1
|
Zhao X, Chen J, Shan M, Sun P, Qu X, Qin LP, Cheng G. Identification of potential IL4I1 inhibitors through structure-based virtual screening and molecular dynamics simulations. J Biomol Struct Dyn 2025:1-24. [PMID: 40411435 DOI: 10.1080/07391102.2025.2501666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 06/24/2024] [Indexed: 05/26/2025]
Abstract
Interleukin-4-induced gene 1 (IL4I1) is an L-phenylalanine oxidase. As the primary enzyme responsible for degrading tryptophan, IL4I1 generates indole metabolites and kynurenic acid, which act as crucial endogenous ligands to activate the aryl hydrocarbon receptor (AHR). This activation enhances tumor survivability while suppressing the body's anti-tumor immune response. Consequently, IL4I1 is now recognized as a promising new target for drug development in the realm of cancer immunomodulation. In this study, we employed a strategy combining AlphaFold2 with molecular dynamics (MD) simulations to model receptor conformations our docking model achieved a regression fit with an R2 coefficient of 0.34, providing a robust framework for structure-based virtual screening aimed at identifying potential IL4I1 inhibitors. We then applied this structure-based virtual screening method to a compound library. After further MD simulation and following Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) calculation of binding free energy and ADMET analysis, five candidate IL4I1 inhibitors were obtained. This study provides an effective in silico approach for the identification of IL4I1 inhibitors and offers a valuable reference for the virtual screening of inhibitors targeting other proteins without known structures.
Collapse
Affiliation(s)
- Xuan Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Mengyi Shan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peng Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - XinHao Qu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu-Ping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Gang Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Han M, Yoon SH, Lee J, Chung TD, Song WJ. Redirecting electron flows in glutamate oxidases by selective anchoring of osmium complexes. Chem Sci 2025; 16:7433-7441. [PMID: 40160359 PMCID: PMC11948343 DOI: 10.1039/d5sc00166h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025] Open
Abstract
l-Glutamate is the most abundant and essential excitatory neurotransmitter in the nervous system. However, its direct electrochemical detection is challenging due to its inherently non-electroactive nature. In this study, we redesigned l-glutamate oxidase (GlutOx) by covalently attaching osmium polypyridyl complexes as electron mediators at selected sites. Most engineered enzymes retained their native catalytic activity, while exhibiting significantly altered catalytic currents during l-glutamate oxidation, depending on the proximity, orientation, and microenvironments of the osmium complexes relative to the FAD cofactors. Notably, two mutants significantly enhanced catalytic currents, revealing selectively and efficiently rerouted electron transfer pathways from the enzyme active site to Os complexes. These findings provide an effective strategy for designing redox-active enzymes for electrochemical biosensors.
Collapse
Affiliation(s)
- Minjung Han
- Department of Chemistry, Seoul National University Seoul 08826 Republic of Korea
| | - Sun-Heui Yoon
- Department of Chemistry, Seoul National University Seoul 08826 Republic of Korea
| | - Jaehee Lee
- Department of Chemistry, Seoul National University Seoul 08826 Republic of Korea
| | - Taek Dong Chung
- Department of Chemistry, Seoul National University Seoul 08826 Republic of Korea
- Advanced Institute of Convergence Technology Suwon-Si Gyeonggi-do 16229 Republic of Korea
| | - Woon Ju Song
- Department of Chemistry, Seoul National University Seoul 08826 Republic of Korea
| |
Collapse
|
3
|
Yang Q, Liu S, Sun A, Li X, Liu T, Han X, Mao J. Salt-Resistant and Ethanol-Resistant Monoamine Oxidases: New Sight for yobN Mining from Bacillus and Biogenic Amine Degradation Mechanism in Fermented Food. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9714-9731. [PMID: 40223567 DOI: 10.1021/acs.jafc.4c13223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Amine oxidases have strong capabilities for the degradation of biogenic amines (BAs) in fermented foods. However, their application is limited by substrate specificity, and high concentrations of ethanol and salt can hinder their effectiveness. This study presents a novel approach utilizing comparative genomics, protein clustering analysis, and full homology modeling to identify three amine oxidases: KCYOBN from the salt-resistant Bacillus subtilis, and LYYOBN1 and LYYOBN2 from the ethanol-resistant Bacillus cereus. These enzymes are highly similar in structure and exhibit broad substrate specificities. KCYOBN maintains over 84% relative activity at 20% (w/v) NaCl, while LYYOBN1 and LYYOBN2 retain over 32 and 21% relative activity at 25% vol ethanol. Variations in key residues are one of the reasons for the differences in tolerance. In fermented foods, KCYOBN degraded 45.97% of the total BAs in cooking wine and 38.33% in fish sauce. LYYOBN1 achieved the highest degradation rate of 32.93% in huangjiu. LYYOBN2 exhibited degradation rates of 30.00% in soy sauce and 35.14% in wine with no significant impact on flavor compounds. The significance of this work lies in the identification of the novel salt-resistant KCYOBN and ethanol-resistant LYYOBN1 and LYYOBN2 through this new method, which can simultaneously degrade multiple BAs and have a broad application potential.
Collapse
Affiliation(s)
- Qilin Yang
- State Key Laboratory of Food Science and Technology, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Shuangping Liu
- State Key Laboratory of Food Science and Technology, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Shaoxing Key Laboratory of Traditional Fermentation Food and Human Health, Jiangnan University (Shaoxing) Industrial Technology Research Institute, Shaoxing, Zhejiang 312000, China
- National Engineering Research Center of Huangjiu, Zhejiang Guyuelongshan Shaoxing Wine Co., Ltd., Shaoxing, Zhejiang 312000, China
| | - Aibao Sun
- National Engineering Research Center of Huangjiu, Zhejiang Guyuelongshan Shaoxing Wine Co., Ltd., Shaoxing, Zhejiang 312000, China
| | - Xin Li
- Jiangsu Hengshun Vinegar Industry Co., Ltd., Zhenjiang 212000, China
| | - Tiantian Liu
- State Key Laboratory of Food Science and Technology, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Shaoxing Key Laboratory of Traditional Fermentation Food and Human Health, Jiangnan University (Shaoxing) Industrial Technology Research Institute, Shaoxing, Zhejiang 312000, China
- National Engineering Research Center of Huangjiu, Zhejiang Guyuelongshan Shaoxing Wine Co., Ltd., Shaoxing, Zhejiang 312000, China
| | - Xiao Han
- State Key Laboratory of Food Science and Technology, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Shaoxing Key Laboratory of Traditional Fermentation Food and Human Health, Jiangnan University (Shaoxing) Industrial Technology Research Institute, Shaoxing, Zhejiang 312000, China
- National Engineering Research Center of Huangjiu, Zhejiang Guyuelongshan Shaoxing Wine Co., Ltd., Shaoxing, Zhejiang 312000, China
| | - Jian Mao
- State Key Laboratory of Food Science and Technology, National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Shaoxing Key Laboratory of Traditional Fermentation Food and Human Health, Jiangnan University (Shaoxing) Industrial Technology Research Institute, Shaoxing, Zhejiang 312000, China
- National Engineering Research Center of Huangjiu, Zhejiang Guyuelongshan Shaoxing Wine Co., Ltd., Shaoxing, Zhejiang 312000, China
| |
Collapse
|
4
|
Yang C, Ding L, He Q, Chen X, Zhu H, Chen F, Yang W, Pan Y, Tai Z, Zhang W, Yu Z, Chen Z, Yu X. Proteomic Profiling of Venoms from Bungarus suzhenae and B. bungaroides: Enzymatic Activities and Toxicity Assessment. Toxins (Basel) 2024; 16:494. [PMID: 39591249 PMCID: PMC11598402 DOI: 10.3390/toxins16110494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/23/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Kraits are venomous snakes of the genus Bungarus from the family Elapidae. Their venom typically demonstrates neurotoxicity; however, the toxicity is significantly influenced by the snake's species and geographical origin. Among the Bungarus species, Bungarus suzhenae and B. bungaroides have been poorly studied, with little to no information available regarding their venom composition. In this study, a proteomic approach was employed using LC-MS/MS to identify proteins from trypsin-digested peptides. The analysis revealed 102 venom-related proteins from 18 distinct functional protein families in the venom of B. suzhenae, with the primary components being three-finger toxins (3-FTx, 25.84%), phospholipase A2 (PLA2, 40.29%), L-amino acid oxidase (LAAO, 10.33%), Kunitz-type serine protease inhibitors (KUN, 9.48%), and snake venom metalloproteinases (SVMPs, 6.13%). In the venom of B. bungaroides, 99 proteins from 17 families were identified, with primary components being 3-FTx (33.87%), PLA2 (37.91%), LAAO (4.21%), and KUN (16.60%). Enzymatic activity assays confirmed the presence of key venom enzymes. Additionally, the LD50 values for B. suzhenae and B. bungaroides were 0.0133 μg/g and 0.752 μg/g, respectively, providing a reference for toxicity studies of these two species. This research elucidates the proteomic differences in the venoms of these two species, offering a foundation for developing antivenoms and clinical treatments for envenomation.
Collapse
Affiliation(s)
- Chenying Yang
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| | - Li Ding
- Laboratory of Amphibians and Reptiles, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
| | - Qiyi He
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| | - Xiya Chen
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| | - Haiting Zhu
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| | - Feng Chen
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| | - Wanzhou Yang
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| | - Yuexin Pan
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| | - Zhiyuan Tai
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| | - Wenhao Zhang
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| | - Zeyuan Yu
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| | - Zening Chen
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection, Guangxi Normal University, Ministry of Education, Guilin 541006, China;
| | - Xiaodong Yu
- Animal Toxin Group, Engineering Research Center of Active Substance and Biotechnology, Ministry of Education, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (C.Y.); (Q.H.); (X.C.); (H.Z.); (F.C.); (W.Y.); (Y.P.); (Z.T.); (W.Z.); (Z.Y.)
| |
Collapse
|
5
|
Ding SM, Yap MKK. Deciphering toxico-proteomics of Asiatic medically significant venomous snake species: A systematic review and interactive data dashboard. Toxicon 2024; 250:108120. [PMID: 39393539 DOI: 10.1016/j.toxicon.2024.108120] [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: 08/07/2024] [Revised: 09/30/2024] [Accepted: 10/06/2024] [Indexed: 10/13/2024]
Abstract
Snakebite envenomation (SBE) is a neglected tropical disease (NTD) with an approximate 1.8 million cases annually. The tremendous figure is concerning, and the currently available treatment for snakebite envenomation is antivenom. However, the current antivenom has limited cross-neutralisation activity due to the variations in snake venom composition across species and geographical locations. The proteomics of medically important venomous species is essential as they study the venom compositions within and among different species. The advancement of sophisticated proteomic approaches allows intensive investigation of snake venoms. Nevertheless, there is a need to consolidate the venom proteomics profiles and distribution analysis to examine their variability patterns. This review systematically analysed the proteomics and toxicity profiles of medically important venomous species from Asia across different geographical locations. An interactive dashboard - Asiatic Proteomics Interactive Datasets was curated to consolidate the distribution patterns of the venom compositions, serve as a comprehensive directory for large-scale comparative meta-analyses. The population proteomics demonstrate higher diversities in the predominant venom toxins. Besides, inter-regional differences were also observed in Bungarus sp., Naja sp., Calliophis sp., and Ophiophagus hannah venoms. The elapid venoms are predominated with three-finger toxins (3FTXs) and phospholipase A2 (PLA2). Intra-regional variation is only significantly observed in Naja naja venoms. Proteomics diversity is more prominent in viper venoms, with widespread dominance observed in snake venom metalloproteinase (SVMP) and snake venom serine protease (SVSP). Correlations exist between the proteomics profiles and the toxicity (LD50) of the medically important venomous species. Additionally, the predominant toxins, alongside their pathophysiological effects, were highlighted and discussed as well. The insights of interactive toxico-proteomics datasets provide comprehensive frameworks of venom dynamics and contribute to developing antivenoms for snakebite envenomation. This could reduce misdiagnosis of SBE and accelerate the researchers' data mining process.
