Santa Fe, Colectora Ruta Nac 168 km 0, 3000 Santa Fe, Argentina.
| |
Collapse
|
37
|
Bardasov IN, Alekseeva AY, Ershov OV, Mar’yasov MA. Antiproliferative Activity of N-Substituted 2,4-Diamino-5-Aryl-5,6,7,8,9,10-Hexahydrobenzo[B][1,8]Naphthyridine-3-Carbonitriles. Pharm Chem J 2020. [DOI: 10.1007/s11094-020-02236-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
38
|
Bashyal P, Thapa SB, Kim TS, Pandey RP, Sohng JK. Exploring the Nucleophilic N- and S-Glycosylation Capacity of Bacillus licheniformis YjiC Enzyme. J Microbiol Biotechnol 2020; 30:1092-1096. [PMID: 32238768 PMCID: PMC9728172 DOI: 10.4014/jmb.2001.01024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022]
Abstract
YjiC, a glycosyltransferase from Bacillus licheniformis, is a well-known versatile enzyme for glycosylation of diverse substrates. Although a number of O-glycosylated products have been produced using YjiC, no report has been updated for nucleophilic N-, S-, and C- glycosylation. Here, we report the additional functional capacity of YjiC for nucleophilic N- and S- glycosylation using a broad substrate spectrum including UDP-α-D-glucose, UDP-N-acetyl glucosamine, UDP-N-acetylgalactosamine, UDP-α-D-glucuronic acid, TDP-α-L-rhamnose, TDP-α-D-viosamine, and GDP-α-Lfucose as donor and various amine and thiol groups containing natural products as acceptor substrates. The results revealed YjiC as a promiscuous enzyme for conjugating diverse sugars at amine and thiol functional groups of small molecules applicable for generating glycofunctionalized chemical diversity libraries. The glycosylated products were analyzed using HPLC and LC/MS and compared with previous reports.
Collapse
Affiliation(s)
- Puspalata Bashyal
- Department of Life Science and Biochemical Engineering, Sun Moon University, Asan 31460, Republic of Korea
| | - Samir Bahadur Thapa
- Department of Life Science and Biochemical Engineering, Sun Moon University, Asan 31460, Republic of Korea
| | - Tae-Su Kim
- Department of Life Science and Biochemical Engineering, Sun Moon University, Asan 31460, Republic of Korea
| | - Ramesh Prasad Pandey
- Department of Life Science and Biochemical Engineering, Sun Moon University, Asan 31460, Republic of Korea,Department of Department of Pharmaceutical Engineering and Biotechnology, Sun Moon University, Asan 31460, Republic of Korea,Corresponding authors J.K.S. Phone: +82-41-530-2246 Fax: +82-41-544-2919 E-mail:
| | - Jae Kyung Sohng
- Department of Life Science and Biochemical Engineering, Sun Moon University, Asan 31460, Republic of Korea,Corresponding authors J.K.S. Phone: +82-41-530-2246 Fax: +82-41-544-2919 E-mail:
| |
Collapse
|
39
|
Identification and Characterization of a Novel N- and O-Glycosyltransferase from Saccharopolyspora erythraea. Molecules 2020; 25:molecules25153400. [PMID: 32727097 PMCID: PMC7435583 DOI: 10.3390/molecules25153400] [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: 06/18/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 11/26/2022] Open
Abstract
Glycosyltransferases are important enzymes which are often used as tools to generate novel natural products. In this study, we describe the identification and characterization of an inverting N- and O-glycosyltransferase from Saccharopolyspora erythraea NRRL2338. When feeding experiments with 1,4-diaminoanthraquinone in Saccharopolyspora erythraea were performed, the formation of new compounds (U3G and U3DG) was observed by HPLC-MS. Structure elucidation by NMR revealed that U3G consists of two compounds, N1-α-glucosyl-1,4-diaminoanthraquinone and N1-β-glucosyl-1,4-diaminoanthraquinone. Based on UV and MS data, U3DG is a N1,N4-diglucosyl-1,4-diaminoanthraquinone. In order to find the responsible glycosyltransferase, gene deletion experiments were performed and we identified the glycosyltransferase Sace_3599, which belongs to the CAZy family 1. When Streptomyces albus J1074, containing the dTDP-d-glucose synthase gene oleS and the plasmid pUWL-A-sace_3599, was used as host, U3 was converted to the same compounds. Protein production in Escherichia coli and purification of Sace_3599 was carried out. The enzyme showed glycosyl hydrolase activity and was able to produce mono- and di-N-glycosylated products in vitro. When UDP-α-d-glucose was used as a sugar donor, U3 was stereoselective converted to N1-β-glucosyl-1,4-diaminoanthraquinone and N1,N4-diglucosyl-1,4-diaminoanthraquinone. The use of 1,4-dihydroxyanthraquinone as a substrate in in vitro experiments also led to the formation of mono-glucosylated and di-glucosylated products, but in lower amounts. Overall, we identified and characterized a novel glycosyltransferase which shows glycohydrolase activity and the ability to glycosylate “drug like” structures forming N- and O-glycosidic bonds.