Collapse
Affiliation(s)
- Sher Min Ding
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | | |
Collapse
|
6
|
Koopmeiners S, Gilzer D, Widmann C, Berelsmann N, Sproß J, Niemann HH, Fischer von Mollard G. Crystal structure and enzyme engineering of the broad substrate spectrum l-amino acid oxidase 4 from the fungus Hebeloma cylindrosporum. FEBS Lett 2024; 598:2306-2320. [PMID: 39152524 DOI: 10.1002/1873-3468.15002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/19/2024]
Abstract
l-Amino acid oxidases (LAAOs) catalyze the oxidative deamination of l-amino acids to α-keto acids. Recombinant production of LAAOs with broad substrate spectrum remains a formidable challenge. We previously achieved this for the highly active and thermostable LAAO4 of Hebeloma cylindrosporum (HcLAAO4). Here, we crystallized a proteolytically truncated surface entropy reduction variant of HcLAAO4 and solved its structure in substrate-free form and in complex with diverse substrates. The ability to support the aliphatic portion of a substrate's side chain by an overall hydrophobic active site is responsible for the broad substrate spectrum of HcLAAO4, including l-amino acids with big aromatic, acidic and basic side chains. Based on the structural findings, we generated an E288H variant with increased activity toward pharmaceutical building blocks of high interest.
Collapse
Affiliation(s)
- Simon Koopmeiners
- Biochemistry III, Department of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Dominic Gilzer
- Structural Biochemistry, Department of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Christiane Widmann
- Structural Biochemistry, Department of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Nils Berelsmann
- Biochemistry III, Department of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Jens Sproß
- Industrial Organic Chemistry and Biotechnology, Department of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Hartmut H Niemann
- Structural Biochemistry, Department of Chemistry, Bielefeld University, Bielefeld, Germany
| | | |
Collapse
|
7
|
Doyle MGJ, Mair BA, Sib A, Bsharat O, Munch M, Derdau V, Rotstein BH, Lundgren RJ. A practical guide for the preparation of C1-labeled α-amino acids using aldehyde catalysis with isotopically labeled CO 2. Nat Protoc 2024; 19:2147-2179. [PMID: 38548937 DOI: 10.1038/s41596-024-00974-4] [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: 07/04/2023] [Accepted: 01/09/2024] [Indexed: 07/10/2024]
Abstract
Isotopically carbon-labeled α-amino acids are valuable synthetic targets that are increasingly needed in pharmacology and medical imaging. Existing preparations rely on early stage introduction of the isotopic label, which leads to prohibitive synthetic costs and time-intensive preparations. Here we describe a protocol for the preparation of C1-labeled α-amino acids using simple aldehyde catalysts in conjunction with [*C]CO2 (* = 14, 13, 11). This late-stage labeling strategy is enabled by the one-pot carboxylate exchange of unprotected α-amino acids with [*C]CO2. The protocol consists of three separate procedures, describing the syntheses of (±)-[1-13C]phenylalanine, (±)-[1-11C]phenylalanine and (±)-[1-14C]phenylalanine from unlabeled phenylalanine. Although the delivery of [*C]CO2 is operationally distinct for each experiment, each procedure relies on the same fundamental chemistry and can be executed by heating the reaction components at 50-90 °C under basic conditions in dimethylsulfoxide. Performed on scales of up to 0.5 mmol, this methodology is amenable to C1-labeling of many proteinogenic α-amino acids and nonnatural derivatives, which is a breakthrough from existing methods. The synthesis of (±)-[1-13C]phenylalanine requires ~2 d, with product typically obtained in a 60-80% isolated yield (n = 3, μ = 71, σ = 8.3) with an isotopic incorporation of 70-88% (n = 18, μ = 72, σ = 9.0). Starting from the preformed imino acid (~3 h preparation time), rapid synthesis of (±)-[1-11C]phenylalanine can be completed in ~1 h with an isolated radiochemical yield of 13%. Finally, (±)-[1-14C]phenylalanine can be accessed in ~2 d with a 51% isolated yield and 11% radiochemical yield.
Collapse
Affiliation(s)
- Michael G J Doyle
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Braeden A Mair
- Department of Biochemistry, Microbiology and Immunology and Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Anna Sib
- Sanofi Germany, R&D, Integrated Drug Discovery, Isotope Chemistry, Industriepark Höchst, Frankfurt, Germany
| | - Odey Bsharat
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Maxime Munch
- Department of Biochemistry, Microbiology and Immunology and Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Volker Derdau
- Sanofi Germany, R&D, Integrated Drug Discovery, Isotope Chemistry, Industriepark Höchst, Frankfurt, Germany
| | - Benjamin H Rotstein
- Department of Biochemistry, Microbiology and Immunology and Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada.
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
| | - Rylan J Lundgren
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
8
|
Guo X, Fu Y, Peng J, Fu Y, Dong S, Ding RB, Qi X, Bao J. Emerging anticancer potential and mechanisms of snake venom toxins: A review. Int J Biol Macromol 2024; 269:131990. [PMID: 38704067 DOI: 10.1016/j.ijbiomac.2024.131990] [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: 11/27/2023] [Revised: 03/13/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Animal-derived venom, like snake venom, has been proven to be valuable natural resources for the drug development. Previously, snake venom was mainly investigated in its pharmacological activities in regulating coagulation, vasodilation, and cardiovascular function, and several marketed cardiovascular drugs were successfully developed from snake venom. In recent years, snake venom fractions have been demonstrated with anticancer properties of inducing apoptotic and autophagic cell death, restraining proliferation, suppressing angiogenesis, inhibiting cell adhesion and migration, improving immunity, and so on. A number of active anticancer enzymes and peptides have been identified from snake venom toxins, such as L-amino acid oxidases (LAAOs), phospholipase A2 (PLA2), metalloproteinases (MPs), three-finger toxins (3FTxs), serine proteinases (SPs), disintegrins, C-type lectin-like proteins (CTLPs), cell-penetrating peptides, cysteine-rich secretory proteins (CRISPs). In this review, we focus on summarizing these snake venom-derived anticancer components on their anticancer activities and underlying mechanisms. We will also discuss their potential to be developed as anticancer drugs in the future.
Collapse
Affiliation(s)
- Xijun Guo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Yuanfeng Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Junbo Peng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ying Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Shuai Dong
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ren-Bo Ding
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Xingzhu Qi
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China.
| | - Jiaolin Bao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
9
|
Tasima LJ, Lima EOVD, Hatakeyama DM, Vidueiros JP, Stuginski DR, Grego KF, Tanaka-Azevedo AM. Seasonality in Crotalus durissus venom. Toxicon 2024; 244:107748. [PMID: 38710309 DOI: 10.1016/j.toxicon.2024.107748] [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: 02/26/2024] [Revised: 04/22/2024] [Accepted: 05/04/2024] [Indexed: 05/08/2024]
Abstract
Rattlesnakes belonging to the genus Crotalus are widely distributed throughout the Americas. In Brazil, symptoms commonly associated with envenomation by Crotalus durissus collilineatus include myalgia, rhabdomyolysis, renal failure, neurotoxicity, and progressive paralysis, which are related to the protein composition of this venom. Snake venom composition exhibits compositional variability that may reflect geographic distribution, age, captivity, diet, sex, and even individual genetics. Although seasonality is also considered a possible source of variation, there are few reports of such variability in snake venom. In this work, venoms of the same eight C. durissus collilineatus were extracted every three months for two years, to analyze seasonal changes in composition and activities. To this end, venom composition was analyzed by protein quantification, SDS-PAGE, and HPLC, and the LAAO, PLA2 and coagulant activities were measured. Venoms of these C. d. collilineatus showed minor seasonal differences in venom activities and no composition differences were found. LAAO and coagulant activities displayed a pattern of seasonal change, while PLA2 activity seemed to have no seasonality tendency. Also, there are sexual differences, in which males seem to be more stable than females in regard to some activities. Individual variability occurs even in seasonal variation of activities, highlighting the importance of controlling circumstances of venom extraction before comparing results between groups of snakes.
Collapse
Affiliation(s)
- Lidia Jorge Tasima
- Laboratory of Herpetology, Butantan Institute, São Paulo, 05503-900, SP, Brazil; Interunidades Em Biotecnologia, Instituto de Ciências Biomédicas-Instituto de Pesquisas Tecnológicas-Instituto Butantan, Universidade de São Paulo, São Paulo, 05508-900, SP, Brazil
| | - Eduardo Oliveira Venâncio de Lima
- Laboratory of Herpetology, Butantan Institute, São Paulo, 05503-900, SP, Brazil; Interunidades Em Biotecnologia, Instituto de Ciências Biomédicas-Instituto de Pesquisas Tecnológicas-Instituto Butantan, Universidade de São Paulo, São Paulo, 05508-900, SP, Brazil
| | - Daniela Miki Hatakeyama
- Laboratory of Herpetology, Butantan Institute, São Paulo, 05503-900, SP, Brazil; Interunidades Em Biotecnologia, Instituto de Ciências Biomédicas-Instituto de Pesquisas Tecnológicas-Instituto Butantan, Universidade de São Paulo, São Paulo, 05508-900, SP, Brazil
| | | | | | | | - Anita Mitico Tanaka-Azevedo
- Laboratory of Herpetology, Butantan Institute, São Paulo, 05503-900, SP, Brazil; Interunidades Em Biotecnologia, Instituto de Ciências Biomédicas-Instituto de Pesquisas Tecnológicas-Instituto Butantan, Universidade de São Paulo, São Paulo, 05508-900, SP, Brazil.
| |
Collapse
|
10
|
Neri-Castro E, Zarzosa V, Benard-Valle M, Rodríguez-Solís AM, Hernández-Orihuela L, Ortiz-Medina JA, Alagón A. Quantifying venom production: A study on Micrurus snakes in Mexico. Toxicon 2024; 240:107658. [PMID: 38395261 DOI: 10.1016/j.toxicon.2024.107658] [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: 01/17/2024] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Our study quantifies venom production in nine Mexican coral snake species (Micrurus), encompassing 76 specimens and 253 extractions. Noteworthy variations were observed, with M. diastema and M. laticollaris displaying diverse yields, ranging from 0.3 mg to 59 mg. For animals for which we have length data, there is a relationship between size and venom quantity. Twenty-eight percent of the observed variability in venom production can be explained by snake size, suggesting that other factors influence the amount of obtained venom. These findings are pivotal for predicting venom effects and guiding antivenom interventions. Our data offer insights into Micrurus venom yields, laying the groundwork for future research and aiding in medical response strategies. This study advances understanding coral snake venom production, facilitating informed medical responses to coral snake bites.
Collapse
Affiliation(s)
- Edgar Neri-Castro
- Investigador por México, Facultad de Ciencias Biológicas, Universidad Juárez del Estado de Durango, Av. Universidad s/n. Fracc. Filadelfia, C.P. 35010, Gómez Palacio, Dgo., Mexico; Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Chamilpa, C.P. 62210, Cuernavaca, Mor., Mexico.
| | - Vanessa Zarzosa
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Chamilpa, C.P. 62210, Cuernavaca, Mor., Mexico
| | - Melisa Benard-Valle
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, 2800, Kongens Lyngby, Denmark
| | - Audrey Michelle Rodríguez-Solís
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Chamilpa, C.P. 62210, Cuernavaca, Mor., Mexico
| | - Lorena Hernández-Orihuela
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Chamilpa, C.P. 62210, Cuernavaca, Mor., Mexico
| | - Javier A Ortiz-Medina
- Departamento de Sistemática y Ecología Acuática, El Colegio de la Frontera Sur, Unidad Chetumal, Avenida Centenario km 5.5, 77014, Chetumal Quintana Roo, Mexico; Unidad de Manejo para la conservación de la vida silvestre Tsáab Kaan, Baca, Yucatán, Mexico; HERP.MX A.C., Villa de Álvarez, Colima, Mexico
| | - Alejandro Alagón
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Chamilpa, C.P. 62210, Cuernavaca, Mor., Mexico
| |
Collapse
|
11
|
Truong NV, Phan TTT, Hsu TS, Phu Duc P, Lin LY, Wu WG. Action mechanism of snake venom l-amino acid oxidase and its double-edged sword effect on cancer treatment: Role of pannexin 1-mediated interleukin-6 expression. Redox Biol 2023; 64:102791. [PMID: 37385076 PMCID: PMC10331595 DOI: 10.1016/j.redox.2023.102791] [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: 05/02/2023] [Accepted: 06/16/2023] [Indexed: 07/01/2023] Open
Abstract
Snake venom l-amino acid oxidases (svLAAOs) have been recognized as promising candidates for anticancer therapeutics. However, multiple aspects of their catalytic mechanism and the overall responses of cancer cells to these redox enzymes remain ambiguous. Here, we present an analysis of the phylogenetic relationships and active site-related residues among svLAAOs and reveal that the previously proposed critical catalytic residue His 223 is highly conserved in the viperid but not the elapid svLAAO clade. To gain further insight into the action mechanism of the elapid svLAAOs, we purify and characterize the structural, biochemical, and anticancer therapeutic potentials of the Thailand elapid snake Naja kaouthia LAAO (NK-LAAO). We find that NK-LAAO, with Ser 223, exhibits high catalytic activity toward hydrophobic l-amino acid substrates. Moreover, NK-LAAO induces substantial oxidative stress-mediated cytotoxicity with the magnitude relying on both the levels of extracellular hydrogen peroxide (H2O2) and intracellular reactive oxygen species (ROS) generated during the enzymatic redox reactions, but not being influenced by the N-linked glycans on its surface. Unexpectedly, we discover a tolerant mechanism deployed by cancer cells to dampen the anticancer activities of NK-LAAO. NK-LAAO treatment amplifies interleukin (IL)-6 expression via the pannexin 1 (Panx1)-directed intracellular calcium (iCa2+) signaling pathway to confer adaptive and aggressive phenotypes on cancer cells. Accordingly, IL-6 silencing renders cancer cells vulnerable to NK-LAAO-induced oxidative stress together with abrogating NK-LAAO-stimulated metastatic acquisition. Collectively, our study urges caution when using svLAAOs in cancer treatment and identifies the Panx1/iCa2+/IL-6 axis as a therapeutic target for improving the effectiveness of svLAAOs-based anticancer therapies.
Collapse
Affiliation(s)
- Nam V Truong
- Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Trinh T T Phan
- Institute of Molecular and Cellular Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Phan Phu Duc
- Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC
| | - Lih-Yuan Lin
- Institute of Molecular and Cellular Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC.
| | - Wen-Guey Wu
- Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University, Hsinchu, 300044, Taiwan, ROC.
| |
Collapse
|
12
|
Yildiz I. Computational Mechanistic Study of l-Aspartate Oxidase by ONIOM Method. ACS OMEGA 2023; 8:19963-19968. [PMID: 37305300 PMCID: PMC10249383 DOI: 10.1021/acsomega.3c01949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023]
Abstract
l-Aspartate oxidase (Laspo) is responsible for the oxidation of l-aspartate into iminoaspartate using flavin as a cofactor. During this process flavin is reduced, and it can be reoxidized by either molecular oxygen or fumarate. The overall fold and the catalytic residues of Laspo are similar to succinate dehydrogenase and fumarate reductase. On the basis of deuterium kinetic isotope effects as well as other kinetic and structural data, it is proposed that the enzyme can catalyze the oxidation of l-aspartate through a mechanism similar to amino acid oxidases. It is suggested that a proton is removed from the α-amino group, while a hydride is transferred from C2 to flavin. It is also suggested that the hydride transfer is a rate-limiting step. However, there is still an ambiguity about the stepwise or concerted mechanism of hydride- and proton-transfer steps. In this study, we formulated some computational models to study the hydride-transfer mechanism using the crystal structure of Escherichia colil-aspartate oxidase in complexes with succinate. The calculations involved our own N-layered integrated molecular orbital and molecular mechanics method, and we evaluated the geometry and energetics of the hydride/proton-transfer processes while probing the roles of active site residues. Based on the calculations, it is concluded that proton- and hydride-transfer steps are decoupled, and a stepwise mechanism might be operative as opposed to the concerted one.
Collapse
Affiliation(s)
- Ibrahim Yildiz
- Chemistry Department, Khalifa
University, P.O. Box, 127788 Abu Dhabi, UAE
| |
Collapse
|
13
|
Muellers SN, Tararina MA, Kuzmanovic U, Galagan JE, Allen KN. Structural Insights into the Substrate Range of a Bacterial Monoamine Oxidase. Biochemistry 2023; 62:851-862. [PMID: 36662673 DOI: 10.1021/acs.biochem.2c00540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Monoamine oxidases (MAOs) play a key role in the breakdown of primary and secondary amines. In eukaryotic organisms, these enzymes are vital to the regulation of monoamine neurotransmitters and the degradation of dietary monoamines. MAOs have also been identified in prokaryotic species, although their role in these organisms is not well understood. Here, we report the biophysical and structural properties of a promiscuous, bacterial MAO from Corynebacterium ammoniagenes (caMAO). caMAO catalyzes the oxidation of a number of monoamine substrates including dopamine and norepinephrine, as well as exhibiting some activity with polyamine substrates such as cadaverine. The X-ray crystal structures of Michaelis complexes with seven substrates show that conserved hydrophobic interactions and hydrogen-bonding pattern (for polar substrates) allow the broad specificity range. The structure of caMAO identifies an unusual cysteine (Cys424) residue in the so-called "aromatic cage", which flanks the flavin isoalloxazine ring in the active site. Site-directed mutagenesis, steady-state kinetics in air-saturated buffer, and UV-vis spectroscopy revealed that Cys424 plays a role in the pH dependence and modulation of electrostatics within the caMAO active site. Notably, bioinformatic analysis shows a propensity for variation at this site within the "aromatic cage" of the flavin amine oxidase (FAO) superfamily. Structural analysis also identified the conservation of a secondary substrate inhibition site, present in a homologous member of the superfamily. Finally, genome neighborhood diagram analysis of caMAO in the context of the FAO superfamily allows us to propose potential roles for these bacterial MAOs in monoamine and polyamine degradation and catabolic pathways related to scavenging of nitrogen.
Collapse
Affiliation(s)
- Samantha N Muellers
- Department of Chemistry, Boston University, Boston, Massachusetts02215, United States
| | - Margarita A Tararina
- Program in Biomolecular Pharmacology, Boston University School of Medicine, Boston, Massachusetts02118, United States
| | - Uros Kuzmanovic
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts02215, United States
| | - James E Galagan
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts02215, United States
| | - Karen N Allen
- Department of Chemistry, Boston University, Boston, Massachusetts02215, United States
| |
Collapse
|
14
|
The secretory phenotypes of envenomed cells: Insights into venom cytotoxicity. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:193-230. [PMID: 36707202 DOI: 10.1016/bs.apcsb.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Snake envenomation is listed as Category A Neglected Tropical Diseases (NTD) by World Health Organization, indicates a severe public health problem. The global figures for envenomation cases are estimated to be more than 1.8 million annually. Even if the affected victims survive the envenomation, they might suffer from permanent morbidity due to local envenomation. One of the most prominent local envenomation is dermonecrosis. Dermonecrosis is a pathophysiological outcome of envenomation that often causes disability in the victims due to surgical amputations, deformities, contracture, and chronic ulceration. The key venom toxins associated with this local symptom are mainly attributed to substantial levels of enzymatic and non-enzymatic toxins as well as their possible synergistic actions. Despite so, the severity of the local tissue damage is based on macroscopic observation of the bite areas. Furthermore, limited knowledge is known about the key biomarkers involved in the pathogenesis of dermonecrosis. The current immunotherapy with antivenom is also ineffective against dermonecrosis. These local effects eventually end up as sequelae. There is also a global shortage of toxins-targeted therapeutics attributed to inadequate knowledge of the actual molecular mechanisms of cytotoxicity. This chapter discusses the characterization of secretory phenotypes of dermonecrosis as an advanced tool to indicate its severity and pathogenesis in envenomation. Altogether, the secretory phenotypes of envenomed cells and tissues represent the precise characteristics of dermonecrosis caused by venom toxins.
Collapse
|
15
|
A current perspective on snake venom composition and constituent protein families. Arch Toxicol 2023; 97:133-153. [PMID: 36437303 DOI: 10.1007/s00204-022-03420-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/09/2022] [Indexed: 11/28/2022]
Abstract
Snake venoms are heterogeneous mixtures of proteins and peptides used for prey subjugation. With modern proteomics there has been a rapid expansion in our knowledge of snake venom composition, resulting in the venom proteomes of 30% of vipers and 17% of elapids being characterised. From the reasonably complete proteomic coverage of front-fanged snake venom composition (179 species-68 species of elapids and 111 species of vipers), the venoms of vipers and elapids contained 42 different protein families, although 18 were only reported in < 5% of snake species. Based on the mean abundance and occurrence of the 42 protein families, they can be classified into 4 dominant, 6 secondary, 14 minor, and 18 rare protein families. The dominant, secondary and minor categories account for 96% on average of a snake's venom composition. The four dominant protein families are: phospholipase A2 (PLA2), snake venom metalloprotease (SVMP), three-finger toxins (3FTx), and snake venom serine protease (SVSP). The six secondary protein families are: L-amino acid oxidase (LAAO), cysteine-rich secretory protein (CRiSP), C-type lectins (CTL), disintegrins (DIS), kunitz peptides (KUN), and natriuretic peptides (NP). Venom variation occurs at all taxonomic levels, including within populations. The reasons for venom variation are complex, as variation is not always associated with geographical variation in diet. The four dominant protein families appear to be the most important toxin families in human envenomation, being responsible for coagulopathy, neurotoxicity, myotoxicity and cytotoxicity. Proteomic techniques can be used to investigate the toxicological profile of a snake venom and hence identify key protein families for antivenom immunorecognition.
Collapse
|
16
|
Mora-Obando D, Lomonte B, Pla D, Guerrero-Vargas JA, Ayerbe-González S, Gutiérrez JM, Sasa M, Calvete JJ. Half a century of research on Bothrops asper venom variation: Biological and biomedical implications. Toxicon 2022; 221:106983. [DOI: 10.1016/j.toxicon.2022.106983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
|
17
|
Costa MN, Silva RN. Cytotoxic activity of l-lysine alpha-oxidase against leukemia cells. Semin Cancer Biol 2022; 86:590-599. [PMID: 34606983 DOI: 10.1016/j.semcancer.2021.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/27/2023]
Abstract
Cancer cells exhibit higher proliferation rates than normal cells, and as a consequence, a higher nutritional demand for metabolites such as amino acids. Such cells demonstrate high expression of amino acid transporters and are significantly dependent on the external uptake of amino acids. Moreover, some types of cancer cells exhibit oncogenic mutations that render them auxotrophic to certain amino acids. This metabolic difference between tumor and normal cells has been explored for developing anticancer drugs. Enzymes capable of depleting certain amino acids in the bloodstream can be employed to inhibit the proliferation of cancer cells and promote cell death. Certain microbial enzymes, such as l-asparaginase and l-amino acid oxidases, have been studied for this purpose. In this paper, we discuss the role of l-asparaginase, the only enzyme currently used as a chemotherapeutic agent. We also review the studies on a new potential antineoplastic agent, l-lysine α-oxidase, an enzyme of l-amino acid oxidase family.