Collapse
|
40
|
Kumar M, Reddy TR, Gurawa A, Kashyap S. Copper(ii)-catalyzed stereoselective 1,2-addition vs. Ferrier glycosylation of "armed" and "disarmed" glycal donors. Org Biomol Chem 2020; 18:4848-4862. [PMID: 32608448 DOI: 10.1039/d0ob01042a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Selective activation of "armed' and ''disarmed" glycal donors enabling the stereo-controlled glycosylations by employing Cu(ii)-catalyst as the promoter has been realized. The distinctive stereochemical outcome in the process is mainly influenced by the presence of diverse protecting groups on the donor and the solvent system employed. The protocol is compatible with a variety of aglycones including carbohydrates, amino acids, and natural products to access deoxy-glycosides and glycoconjugates with high α-anomeric selectivity. Notably, the synthetic practicality of the method is amply verified for the stereoselective assembling of trisaccharides comprising 2-deoxy components. Mechanistic studies involving deuterated experiments validate the syn-diastereoselective 1,2-addition of acceptors on the double bond of armed donors.
Collapse
Affiliation(s)
- Manoj Kumar
- Carbohydrate Chemistry Research Laboratory (CCRL), Department of Chemistry, Malaviya National Institute of Technology (MNIT), Jaipur-302017, India.
| | - Thurpu Raghavender Reddy
- Carbohydrate Chemistry Research Laboratory (CCRL), Department of Chemistry, Malaviya National Institute of Technology (MNIT), Jaipur-302017, India.
| | - Aakanksha Gurawa
- Carbohydrate Chemistry Research Laboratory (CCRL), Department of Chemistry, Malaviya National Institute of Technology (MNIT), Jaipur-302017, India.
| | - Sudhir Kashyap
- Carbohydrate Chemistry Research Laboratory (CCRL), Department of Chemistry, Malaviya National Institute of Technology (MNIT), Jaipur-302017, India.
| |
Collapse
|
41
|
Gucchait A, Kundu M, Manna T, Shit P, Misra AK. Influence of Functional Groups towards the β-Selective Glycosylation of 2-Azido-2-deoxy Glycosyl Thioglycosides. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000392] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Arin Gucchait
- Division of Molecular Medicine; Bose Institute; P-1/12, C.I.T. Scheme VII M 700054 Kolkata India
| | - Monalisa Kundu
- Division of Molecular Medicine; Bose Institute; P-1/12, C.I.T. Scheme VII M 700054 Kolkata India
| | - Tapasi Manna
- Division of Molecular Medicine; Bose Institute; P-1/12, C.I.T. Scheme VII M 700054 Kolkata India
| | - Pradip Shit
- Division of Molecular Medicine; Bose Institute; P-1/12, C.I.T. Scheme VII M 700054 Kolkata India
| | - Anup Kumar Misra
- Division of Molecular Medicine; Bose Institute; P-1/12, C.I.T. Scheme VII M 700054 Kolkata India
| |
Collapse
|
42
|
Martins-Teixeira MB, Carvalho I. Antitumour Anthracyclines: Progress and Perspectives. ChemMedChem 2020; 15:933-948. [PMID: 32314528 DOI: 10.1002/cmdc.202000131] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Indexed: 12/31/2022]
Abstract
Anthracyclines are ranked among the most effective chemotherapeutics against cancer. They are glycoside drugs comprising the amino sugar daunosamine linked to a hydroxy anthraquinone aglycone, and act by DNA intercalation, oxidative stress generation and topoisomerase II poisoning. Regardless of their therapeutic value, multidrug resistance and severe cardiotoxicity are important limitations of anthracycline treatment that have prompted the discovery of novel analogues. This review covers the most clinically relevant anthracyclines and their development over decades, since the first discovered natural prototypes to recent semisynthetic and synthetic derivatives. These include registered drugs, drug candidates undergoing clinical trials, and compounds under pre-clinical investigation. The impact of the structural modifications on antitumour activity, toxicity and resistance profile is addressed.