Collapse
Affiliation(s)
- Mariana N Costa
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | - Roberto N Silva
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil.
| |
Collapse
|
18
|
Mamounis KJ, Caldas Nogueira ML, Marchi Salvador DP, Andreo-Vidal A, Sanchez-Amat A, Davidson VL. Structural Determinants of the Specific Activities of an L-Amino Acid Oxidase from Pseudoalteromonas luteoviolacea CPMOR-1 with Broad Substrate Specificity. Molecules 2022; 27:molecules27154726. [PMID: 35897902 PMCID: PMC9331233 DOI: 10.3390/molecules27154726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022] Open
Abstract
The Pseudoalteromonas luteoviolacea strain CPMOR-1 expresses a flavin adenine dinucleotide (FAD)-dependent L-amino acid oxidase (LAAO) with broad substrate specificity. Steady-state kinetic analysis of its reactivity towards the 20 proteinogenic amino acids showed some activity to all except proline. The relative specific activity for amino acid substrates was not correlated only with Km or kcat values, since the two parameters often varied independently of each other. Variation in Km was attributed to the differential binding affinity. Variation in kcat was attributed to differential positioning of the bound substrate relative to FAD that decreased the reaction rate. A structural model of this LAAO was compared with structures of other FAD-dependent LAAOs that have different substrate specificities: an LAAO from snake venom that prefers aromatic amino acid substrates and a fungal LAAO that is specific for lysine. While the amino acid sequences of these LAAOs are not very similar, their overall structures are comparable. The differential activity towards specific amino acids was correlated with specific residues in the active sites of these LAAOs. Residues in the active site that interact with the amino and carboxyl groups attached to the α-carbon of the substrate amino acid are conserved in all of the LAAOs. Residues that interact with the side chains of the amino acid substrates show variation. This provides insight into the structural determinants of the LAAOs that dictate their different substrate preferences. These results are of interest for harnessing these enzymes for possible applications in biotechnology, such as deracemization.
Collapse
Affiliation(s)
- Kyle J. Mamounis
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA; (K.J.M.); (M.L.C.N.)
| | - Maria Luiza Caldas Nogueira
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA; (K.J.M.); (M.L.C.N.)
| | - Daniela Priscila Marchi Salvador
- Department of Molecular Biology, Center of Exact and Natural Sciences, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil;
| | - Andres Andreo-Vidal
- Department of Genetics and Microbiology, University of Murcia, 30100 Murcia, Spain; (A.A.-V.); (A.S.-A.)
| | - Antonio Sanchez-Amat
- Department of Genetics and Microbiology, University of Murcia, 30100 Murcia, Spain; (A.A.-V.); (A.S.-A.)
| | - Victor L. Davidson
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA; (K.J.M.); (M.L.C.N.)
- Correspondence: ; Tel.: +1-407-266-7111; Fax: +1-407-266-7002
| |
Collapse
|
19
|
Rosini E, Pollegioni L. Reactive oxygen species as a double-edged sword: The role of oxidative enzymes in antitumor therapy. Biofactors 2022; 48:384-399. [PMID: 34608689 DOI: 10.1002/biof.1789] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 09/20/2021] [Indexed: 12/18/2022]
Abstract
A number of approaches have been developed over the years to manage cancer, such as chemotherapy using low-molecular-mass molecules and radiotherapy. Here, enzymes can also find useful applications. Among them, oxidases have attracted attention because of their ability to produce reactive oxygen species (ROS, especially hydrogen peroxide) in tumors and potentially modulate the production of this cytotoxic compound when enzymes active on substrates present in low amounts are used, such as the d-amino acid oxidase and d-amino acid couple system. These treatments have been also developed for additional cancer treatment approaches, such as phototherapy, nutrient starvation, and metal-induced hydroxyl radical production. In addition, to improve tumor specificity and decrease undesired side effects, oxidases have been targeted by means of nanotechnologies and protein engineering (i.e., by designing chimeric proteins able to accumulate in the tumor). The most recent advances obtained by using six different oxidases (i.e., the FAD-containing enzymes glucose oxidase, d- and l-amino acid oxidases, cholesterol oxidase and xanthine oxidase, and the copper-containing amine oxidase) have been reported. Anticancer therapy based on oxidase-based ROS production has now reached maturity and can be applied in the clinic.
Collapse
Affiliation(s)
- Elena Rosini
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
20
|
Kitani Y, Osaka Y, Ishizaki S. Seawater activates l-amino acid oxidase from the serum of the red-spotted grouper Epinephelusakaara. FISH & SHELLFISH IMMUNOLOGY 2022; 120:222-232. [PMID: 34838986 DOI: 10.1016/j.fsi.2021.11.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/15/2021] [Accepted: 11/23/2021] [Indexed: 06/13/2023]
Abstract
l-amino acid oxidases (LAOs) catalyze the oxidative deamination of l-amino acid and generate α-keto acid, ammonia, and hydrogen peroxide as byproducts. LAOs showed the variety of bioactivity by the resulting hydrogen peroxide. The serum of the red-spotted grouper Epinephelus akaara contains an LAO (Ea-LAO) with the potential to kill bacterial pathogens Aeromonas salmonicida and Vibrio anguillarum via hydrogen peroxide. However, it is unknown how the grouper tolerates the harmful effects of the serum Ea-LAO byproducts. In this study, we analyzed the kinetics of fish LAOs to understand how they escape the toxicity of byproducts. The LAO activity of grouper serum was suppressed in low-salt solutions such as NaCl, CaCl2, MgCl2, and diluted seawater. The activity was non-linearly increased and fitted to the four-parameter log-logistic model. The EC50 of the seawater was calculated to have a 0.72-fold concentration. This result suggested that the Ea-LAO could be activated by mixing with seawater. The results of circular dichroism spectroscopy showed that the α helix content was estimated to be 12.1% and 5.3% in a salt-free buffer (inactive condition) and the original concentration of seawater (active condition), respectively, indicating that the secondary structure of the Ea-LAO in the active condition was randomized. In addition, the Ea-LAO showed reversible LAO activity regulation according to the salt concentration in the environment. Taken together, this indicates that the Ea-LAO is normally on standby as an inactive form, and it could activate as a host-defense molecule to avoid pathogen invasion via a wound when mixed with seawater.
Collapse
Affiliation(s)
- Yoichiro Kitani
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ogi Mu 4-1, Noto-Cho, Ishikawa, 927-0553, Japan.
| | - Yuto Osaka
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ogi Mu 4-1, Noto-Cho, Ishikawa, 927-0553, Japan
| | - Shoichiro Ishizaki
- Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, Konan 4-5-7, Minato, Tokyo, 108-8477, Japan
| |
Collapse
|
21
|
Abstract
L-Amino acid oxidase (LAAO) is a flavin adenine dinucleotide (FAD)-dependent enzyme active on most proteinogenic L-amino acids, catalysing their conversion to α-keto acids by oxidative deamination of the substrate. For this oxidation reaction, molecular oxygen is used as the electron acceptor, generating hydrogen peroxide. LAAO can be used to detect L-amino acids, for the production of hydrogen peroxide as an oxidative agent or antimicrobial agent, and for the production of enantiopure amino acids from racemates. In this work, we characterised a previously reported LAAO from the bacterium Pseudoalteromonas luteoviolacea. The substrate scope and kinetic properties of the enzyme were determined, and the thermostability was evaluated. Additionally, we elucidated the crystal structure of this bacterial LAAO, enabling us to test the role of active site residues concerning their function in catalysis. The obtained insights and ease of expression of this thermostable LAAO provides a solid basis for the development of engineered LAAO variants tuned for biosensing and/or biocatalysis.
Collapse
|
22
|
Yildiz I. Computational Analysis of the Nicotine Oxidoreductase Mechanism by the ONIOM Method. ACS OMEGA 2021; 6:22422-22428. [PMID: 34497931 PMCID: PMC8412962 DOI: 10.1021/acsomega.1c03357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
Nicotine oxidoreductase (NicA2) is a monoamine oxidase (MAO)-based flavoenzyme that catalyzes the oxidation of S-nicotine into N-methylmyosmine. Due to its nanomolar binding affinity toward nicotine, it is seen as an ideal candidate for the treatment of nicotine addiction. Based on the crystal structure of the substrate-bound enzyme, hydrophobic interactions mainly govern the binding of the substrate in the active site through Trp108, Trp364, Trp427, and Leu217 residues. In addition, Tyr308 forms H-bonding with the pyridyl nitrogen of the substrate. Experimental and computational studies support the hydride transfer mechanism for MAO-based enzymes. In this mechanism, a hydride ion transfers from the substrate to the flavin cofactor. In this study, computational models involving the ONIOM method were formulated to study the hydride transfer mechanism based on the crystal structure of the enzyme-substrate complex. The geometry and energetics of the hydride transfer mechanism were analyzed, and the roles of active site residues were highlighted.
Collapse
Affiliation(s)
- Ibrahim Yildiz
- Chemistry Department, Khalifa
University, P.O. Box 127788 Abu Dhabi, United Arab Emirates
| |
Collapse
|
23
|
Heß MC, Grollius M, Duhay V, Koopmeiners S, Bloess S, Fischer von Mollard G. Analysis of N-glycosylation in fungal l-amino acid oxidases expressed in the methylotrophic yeast Pichia pastoris. Microbiologyopen 2021; 10:e1224. [PMID: 34459552 PMCID: PMC8364938 DOI: 10.1002/mbo3.1224] [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: 04/14/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 11/11/2022] Open
Abstract
l-amino acid oxidases (LAAOs) catalyze the oxidative deamination of l-amino acids to corresponding α-keto acids. Here, we describe the heterologous expression of four fungal LAAOs in Pichia pastoris. cgLAAO1 from Colletotrichum gloeosporioides and ncLAAO1 from Neurospora crassa were able to convert substrates not recognized by recombinant 9His-hcLAAO4 from the fungus Hebeloma cylindrosporum described earlier thereby broadening the substrate spectrum for potential applications. 9His-frLAAO1 from Fibroporia radiculosa and 9His-laLAAO2 from Laccaria amethystine were obtained only in low amounts. All four enzymes were N-glycosylated. We generated mutants of 9His-hcLAAO4 lacking N-glycosylation sites to further understand the effects of N-glycosylation. All four predicted N-glycosylation sites were glycosylated in 9His-hcLAAO4 expressed in P. pastoris. Enzymatic activity was similar for fully glycosylated 9His-hcLAAO4 and variants without one or all N-glycosylation sites after acid activation of all samples. However, activity without acid treatment was low in a variant without N-glycans. This was caused by the absence of a hypermannosylated N-glycan on asparagine residue N54. The lack of one or all of the other N-glycans was without effect. Our results demonstrate that adoption of a more active conformation requires a specific N-glycosylation during biosynthesis.
Collapse
Affiliation(s)
- Marc Christian Heß
- Biochemistry IIIDepartment of ChemistryBielefeld UniversityBielefeldGermany
| | - Marvin Grollius
- Biochemistry IIIDepartment of ChemistryBielefeld UniversityBielefeldGermany
| | - Valentin Duhay
- Biochemistry IIIDepartment of ChemistryBielefeld UniversityBielefeldGermany
| | - Simon Koopmeiners
- Biochemistry IIIDepartment of ChemistryBielefeld UniversityBielefeldGermany
| | - Svenja Bloess
- Biochemistry IIIDepartment of ChemistryBielefeld UniversityBielefeldGermany
| | | |
Collapse
|
24
|
Bothrops Jararaca Snake Venom Modulates Key Cancer-Related Proteins in Breast Tumor Cell Lines. Toxins (Basel) 2021; 13:toxins13080519. [PMID: 34437390 PMCID: PMC8402457 DOI: 10.3390/toxins13080519] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer is characterized by the development of abnormal cells that divide in an uncontrolled way and may spread into other tissues where they may infiltrate and destroy normal body tissue. Several previous reports have described biochemical anti-tumorigenic properties of crude snake venom or its components, including their capability of inhibiting cell proliferation and promoting cell death. However, to the best of our knowledge, there is no work describing cancer cell proteomic changes following treatment with snake venoms. In this work we describe the quantitative changes in proteomics of MCF7 and MDA-MB-231 breast tumor cell lines following treatment with Bothrops jararaca snake venom, as well as the functional implications of the proteomic changes. Cell lines were treated with sub-toxic doses at either 0.63 μg/mL (low) or 2.5 μg/mL (high) of B. jararaca venom for 24 h, conditions that cause no cell death per se. Proteomics analysis was conducted on a nano-scale liquid chromatography coupled on-line with mass spectrometry (nLC-MS/MS). More than 1000 proteins were identified and evaluated from each cell line treated with either the low or high dose of the snake venom. Protein profiling upon venom treatment showed differential expression of several proteins related to cancer cell metabolism, immune response, and inflammation. Among the identified proteins we highlight histone H3, SNX3, HEL-S-156an, MTCH2, RPS, MCC2, IGF2BP1, and GSTM3. These data suggest that sub-toxic doses of B. jararaca venom have potential to modulate cancer-development related protein targets in cancer cells. This work illustrates a novel biochemical strategy to identify therapeutic targets against cancer cell growth and survival.
Collapse
|
25
|
Leu JH, Tsai CH, Yang CH, Chou HY, Wang HC. Identification and characterization of l-amino acid oxidase 2 gene in orange-spotted grouper (Epinephelus coioides). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 120:104058. [PMID: 33657430 DOI: 10.1016/j.dci.2021.104058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 06/12/2023]
Abstract
Recently, l-amino acid oxidases (LAAOs) have been identified in several fish species as first-line defense molecules against bacterial infection. Here, we report the cloning and characterization of a fish LAAO gene, EcLAAO2, from orange-spotted grouper (Epinephelus coioides). The full-length cDNA is 3030 bp, with an ORF encoding a protein of 511 amino acids. EcLAAO2 is mainly expressed in the fin, gill, and intestine. Its expression is upregulated in several immune organs after challenge with lipopolysaccharide (LPS) and poly (I:C). The recombinant EcLAAO2 protein (rEcLAAO2), expressed and purified from a baculovirus expression system, was determined to be a glycosylated dimer. According to a hydrogen peroxide-production assay, the recombinant protein was identified as having LAAO enzyme activity with substrate preference for L-Phe and L-Trp, but not L-Lys as other known fish LAAOs. rEcLAAO2 could effectively inhibit the growth of Vibrio parahaemolyticus, Staphylococcus aureus, and Bacillus subtilis while exhibiting less effective inhibition of the growth of Escherichia coli. Finally, protein models based on sequence homology were constructed to predict the three-dimensional structure of EcLAAO2 as well as to explain the difference in substrate specificity between EcLAAO2 and other reported fish LAAOs. In conclusion, this study identifies EcLAAO2 as a novel fish LAAO with a substrate preference distinct from other known fish LAAOs and reveals that it may function against invading pathogens.
Collapse
Affiliation(s)
- Jiann-Horng Leu
- Institute of Marine Biology, National Taiwan Ocean University, Keelung, Taiwan, ROC; Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, Taiwan, ROC.
| | - Chi-Hang Tsai
- Institute of Marine Biology, National Taiwan Ocean University, Keelung, Taiwan, ROC
| | - Chia-Hsun Yang
- Institute of Marine Biology, National Taiwan Ocean University, Keelung, Taiwan, ROC
| | - Hsin-Yiu Chou
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, Taiwan, ROC; Department of Aquaculture, National Taiwan Ocean University, Keelung, Taiwan, ROC
| | - Hao-Ching Wang
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 115, Taiwan, ROC; Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan, ROC; International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan, Taiwan, ROC.
| |
Collapse
|
26
|
Zeitler L, Fiore A, Meyer C, Russier M, Zanella G, Suppmann S, Gargaro M, Sidhu SS, Seshagiri S, Ohnmacht C, Köcher T, Fallarino F, Linkermann A, Murray PJ. Anti-ferroptotic mechanism of IL4i1-mediated amino acid metabolism. eLife 2021; 10:64806. [PMID: 33646117 PMCID: PMC7946422 DOI: 10.7554/elife.64806] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Interleukin-4-induced-1 (IL4i1) is an amino acid oxidase secreted from immune cells. Recent observations have suggested that IL4i1 is pro-tumorigenic via unknown mechanisms. As IL4i1 has homologs in snake venoms (L-amino acid oxidases [LAAO]), we used comparative approaches to gain insight into the mechanistic basis of how conserved amino acid oxidases regulate cell fate and function. Using mammalian expressed recombinant proteins, we found that venom LAAO kills cells via hydrogen peroxide generation. By contrast, mammalian IL4i1 is non-cytotoxic and instead elicits a cell protective gene expression program inhibiting ferroptotic redox death by generating indole-3-pyruvate (I3P) from tryptophan. I3P suppresses ferroptosis by direct free radical scavenging and through the activation of an anti-oxidative gene expression program. Thus, the pro-tumor effects of IL4i1 are likely mediated by local anti-ferroptotic pathways via aromatic amino acid metabolism, arguing that an IL4i1 inhibitor may modulate tumor cell death pathways.
Collapse
Affiliation(s)
- Leonie Zeitler
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Claudia Meyer
- Universitätsklinikum Carl Gustav Carus Dresden, Dresden, Germany
| | - Marion Russier
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Gaia Zanella
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | | | - Sachdev S Sidhu
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | | | - Caspar Ohnmacht
- Helmholtz Zentrum München Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Thomas Köcher
- Vienna BioCenter Core Facilities GmbH, Vienna, Austria
| | | | | | - Peter J Murray
- Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
27
|
Cytotoxicity of snake venom enzymatic toxins: phospholipase A2 and l-amino acid oxidase. Biochem Soc Trans 2021; 48:719-731. [PMID: 32267491 PMCID: PMC7200639 DOI: 10.1042/bst20200110] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/07/2020] [Accepted: 03/16/2020] [Indexed: 12/23/2022]
Abstract
The phospholipase A2 (PLA2) and l-amino acid oxidase (LAAO) are two major enzymes found in the venoms from most snake species. These enzymes have been structurally and functionally characterised for their pharmacological activities. Both PLA2 and LAAO from different venoms demonstrate considerable cytotoxic effects on cancer cells via induction of apoptosis, cell cycle arrest and suppression of proliferation. These enzymes produce more pronounced cytotoxic effects in cancer cells than normal cells, thus they can be potential sources as chemotherapeutic agents. It is proposed that PLA2 and LAAO contribute to an elevated oxidative stress due to their catalytic actions, for instance, the ability of PLA2 to produce reactive oxygen species during lipolysis and formation of H2O2 from LAAO catalytic activity which consequently lead to cell death. Nonetheless, the cell-death signalling pathways associated with exposure to these enzymatic toxins are not fully elucidated yet. Here in this review, we will discuss the cytotoxic effects of PLA2 and LAAO in relationship to their catalytic mechanisms and the underlying mechanisms of cytotoxic actions.
Collapse
|
28
|
Yildiz I, Yildiz BS. Mechanistic study of L-6-hydroxynicotine oxidase by DFT and ONIOM methods. J Mol Model 2021; 27:53. [PMID: 33507404 DOI: 10.1007/s00894-020-04646-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/10/2020] [Indexed: 10/22/2022]
Abstract
L-6-Hydroxynicotine oxidase (LHNO) is a member of monoamine oxidase (MAO) family and catalyzes conversion of (S)-6-hydroxynicotine to 6-hydroxypseudooxynicotine during bacterial degradation of nicotine. Recent studies indicated that the enzyme catalyzes oxidation of carbon-nitrogen bond instead of previously proposed carbon-carbon bond. Based on kinetics and mutagenesis studies, Asn166, Tyr311, and Lys287 as well as an active site water molecule have roles in the catalysis of the enzyme. A number of studies including experimental and computational methods support hydride transfer mechanism in MAO family as a common mechanism in which a hydride ion transfer from amine substrate to flavin cofactor is the rate-limiting step. In this study, we formulated computational models to study the hydride transfer mechanism using crystal structure of enzyme-substrate complex. The calculations involved ONIOM and DFT methods, and we evaluated the geometry and energetics of the hydride transfer process while probing the roles of active site residues. Based on the calculations involving hydride, radical, and polar mechanisms, it was concluded that hydride transfer mechanism is the only viable mechanism for LHNO.
Collapse
Affiliation(s)
- Ibrahim Yildiz
- Chemistry Department, Khalifa University, PO Box 127788, Abu Dhabi, United Arab Emirates.
| | - Banu Sizirici Yildiz
- CIVE Department, Khalifa University, PO Box 127788, Abu Dhabi, United Arab Emirates
| |
Collapse
|
29
|
Tararina MA, Dam KK, Dhingra M, Janda KD, Palfey BA, Allen KN. Fast Kinetics Reveals Rate-Limiting Oxidation and the Role of the Aromatic Cage in the Mechanism of the Nicotine-Degrading Enzyme NicA2. Biochemistry 2021; 60:259-273. [PMID: 33464876 DOI: 10.1021/acs.biochem.0c00855] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In Pseudomonas putida, the flavoprotein nicotine oxidoreductase (NicA2) catalyzes the oxidation of (S)-nicotine to N-methyl-myosmine, which is nonenzymatically hydrolyzed to pseudooxynicotine. Structural analysis reveals a monoamine oxidase (MAO)-like fold with a conserved FAD-binding domain and variable substrate-binding domain. The flavoenzyme has a unique variation of the classic aromatic cage with flanking residue pair W427/N462. Previous mechanistic studies using O2 as the oxidizing substrate show that NicA2 has a low apparent Km of 114 nM for (S)-nicotine with a very low apparent turnover number (kcat of 0.006 s-1). Herein, the mechanism of NicA2 was analyzed by transient kinetics. Single-site variants of W427 and N462 were used to probe the roles of these residues. Although several variants had moderately higher oxidase activity (7-12-fold), their reductive half-reactions using (S)-nicotine were generally significantly slower than that of wild-type NicA2. Notably, the reductive half-reaction of wild-type NicA2 is 5 orders of magnitude faster than the oxidative half-reaction with an apparent pseudo-first-order rate constant for the reaction of oxygen similar to kcat. X-ray crystal structures of the N462V and N462Y/W427Y variants complexed with (S)-nicotine (at 2.7 and 2.3 Å resolution, respectively) revealed no significant active-site rearrangements. A second substrate-binding site was identified in N462Y/W427Y, consistent with observed substrate inhibition. Together, these findings elucidate the mechanism of a flavoenzyme that preferentially oxidizes tertiary amines with an efficient reductive half-reaction and a very slow oxidative half-reaction when O2 is the oxidizing substrate, suggesting that the true oxidizing agent is unknown.