Collapse
Affiliation(s)
- Maristela B Martins-Teixeira
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo Avenida do Café s/n Monte Alegre, Ribeirão Preto, 14040903, Brazil
| | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo Avenida do Café s/n Monte Alegre, Ribeirão Preto, 14040903, Brazil
| |
Collapse
|
43
|
Wiltschi B, Cernava T, Dennig A, Galindo Casas M, Geier M, Gruber S, Haberbauer M, Heidinger P, Herrero Acero E, Kratzer R, Luley-Goedl C, Müller CA, Pitzer J, Ribitsch D, Sauer M, Schmölzer K, Schnitzhofer W, Sensen CW, Soh J, Steiner K, Winkler CK, Winkler M, Wriessnegger T. Enzymes revolutionize the bioproduction of value-added compounds: From enzyme discovery to special applications. Biotechnol Adv 2020; 40:107520. [DOI: 10.1016/j.biotechadv.2020.107520] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 10/18/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022]
|
44
|
Mrudulakumari Vasudevan U, Lee EY. Flavonoids, terpenoids, and polyketide antibiotics: Role of glycosylation and biocatalytic tactics in engineering glycosylation. Biotechnol Adv 2020; 41:107550. [PMID: 32360984 DOI: 10.1016/j.biotechadv.2020.107550] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/19/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
Abstract
Flavonoids, terpenoids, and polyketides are structurally diverse secondary metabolites used widely as pharmaceuticals and nutraceuticals. Most of these molecules exist in nature as glycosides, in which sugar residues act as a decisive factor in their architectural complexity and bioactivity. Engineering glycosylation through selective trimming or extension of the sugar residues in these molecules is a prerequisite to their commercial production as well to creating novel derivatives with specialized functions. Traditional chemical glycosylation methods are tedious and can offer only limited end-product diversity. New in vitro and in vivo biocatalytic tools have emerged as outstanding platforms for engineering glycosylation in these three classes of secondary metabolites to create a large repertoire of versatile glycoprofiles. As knowledge has increased about secondary metabolite-associated promiscuous glycosyltransferases and sugar biosynthetic machinery, along with phenomenal progress in combinatorial biosynthesis, reliable industrial production of unnatural secondary metabolites has gained momentum in recent years. This review highlights the significant role of sugar residues in naturally occurring flavonoids, terpenoids, and polyketide antibiotics. General biocatalytic tools used to alter the identity and pattern of sugar molecules are described, followed by a detailed illustration of diverse strategies used in the past decade to engineer glycosylation of these valuable metabolites, exemplified with commercialized products and patents. By addressing the challenges involved in current bio catalytic methods and considering the perspectives portrayed in this review, exceptional drugs, flavors, and aromas from these small molecules could come to dominate the natural-product industry.
Collapse
Affiliation(s)
| | - Eun Yeol Lee
- Department of Chemical Engineering, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| |
Collapse
|
45
|
Cheng J, Zhao W, Yao H, Shen Y, Zhang Y, Li YZ, QI Q, Wongprasert K, Tang YJ. Discovery of 4,6-O-Thenylidene-β-d-glucopyranoside-(2″-acetamido, 3″-acetyl-di-S-5-fluorobenzothizole/5-fluorobenzoxazole)-4′-demethylepipodophyllotoxin as Potential Less Toxic Antitumor Candidate Drugs by Reducing DNA Damage and Less Inhibition of PI3K. J Med Chem 2020; 63:2877-2893. [DOI: 10.1021/acs.jmedchem.9b01354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Jie Cheng
- Hubei Key Laboratory of Industrial Microbiology, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Wei Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Hui Yao
- Hubei Key Laboratory of Industrial Microbiology, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| | - Yuemao Shen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Youming Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Yue-zhong Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Qingsheng QI
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Kanokpan Wongprasert
- Department of Anatomy, Faculty of Science, Mahidol University, Rama Sixth Road, Bangkok 10400, Thailand
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| |
Collapse
|
46
|
Vera C, Guerrero C, Aburto C, Cordova A, Illanes A. Conventional and non-conventional applications of β-galactosidases. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140271. [DOI: 10.1016/j.bbapap.2019.140271] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/15/2019] [Accepted: 08/30/2019] [Indexed: 02/04/2023]
|
47
|
Hamama WS, Gouda MA, Kamal El‐din HA, Zoorob HH. Highlights on the synthesis of novel phenothiazine‐based azines scaffold as antioxidant agents. J Heterocycl Chem 2020. [DOI: 10.1002/jhet.3771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Wafaa S. Hamama
- Department of Chemistry, Faculty of ScienceMansoura University El‐Gomhoria Street Mansoura Egypt
| | - Moustafa A. Gouda