Collapse
Affiliation(s)
- Margarita A Tararina
- Program in Biomolecular Pharmacology, Boston University School of Medicine, 72 East Concord Street, Boston, Massachusetts 02118, United States
| | - Katie K Dam
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - Manaswni Dhingra
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | | | - Bruce A Palfey
- Department of Biological Chemistry, University of Michigan, 5220E MSRB III 1150 West Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Karen N Allen
- Program in Biomolecular Pharmacology, Boston University School of Medicine, 72 East Concord Street, Boston, Massachusetts 02118, United States.,Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| |
Collapse
|
30
|
Osaka Y, Kitani Y. Blood loss induces l-amino acid oxidase gene expression in the head kidney of the red-spotted grouper, Epinephelus akaara. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 114:103842. [PMID: 32866581 DOI: 10.1016/j.dci.2020.103842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/03/2020] [Accepted: 08/24/2020] [Indexed: 06/11/2023]
Abstract
In fish, the innate immune system is more important than the adaptive immune system because it responds quickly and nonspecifically to protect against pathogens. Thus, a variety of innate immune molecules have been found in fish. Recently, l-amino acid oxidases (LAOs) were discovered as a new member of the antibacterial protein from fish skin mucus and serum. In this study, we newly found an antibacterial LAO in red-spotted grouper (Epinephelus akaara) serum. It showed a broad range of substrate specificity with aromatic and hydrophobic amino acids. The grouper LAO gene had a low expression level in the kidney under normal conditions; however, it was significantly upregulated by blood loss 1 day after bleeding. In addition, the LAO activity in the serum recovered within 3 days in the same experiment. This quick recovery may indicate that the LAO is an essential innate immune molecule in the whole grouper body.
Collapse
Affiliation(s)
- Yuto Osaka
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ogi Mu 4-1 Noto-Cho, Ishikawa, 927-0553, Japan
| | - Yoichiro Kitani
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ogi Mu 4-1 Noto-Cho, Ishikawa, 927-0553, Japan.
| |
Collapse
|
31
|
Pająk M. Methylated derivatives of l-tyrosine in reaction catalyzed by l-amino acid oxidase: isotope and inhibitory effects. J Biochem 2020; 168:509-514. [PMID: 32569353 DOI: 10.1093/jb/mvaa066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/10/2020] [Indexed: 11/14/2022] Open
Abstract
l-Amino acid oxidase (LAAO) is widely distributed in nature and shows important biological activity. It induces cell apoptosis and has antibacterial properties. This study was designed to investigate the effect of methyl substituent on its activity as methylated derivatives of l-tyrosine, labelled with short-lived B+ emitters, have been used in oncological diagnostics. To study isotope effects in the oxidative deamination of O-methyl-l-tyrosine, the deuterated isotopomer, i.e. O-methyl-[2-2H]-l-tyrosine, was synthesized by isotope exchange, catalyzed enzymatically by tryptophanase. Isotope effects were determined using the spectrophotometric non-competitive method. The values of isotope effects indicate that the α-C-H bond cleavage occurs in the rate determining step of the investigated reaction and α-hydrogen plays a role in the substrate binding process at the enzyme active site. The inhibitory effect on LAAO activity was studied with α-methyl-l-tyrosine and N-methyl-l-tyrosine. The mode of inhibition was determined based on Lineweavear-Burk plots intersections. α-Methyl-l-tyrosine has been found a mixed type inhibitor of the investigated enzyme, whereas N-methyl-l-tyrosine is a non-competitive inhibitor of LAAO.
Collapse
Affiliation(s)
- Małgorzata Pająk
- Faculty of Chemistry, University of Warsaw, Pasteur 1 Str, 02-093 Warsaw, Poland
| |
Collapse
|
32
|
Kitani Y, Nagashima Y. l-Amino acid oxidase as a fish host-defense molecule. FISH & SHELLFISH IMMUNOLOGY 2020; 106:685-690. [PMID: 32822860 DOI: 10.1016/j.fsi.2020.08.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 06/11/2023]
Abstract
An l-amino acid oxidase (LAO) is an amino acid metabolism enzyme that also performs a variety of biological activities. Recently, LAOs have been discovered to be deeply involved in innate immunity in fish because of their antibacterial and antiparasitic activity. The determinant of potent antibacterial/antiparasitic activity is the H2O2 byproduct of LAO enzymatic activity that utilizes the l-amino acid as a substrate. In addition, fish LAOs are upregulated by pathogenic bacteria or parasite infection. Furthermore, some fish LAOs show that the target specificity depends on the virulence of the bacteria. All results reflect that LAOs are new innate immune molecules. This review also describes the potential of the immunomodulatory functions of fish LAOs, not only the innate immune function by a direct oxidation attack of H2O2.
Collapse
Affiliation(s)
- Yoichiro Kitani
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ogi Mu 4-1 Noto-Cho, Ishikawa, 927-0553, Japan.
| | - Yuji Nagashima
- Department of Agro-Food Science, Niigata Agro-Food University, Hirakidai 2416, Tainai, Niigata, 995-2702, Japan
| |
Collapse
|
33
|
Kondo H, Kitagawa M, Matsumoto Y, Saito M, Amano M, Sugiyama S, Tamura T, Kusakabe H, Inagaki K, Imada K. Structural basis of strict substrate recognition of l-lysine α-oxidase from Trichoderma viride. Protein Sci 2020; 29:2213-2225. [PMID: 32894626 DOI: 10.1002/pro.3946] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 11/07/2022]
Abstract
l-Lysine oxidase (LysOX) is a FAD-dependent homodimeric enzyme that catalyzes the oxidative deamination of l-lysine to produce α-keto-ε-aminocaproate with ammonia and hydrogen peroxide. LysOX shows strict substrate specificity for l-lysine, whereas most l-amino acid oxidases (LAAOs) exhibit broad substrate specificity for l-amino acids. Previous studies of LysOX showed that overall structural similarity to the well-studied snake venom LAAOs. However, the molecular mechanism of strict specificity for l-lysine was still unclear. We here determined the structure of LysOX in complex with l-lysine at 1.7 Å resolution. The structure revealed that the hydrogen bonding network formed by D212, D315, and A440 with two water molecules is responsible for the recognition of the side chain amino group. In addition, a narrow hole formed by five hydrophobic residues in the active site contributes to strict substrate specificity. Mutation studies demonstrated that D212 and D315 are essential for l-lysine recognition, and the D212A/D315A double mutant LysOX showed different substrate specificity from LysOX. Moreover, the structural basis of the substrate specificity change has also been revealed by the structural analysis of the mutant variant and its substrate complexes. These results clearly explain the molecular mechanism of the strict specificity of LysOX and suggest that LysOX is a potential candidate for a template to design LAAOs specific to other l-amino acids.
Collapse
Affiliation(s)
- Hiroki Kondo
- Department of Macromolecular Science, Graduate School of Science, Osaka University, Osaka, Japan
| | - Masaki Kitagawa
- Department of Macromolecular Science, Graduate School of Science, Osaka University, Osaka, Japan
| | - Yuya Matsumoto
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Masaya Saito
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Marie Amano
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Shigeru Sugiyama
- Faculty of Science and Technology, Kochi University, Kochi, Japan
| | - Takashi Tamura
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | | | - Kenji Inagaki
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Katsumi Imada
- Department of Macromolecular Science, Graduate School of Science, Osaka University, Osaka, Japan
| |
Collapse
|
34
|
Molecular Deceleration Regulates Toxicant Release to Prevent Cell Damage in Pseudomonas putida S16 (DSM 28022). mBio 2020; 11:mBio.02012-20. [PMID: 32873764 PMCID: PMC7468206 DOI: 10.1128/mbio.02012-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The underlying molecular mechanisms of flavin-dependent amine oxidases remain relatively poorly understood, even though many of these enzymes have been reported. The nicotine oxidoreductase NicA2 is a crucial enzyme for the first step of nicotine degradation in Pseudomonas putida S16 (DSM 28022). Here, we present the crystal structure of a ternary complex comprising NicA2 residues 21 to 482, flavin adenine dinucleotide (FAD), and nicotine at 2.25 Å resolution. Unlike other, related structures, NicA2 does not have an associated diacyl glycerophospholipid, wraps its substrate more tightly, and has an intriguing exit passage in which nine bulky amino acid residues occlude the release of its toxic product, pseudooxynicotine (PN). The replacement of these bulky residues by amino acids with small side chains effectively increases the catalytic turnover rate of NicA2. Our results indicate that the passage in wild-type NicA2 effectively controls the rate of PN release and thus prevents its rapid intracellular accumulation. It gives ample time for PN to be converted to less-harmful substances by downstream enzymes such as pseudooxynicotine amine oxidase (Pnao) before its accumulation causes cell damage or even death. The temporal metabolic regulation mode revealed in this study may shed light on the production of cytotoxic compounds.IMPORTANCE Flavin-dependent amine oxidases have received extensive attention because of their importance in drug metabolism, Parkinson's disease, and neurotransmitter catabolism. However, the underlying molecular mechanisms remain relatively poorly understood. Here, combining the crystal structure of NicA2 (an enzyme in the first step of the bacterial nicotine degradation pathway in Pseudomonas putida S16 (DSM 28022)), biochemical analysis, and mutant construction, we found an intriguing exit passage in which bulky amino acid residues occlude the release of the toxic product of NicA2, in contrast to other, related structures. The selective product exportation register for NicA2 has proven to be beneficial to cell growth. Those seeking to produce cytotoxic compounds could greatly benefit from the use of such an export register mechanism.
Collapse
|
35
|
Yasukawa K, Kawahara N, Motojima F, Nakano S, Asano Y. Porcine kidney d-amino acid oxidase-derived R-amine oxidases with new substrate specificities. Enzymes 2020; 47:117-136. [PMID: 32951821 DOI: 10.1016/bs.enz.2020.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An R-stereoselective amine oxidase and variants with markedly altered substrate specificity toward (R)-amines were generated from porcine d-amino acid oxidase (pkDAO), based on the X-ray crystallographic analysis of the wild-type enzyme. The new R-amine oxidase, a pkDAO variant (Y228L/R283G), acted on α-MBA and its derivatives, α-ethylbenzylamine, alkylamine, and cyclic secondary amines, totally losing the activities toward the original substrates, d-amino acids. The variant is enantiocomplementary to the flavin-type S-stereoselective amine oxidase variant from Aspergillus niger. Moreover, we solved the structure of pkDAO variants and successfully applied the obtained information to generate more variants through rational protein engineering, and used them in the synthesis of pharmaceutically attractive chiral compounds. The pkDAO variant Y228L/R283G and a variant I230A/R283G were used to synthesize (S)-amine and (R)-4-CBHA through deracemization, from racemic α-methylbenzylamine and benzhydrylamine, respectively, by selective oxidation of one of the enantiomers in the presence of a chemical reductant such as NaBH4. From a mechanistic point of view, we speculated that the imine intermediate, synthesized by oxidases or dehydrogenases, could be converted into primary α-aminonitrile by nucleophilic addition of cyanide in aqueous solutions. Nitriles and some unnatural amino acids were synthesized through a cascade reaction by oxidative cyanation reaction with the variant and a wide substrate specificity nitrilase.
Collapse
Affiliation(s)
- Kazuyuki Yasukawa
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, Imizu, Toyama, Japan; Asano Active Enzyme Molecule Project, ERATO, JST, Imizu, Toyama, Japan
| | - Nobuhiro Kawahara
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, Imizu, Toyama, Japan; Asano Active Enzyme Molecule Project, ERATO, JST, Imizu, Toyama, Japan
| | - Fumihiro Motojima
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, Imizu, Toyama, Japan; Asano Active Enzyme Molecule Project, ERATO, JST, Imizu, Toyama, Japan
| | - Shogo Nakano
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, Imizu, Toyama, Japan; Asano Active Enzyme Molecule Project, ERATO, JST, Imizu, Toyama, Japan
| | - Yasuhisa Asano
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, Imizu, Toyama, Japan; Asano Active Enzyme Molecule Project, ERATO, JST, Imizu, Toyama, Japan.
| |
Collapse
|
36
|
Wu Y, Shi J, Mei S, Katimba HA, Sun Y, Wang X, Liang K, Jiang Z. Concerted Chemoenzymatic Synthesis of α-Keto Acid through Compartmentalizing and Channeling of Metal–Organic Frameworks. ACS Catal 2020. [DOI: 10.1021/acscatal.0c01985] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yizhou Wu
- Key Laboratory for Green Chemical Technology of Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, 92# Weijin Road, Nankai District, Tianjin, 300072, China
| | - Jiafu Shi
- School of Environmental Science and Engineering, Tianjin University, 92# Weijin Road, Nankai District, Tianjin, 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 10090, China
| | - Shuang Mei
- Key Laboratory for Green Chemical Technology of Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, 92# Weijin Road, Nankai District, Tianjin, 300072, China
| | - Hija Athman Katimba
- Key Laboratory for Green Chemical Technology of Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, 92# Weijin Road, Nankai District, Tianjin, 300072, China
| | - Yiying Sun
- Key Laboratory for Green Chemical Technology of Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, 92# Weijin Road, Nankai District, Tianjin, 300072, China
| | - Xueying Wang
- School of Environmental Science and Engineering, Tianjin University, 92# Weijin Road, Nankai District, Tianjin, 300072, China
| | - Kang Liang
- School of Chemical Engineering, Graduate School of Biomedical Engineering and Australian Centre for NanoMedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Zhongyi Jiang
- Key Laboratory for Green Chemical Technology of Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, 92# Weijin Road, Nankai District, Tianjin, 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, China
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou 350207, China
| |
Collapse
|
37
|
Idrees M, Mohammad AR, Karodia N, Rahman A. Multimodal Role of Amino Acids in Microbial Control and Drug Development. Antibiotics (Basel) 2020; 9:E330. [PMID: 32560458 PMCID: PMC7345125 DOI: 10.3390/antibiotics9060330] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 12/16/2022] Open
Abstract
Amino acids are ubiquitous vital biomolecules found in all kinds of living organisms including those in the microbial world. They are utilised as nutrients and control many biological functions in microorganisms such as cell division, cell wall formation, cell growth and metabolism, intermicrobial communication (quorum sensing), and microbial-host interactions. Amino acids in the form of enzymes also play a key role in enabling microbes to resist antimicrobial drugs. Antimicrobial resistance (AMR) and microbial biofilms are posing a great threat to the world's human and animal population and are of prime concern to scientists and medical professionals. Although amino acids play an important role in the development of microbial resistance, they also offer a solution to the very same problem i.e., amino acids have been used to develop antimicrobial peptides as they are highly effective and less prone to microbial resistance. Other important applications of amino acids include their role as anti-biofilm agents, drug excipients, drug solubility enhancers, and drug adjuvants. This review aims to explore the emerging paradigm of amino acids as potential therapeutic moieties.
Collapse
Affiliation(s)
- Muhammad Idrees
- Faculty of Science and Technology, University of Wolverhampton, Wolverhampton WV1 1LY, UK; (M.I.); (N.K.)
| | | | - Nazira Karodia
- Faculty of Science and Technology, University of Wolverhampton, Wolverhampton WV1 1LY, UK; (M.I.); (N.K.)
| | - Ayesha Rahman
- Faculty of Science and Technology, University of Wolverhampton, Wolverhampton WV1 1LY, UK; (M.I.); (N.K.)
| |
Collapse
|
38
|
Tararina MA, Allen KN. Bioinformatic Analysis of the Flavin-Dependent Amine Oxidase Superfamily: Adaptations for Substrate Specificity and Catalytic Diversity. J Mol Biol 2020; 432:3269-3288. [PMID: 32198115 DOI: 10.1016/j.jmb.2020.03.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/24/2020] [Accepted: 03/06/2020] [Indexed: 12/29/2022]
Abstract
The flavin-dependent amine oxidase (FAO) superfamily consists of over 9000 nonredundant sequences represented in all domains of life. Of the thousands of members identified, only 214 have been functionally annotated to date, and 40 unique structures are represented in the Protein Data Bank. The few functionally characterized members share a catalytic mechanism involving the oxidation of an amine substrate through transfer of a hydride to the FAD cofactor, with differences observed in substrate specificities. Previous studies have focused on comparing a subset of superfamily members. Here, we present a comprehensive analysis of the FAO superfamily based on reaction mechanism and substrate recognition. Using a dataset of 9192 sequences, a sequence similarity network, and subsequently, a genome neighborhood network were constructed, organizing the superfamily into eight subgroups that accord with substrate type. Likewise, through phylogenetic analysis, the evolutionary relationship of subgroups was determined, delineating the divergence between enzymes based on organism, substrate, and mechanism. In addition, using sequences and atomic coordinates of 22 structures from the Protein Data Bank to perform sequence and structural alignments, active-site elements were identified, showing divergence from the canonical aromatic-cage residues to accommodate large substrates. These specificity determinants are held in a structural framework comprising a core domain catalyzing the oxidation of amines with an auxiliary domain for substrate recognition. Overall, analysis of the FAO superfamily reveals a modular fold with cofactor and substrate-binding domains allowing for diversity of recognition via insertion/deletions. This flexibility allows facile evolution of new activities, as shown by reinvention of function between subfamilies.
Collapse
Affiliation(s)
- Margarita A Tararina
- Program in Biomolecular Pharmacology, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Karen N Allen
- Program in Biomolecular Pharmacology, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA; Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, MA 02215, USA.
| |
Collapse
|
39
|
Ullah A. Structure-Function Studies and Mechanism of Action of Snake Venom L-Amino Acid Oxidases. Front Pharmacol 2020; 11:110. [PMID: 32158389 PMCID: PMC7052187 DOI: 10.3389/fphar.2020.00110] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/28/2020] [Indexed: 12/30/2022] Open
Abstract
Snake venom L-amino acid oxidases (SV-LAAOs) are the least studied venom enzymes. These enzymes catalyze the stereospecific oxidation of an L-amino acid to their corresponding α-keto acid with the liberation of hydrogen peroxide (H2O2) and ammonia (NH3). They display various pathological and physiological activities including induction of apoptosis, edema, platelet aggregation/inhibition, hemorrhagic, and anticoagulant activities. They also show antibacterial, antiviral and leishmanicidal activity and have been used as therapeutic agents in some disease conditions like cancer and anti-HIV drugs. Although the crystal structures of six SV-LAAOs are present in the Protein Data Bank (PDB), there is no single article that describes all of them in particular. To better understand their structural properties and correlate it with their function, the current work describes structure characterization, structure-based mechanism of catalysis, inhibition and substrate specificity of SV-LAAOs. Sequence analysis indicates a high sequence identity (>84%) among SV-LAAOs, comparatively lower sequence identity with Pig kidney D-amino acid oxidase (<50%) and very low sequence identity (<24%) with bacterial LAAOs, Fugal (L-lysine oxidase), and Zea mays Polyamine oxidase (PAAO). The three-dimensional structure of these enzymes are composed of three-domains, a FAD-binding domain, a substrate-binding domain and a helical domain. The sequence and structural analysis indicate that the amino acid residues in the loops vary in length and composition due to which the surface charge distribution also varies that may impart variable substrate specificity to these enzymes. The active site cavity volume and its average depth also vary in these enzymes. The inhibition of these enzymes by synthetic inhibitors will lead to the production of more potent antivenoms against snakebite envenomation.
Collapse
Affiliation(s)
- Anwar Ullah
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
40
|
Molla G, Chaves‐Sanjuan A, Savinelli A, Nardini M, Pollegioni L. Structure and kinetic properties of humand‐aspartate oxidase, the enzyme‐controllingd‐aspartate levels in brain. FASEB J 2019; 34:1182-1197. [DOI: 10.1096/fj.201901703r] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/05/2019] [Accepted: 11/10/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Gianluca Molla
- Dipartimento di Biotecnologie e Scienze della Vita Università degli studi dell’Insubria Varese Italy
| | | | - Antonio Savinelli
- Dipartimento di Biotecnologie e Scienze della Vita Università degli studi dell’Insubria Varese Italy
| | - Marco Nardini
- Dipartimento di Bioscienze Università degli studi di Milano Milano Italy
| | - Loredano Pollegioni
- Dipartimento di Biotecnologie e Scienze della Vita Università degli studi dell’Insubria Varese Italy
| |
Collapse
|
41
|
Presset M, Djordjevic D, Dupont A, Le Gall E, Molinier-Frenkel V, Castellano F. Identification of inhibitors of the immunosuppressive enzyme IL4I1. Bioorg Chem 2019; 94:103463. [PMID: 31812258 DOI: 10.1016/j.bioorg.2019.103463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Marc Presset
- Electrochimie et Synthèse Organique, Université Paris Est, ICMPE (UMR 7182), CNRS, UPEC, 2 rue Henri Dunant, F-94320 Thiais, France
| | - Diana Djordjevic
- INSERM, U955, Equipe 09, Créteil, France; Université Paris Est, Faculté de Médecine, Créteil, France
| | - Aurélie Dupont
- INSERM, U955, Equipe 09, Créteil, France; Université Paris Est, Faculté de Médecine, Créteil, France
| | - Erwan Le Gall
- Electrochimie et Synthèse Organique, Université Paris Est, ICMPE (UMR 7182), CNRS, UPEC, 2 rue Henri Dunant, F-94320 Thiais, France
| | - Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09, Créteil, France; Université Paris Est, Faculté de Médecine, Créteil, France; AP-HP, Hôpital H. Mondor - A. Chenevier, Laboratoire d'Immunologie, Créteil, France.
| | - Flavia Castellano
- INSERM, U955, Equipe 09, Créteil, France; Université Paris Est, Faculté de Médecine, Créteil, France.
| |
Collapse
|
42
|
Zhang R, Zhou Y, Yan X, Fan K. Advances in chiral nanozymes: a review. Mikrochim Acta 2019; 186:782. [DOI: 10.1007/s00604-019-3922-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/12/2019] [Indexed: 02/08/2023]
|
43
|
Khedrinia M, Aryapour H, Mianabadi M. Prediction of novel inhibitors for Crotalus adamanteus l-amino acid oxidase by repurposing FDA-approved drugs: a virtual screening and molecular dynamics simulation investigation. Drug Chem Toxicol 2019; 44:470-479. [PMID: 31668098 DOI: 10.1080/01480545.2019.1614022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
One of the deadliest enzymes in the snake venom is l-amino acid oxidase (LAAO) which plays an important role in the pathophysiological effects during snake envenomation. Some effects of this enzyme on the human body are apoptosis, platelet aggregation, edema, hemorrhage, and cytotoxicity. Hence, inhibiting the enzyme activity to reduce its degradation effects is of great medical and pharmacological importance. On the other hand, drug repurposing is a process to find the new existing drug for a new medical indication. Since Crotalus adamanteus LAAO has no crystal structure in the protein data bank, first, its 3D structure was constructed by homology modeling using 1REO as the template and then modeled structure was evaluated by several algorithms. We screened the FDA-approved drugs by structure-based virtual screening, molecular dynamics (MD) simulation, and Molecular Mechanics Poisson Boltzmann Surface Area (MM/PBSA) to identify new inhibitors for the snake venom LAAO. Interestingly, docking results revealed that half of the hits belong to the propionic acid derivatives drugs. In addition, MD simulation was performed to assess the interaction profile of the docked protein-hits complexes. Meanwhile, Arg88, Gln112, Lys345, Trp356 form consistent hydrogen bond interactions with Dexketoprofen, Flurbiprofen, Ketoprofen, Morphine, and Citric acid during simulation. According to the results, each of the four compounds can be an appropriate inhibitor of LAAO and since our study was based on drug repurposing could be evaluated in phase II clinical trials.