- Department of Chemistry, Faculty of ScienceMansoura University El‐Gomhoria Street Mansoura Egypt
- Department of Chemistry, Faculty of Science and Arts, UllaTaibah University Medina Saudi Arabia
| | - Hadwah A. Kamal El‐din
- Department of Chemistry, Faculty of ScienceMansoura University El‐Gomhoria Street Mansoura Egypt
| | - Hanafi H. Zoorob
- Department of Chemistry, Faculty of ScienceMansoura University El‐Gomhoria Street Mansoura Egypt
| |
Collapse
|
48
|
Mendoza F, Jaña GA. Unveiling the Dynamical and Structural Features That Determine the Orientation of the Acceptor Substrate in the Landomycin Glycosyltransferase LanGT2 and Its Variant with C-Glycosylation Activity. J Chem Inf Model 2019; 60:933-943. [DOI: 10.1021/acs.jcim.9b00865] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Fernanda Mendoza
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Autopista Concepción-Talcahuano 7100, Talcahuano, Chile
| | - Gonzalo A. Jaña
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Autopista Concepción-Talcahuano 7100, Talcahuano, Chile
| |
Collapse
|
49
|
Weiz G, Mazzaferro LS, Kotik M, Neher BD, Halada P, Křen V, Breccia JD. The flavonoid degrading fungus Acremonium sp. DSM 24697 produces two diglycosidases with different specificities. Appl Microbiol Biotechnol 2019; 103:9493-9504. [PMID: 31705182 DOI: 10.1007/s00253-019-10180-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/30/2019] [Accepted: 10/09/2019] [Indexed: 01/01/2023]
Abstract
AbstractDiglycosidases hydrolyze the heterosidic linkage of diglycoconjugates, releasing the disaccharide and the aglycone. Usually, these enzymes do not hydrolyze or present only low activities towards monoglycosylated compounds. The flavonoid degrading fungus Acremonium sp. DSM 24697 produced two diglycosidases, which were termed 6-O-α-rhamnosyl-β-glucosidase I and II (αRβG I and II) because of their function of releasing the disaccharide rutinose (6-O-α-L-rhamnosyl-β-D-glucose) from the diglycoconjugates hesperidin or rutin. In this work, the genome of Acremonium sp. DSM 24697 was sequenced and assembled with a size of ~ 27 Mb. The genes encoding αRβG I and II were expressed in Pichia pastoris KM71 and the protein products were purified with apparent molecular masses of 42 and 82 kDa, respectively. A phylogenetic analysis showed that αRβG I grouped in glycoside hydrolase family 5, subfamily 23 (GH5), together with other fungal diglycosidases whose substrate specificities had been reported to be different from αRβG I. On the other hand, αRβG II grouped in glycoside hydrolase family 3 (GH3) and thus is the first GH3 member that hydrolyzes the heterosidic linkage of rutinosylated compounds. The substrate scopes of the enzymes were different: αRβG I showed exclusive specificity toward 7-O-β-rutinosyl flavonoids, whereas αRβG II hydrolyzed both 7-O-β-rutinosyl- and 3-O-β-rutinosyl- flavonoids. None of the enzymes displayed activity toward 7-O-β-neohesperidosyl- flavonoids. The recombinant enzymes also exhibited transglycosylation activities, transferring rutinose from hesperidin or rutin onto various alcoholic acceptors. The different substrate scopes of αRβG I and II may be part of an optimized strategy of the original microorganism to utilize different carbon sources.
Collapse
Affiliation(s)
- Gisela Weiz
- Facultad de Ciencias Exactas y Naturales, Instituto de Ciencias de la Tierra y Ambientales de La Pampa (INCITAP), Universidad Nacional de La Pampa - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLPam-CONICET), Av. Uruguay 151, 6300, Santa Rosa, La Pampa, Argentina
| | - Laura S Mazzaferro
- Facultad de Ciencias Exactas y Naturales, Instituto de Ciencias de la Tierra y Ambientales de La Pampa (INCITAP), Universidad Nacional de La Pampa - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLPam-CONICET), Av. Uruguay 151, 6300, Santa Rosa, La Pampa, Argentina
| | - Michael Kotik
- Laboratory of Biotransformation, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Bárbara D Neher
- Facultad de Ciencias Exactas y Naturales, Instituto de Ciencias de la Tierra y Ambientales de La Pampa (INCITAP), Universidad Nacional de La Pampa - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLPam-CONICET), Av. Uruguay 151, 6300, Santa Rosa, La Pampa, Argentina
| | - Petr Halada
- Laboratory of Molecular Structure Characterization, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Vladimír Křen
- Laboratory of Biotransformation, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Javier D Breccia
- Facultad de Ciencias Exactas y Naturales, Instituto de Ciencias de la Tierra y Ambientales de La Pampa (INCITAP), Universidad Nacional de La Pampa - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLPam-CONICET), Av. Uruguay 151, 6300, Santa Rosa, La Pampa, Argentina.
| |
Collapse
|
50
|
Saha M, Singha S, Chakraborty M, Mazumdar S, Kumar S, Karmakar P, Das S. Characterization of a MnII complex of Alizarin suggests attributes explaining a superior anticancer activity: A comparison with anthracycline drugs. Polyhedron 2019. [DOI: 10.1016/j.poly.2019.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|