Collapse
Affiliation(s)
- Mostafa Khedrinia
- Department of Biology, Faculty of Science, Golestan University, Gorgan, Iran
| | - Hassan Aryapour
- Department of Biology, Faculty of Science, Golestan University, Gorgan, Iran
| | - Manijeh Mianabadi
- Department of Biology, Faculty of Science, Golestan University, Gorgan, Iran
| |
Collapse
|
44
|
Wiezel GA, Rustiguel JK, Morgenstern D, Zoccal KF, Faccioli LH, Nonato MC, Ueberheide B, Arantes EC. Insights into the structure, function and stability of bordonein-L, the first L-amino acid oxidase from Crotalus durissus terrificus snake venom. Biochimie 2019; 163:33-49. [DOI: 10.1016/j.biochi.2019.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/06/2019] [Indexed: 01/18/2023]
|
45
|
Pokrovsky VS, Chepikova OE, Davydov DZ, Zamyatnin AA, Lukashev AN, Lukasheva EV. Amino Acid Degrading Enzymes and their Application in Cancer Therapy. Curr Med Chem 2019; 26:446-464. [PMID: 28990519 DOI: 10.2174/0929867324666171006132729] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 09/12/2017] [Accepted: 09/28/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Amino acids are essential components in various biochemical pathways. The deprivation of certain amino acids is an antimetabolite strategy for the treatment of amino acid-dependent cancers which exploits the compromised metabolism of malignant cells. Several studies have focused on the development and preclinical and clinical evaluation of amino acid degrading enzymes, namely L-asparaginase, L-methionine γ-lyase, L-arginine deiminase, L-lysine α-oxidase. Further research into cancer cell metabolism may therefore define possible targets for controlling tumor growth. OBJECTIVE The purpose of this review was to summarize recent progress in the relationship between amino acids metabolism and cancer therapy, with a particular focus on Lasparagine, L-methionine, L-arginine and L-lysine degrading enzymes and their formulations, which have been successfully used in the treatment of several types of cancer. METHODS We carried out a structured search among literature regarding to amino acid degrading enzymes. The main aspects of search were in vitro and in vivo studies, clinical trials concerning application of these enzymes in oncology. RESULTS Most published research are on the subject of L-asparaginase properties and it's use for cancer treatment. L-arginine deiminase has shown promising results in a phase II trial in advanced melanoma and hepatocellular carcinoma. Other enzymes, in particular Lmethionine γ-lyase and L-lysine α-oxidase, were effective in vitro and in vivo. CONCLUSION The findings of this review revealed that therapy based on amino acid depletion may have the potential application for cancer treatment but further clinical investigations are required to provide the efficacy and safety of these agents.
Collapse
Affiliation(s)
- Vadim S Pokrovsky
- Blokhin Cancer Research Center, Moscow, Russian Federation.,Orekhovich Institute of Biomedical Chemistry, Moscow, Russian Federation.,People's Friendship University, Russia (RUDN University), Moscow, Russian Federation
| | - Olga E Chepikova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | | | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.,Belozersky Institute of Physico- Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Alexander N Lukashev
- People's Friendship University, Russia (RUDN University), Moscow, Russian Federation.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Elena V Lukasheva
- People's Friendship University, Russia (RUDN University), Moscow, Russian Federation
| |
Collapse
|
46
|
Costa TR, Carone SEI, Tucci LFF, Menaldo DL, Rosa-Garzon NG, Cabral H, Sampaio SV. Kinetic investigations and stability studies of two Bothrops L-amino acid oxidases. J Venom Anim Toxins Incl Trop Dis 2018; 24:37. [PMID: 30534149 PMCID: PMC6280375 DOI: 10.1186/s40409-018-0172-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/07/2018] [Indexed: 01/29/2023] Open
Abstract
Background L-amino acid oxidases isolated from snake venoms (SV-LAAOs) are enzymes that have great therapeutic potential and are currently being investigated as tools for developing new strategies to treat various diseases, including cancer and bacterial infections. The main objective of this study was to make a brief evaluation of the enzymatic stability of two Bothrops LAAOs, one isolated from Bothrops jararacussu (BjussuLAAO-II) and the other from Bothrops moojeni (BmooLAAO-I) venoms. Methods and results The enzymatic activity and stability of both LAAOs were evaluated by microplate colorimetric assays, for which BjussuLAAO-II and BmooLAAO-I were incubated with different L-amino acid substrates, in the presence of different ions, and at different pH ranges and temperatures. BjussuLAAO-II and BmooLAAO-I demonstrated higher affinity for hydrophobic amino acids, such as Phe and Leu. The two enzymes showed high enzymatic activity in a wide temperature range, from 25 to 75 °C, and presented optimum pH around 7.0. Additionally, Zn2+, Al3+, Cu2+ and Ni2+ ions negatively modulated the enzymatic activity of both LAAOs. As to stability, BjussuLAAO-II and BmooLAAO-I showed high enzymatic activity for 42 days stored at 4 °C in neutral pH solution. Moreover, the glycan portions of both LAAOs were analyzed by capillary electrophoresis, which revealed that BjussuLAAO-II presented two main glycan portions with relative masses of 7.78 and 8.13 CGU, while BmooLAAO-I showed three portions of 7.58, 7.94 and 8.37 CGU. Conclusions Our results showed that, when stored properly, BjussuLAAO-II and BmooLAAO-I present enzymatic stability over a long time period, which is very important to allow the use of these enzymes in pharmacological studies of great impact in the medical field.
Collapse
Affiliation(s)
- Tássia R Costa
- 1Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (FCFRP-USP), Avenida do Café, s/n, B. Monte Alegre, Ribeirão Preto, SP CEP 14040-903 Brazil
| | - Sante E I Carone
- 1Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (FCFRP-USP), Avenida do Café, s/n, B. Monte Alegre, Ribeirão Preto, SP CEP 14040-903 Brazil
| | - Luiz F F Tucci
- 1Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (FCFRP-USP), Avenida do Café, s/n, B. Monte Alegre, Ribeirão Preto, SP CEP 14040-903 Brazil
| | - Danilo L Menaldo
- 1Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (FCFRP-USP), Avenida do Café, s/n, B. Monte Alegre, Ribeirão Preto, SP CEP 14040-903 Brazil
| | - Nathalia G Rosa-Garzon
- 2Departamento de Ciências Farmacêuticas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (FCFRP-USP), Ribeirão Preto, SP Brazil
| | - Hamilton Cabral
- 2Departamento de Ciências Farmacêuticas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (FCFRP-USP), Ribeirão Preto, SP Brazil
| | - Suely V Sampaio
- 1Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (FCFRP-USP), Avenida do Café, s/n, B. Monte Alegre, Ribeirão Preto, SP CEP 14040-903 Brazil
| |
Collapse
|
47
|
Estevão-Costa MI, Sanz-Soler R, Johanningmeier B, Eble JA. Snake venom components in medicine: From the symbolic rod of Asclepius to tangible medical research and application. Int J Biochem Cell Biol 2018; 104:94-113. [PMID: 30261311 DOI: 10.1016/j.biocel.2018.09.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/03/2018] [Accepted: 09/19/2018] [Indexed: 12/21/2022]
Abstract
Both mythologically and logically, snakes have always fascinated man. Snakes have attracted both awe and fear not only because of the elegant movement of their limbless bodies, but also because of the potency of their deadly venoms. Practically, in 2017, the world health organization (WHO) listed snake envenomation as a high priority neglected disease, as snakes inflict up to 2.7 million poisonous bites, around 100.000 casualties, and about three times as many invalidities on man. The venoms of poisonous snakes are a cocktail of potent compounds which specifically and avidly target numerous essential molecules with high efficacy. The individual effects of all venom toxins integrate into lethal dysfunctions of almost any organ system. It is this efficacy and specificity of each venom component, which after analysis of its structure and activity may serve as a potential lead structure for chemical imitation. Such toxin mimetics may help in influencing a specific body function pharmaceutically for the sake of man's health. In this review article, we will give some examples of snake venom components which have spurred the development of novel pharmaceutical compounds. Moreover, we will provide examples where such snake toxin-derived mimetics are in clinical use, trials, or consideration for further pharmaceutical exploitation, especially in the fields of hemostasis, thrombosis, coagulation, and metastasis. Thus, it becomes clear why a snake captured its symbolic place at the Asclepius rod with good reason still nowadays.
Collapse
Affiliation(s)
- Maria-Inacia Estevão-Costa
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Raquel Sanz-Soler
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Benjamin Johanningmeier
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany.
| |
Collapse
|
48
|
l-Amino acid oxidase from Cerastes vipera snake venom: Isolation, characterization and biological effects on bacteria and tumor cell lines. Toxicon 2018; 150:270-279. [DOI: 10.1016/j.toxicon.2018.06.064] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 05/27/2018] [Accepted: 06/07/2018] [Indexed: 11/22/2022]
|
49
|
Zainal Abidin SA, Rajadurai P, Chowdhury MEH, Ahmad Rusmili MR, Othman I, Naidu R. Cytotoxic, Antiproliferative and Apoptosis-inducing Activity of L-Amino Acid Oxidase from MalaysianCalloselasma rhodostomaon Human Colon Cancer Cells. Basic Clin Pharmacol Toxicol 2018; 123:577-588. [DOI: 10.1111/bcpt.13060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 06/11/2018] [Indexed: 02/04/2023]
Affiliation(s)
- Syafiq Asnawi Zainal Abidin
- Jeffrey Cheah School of Medicine and Health Sciences; Monash University Malaysia; Bandar Sunway Selangor Darul Ehsan Malaysia
| | - Pathmanathan Rajadurai
- Jeffrey Cheah School of Medicine and Health Sciences; Monash University Malaysia; Bandar Sunway Selangor Darul Ehsan Malaysia
| | - Md. Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences; Monash University Malaysia; Bandar Sunway Selangor Darul Ehsan Malaysia
| | | | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences; Monash University Malaysia; Bandar Sunway Selangor Darul Ehsan Malaysia
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences; Monash University Malaysia; Bandar Sunway Selangor Darul Ehsan Malaysia
| |
Collapse
|
50
|
Zainal Abidin SA, Rajadurai P, Hoque Chowdhury ME, Othman I, Naidu R. Cytotoxic, Anti-Proliferative and Apoptosis Activity of l-Amino Acid Oxidase from Malaysian Cryptelytrops purpureomaculatus (CP-LAAO) Venom on Human Colon Cancer Cells. Molecules 2018; 23:molecules23061388. [PMID: 29890640 PMCID: PMC6100610 DOI: 10.3390/molecules23061388] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/24/2018] [Accepted: 05/31/2018] [Indexed: 01/16/2023] Open
Abstract
The aim of this study is to investigate the potential anti-cancer activity of l-amino acid oxidase (CP-LAAO) purified from the venom of Cryptelytrops purpureomaculatus on SW480 and SW620 human colon cancer cells. Mass spectrometry guided purification was able to identify and purify CP-LAAO. Amino acid variations identified from the partial protein sequence of CP-LAAO may suggest novel variants of these proteins. The activity of the purified CP-LAAO was confirmed with o-phenyldiamine (OPD)-based spectrophotometric assay. CP-LAAO demonstrated time- and dose-dependent cytotoxic activity and the EC50 value was determined at 13 µg/mL for both SW480 and SW620 cells. Significant increase of caspase-3 activity, reduction of Bcl-2 levels, as well as morphological changes consistent with apoptosis were demonstrated by CP-LAAO. Overall, these data provide evidence on the potential anti-cancer activity of CP-LAAO from the venom of Malaysian C. purpureomaculatus for therapeutic intervention of human colon cancer.
Collapse
Affiliation(s)
- Syafiq Asnawi Zainal Abidin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia.
| | - Pathmanathan Rajadurai
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia.
| | - Md Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia.
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia.
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia.
| |
Collapse
|