1
|
Liu J, Xue Y, Yan H, Zhou J, Long X, Tang Y. Natural Flavonoids from Licorice as Potent Inhibitors of β-Glucuronidase Elucidated Through Computational Studies. Molecules 2025; 30:1324. [PMID: 40142099 PMCID: PMC11945163 DOI: 10.3390/molecules30061324] [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: 11/30/2024] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Gut bacterial β-glucuronidase is an important molecular target in several therapeutic applications. β-glucuronidase inhibitors can effectively alleviate gastrointestinal toxicity caused by certain drugs. Licorice, a traditional Chinese medicine, harmonizes various herbs and mitigates the toxicity of hundreds of herbs. In this study, a comprehensive computational strategy was employed to evaluate four licorice flavonoids (liquiritigenin, isoliquiritigenin, liquiritin, and isoliquiritin) as potential Escherichia coli β-glucuronidase (EcGUS) inhibitors. Density functional theory was used to determine their geometries, thermal parameters, dipole moments, polarizabilities, and molecular electrostatic potentials. The inhibitory mechanisms of these four flavonoids on EcGUS were investigated using molecular docking, molecular dynamics simulations, and free energy calculations. The results show that all four flavonoids stably bind to EcGUS. Moreover, all molecules, except liquiritigenin, are potent and selective inhibitors of EcGUS. Further calculations suggest that isoliquiritin exhibits the strongest binding affinity for EcGUS among the four licorice flavonoids. Thus, isoliquiritin is a promising candidate for the development of EcGUS inhibitors. These findings will aid in designing and developing novel flavonoid-based inhibitors of EcGUS to alleviate gastrointestinal toxicity caused by drugs.
Collapse
Affiliation(s)
- Jingli Liu
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (J.L.); (Y.X.); (H.Y.); (J.Z.); (X.L.)
| | - Yingying Xue
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (J.L.); (Y.X.); (H.Y.); (J.Z.); (X.L.)
| | - Hao Yan
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (J.L.); (Y.X.); (H.Y.); (J.Z.); (X.L.)
| | - Jing Zhou
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (J.L.); (Y.X.); (H.Y.); (J.Z.); (X.L.)
| | - Xu Long
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (J.L.); (Y.X.); (H.Y.); (J.Z.); (X.L.)
| | - Yuping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| |
Collapse
|
2
|
Chen L, Lai J, Luo Y, Shu T, Lv B, Li C. Efficient glycyrrhetinic acid biomanufacturing through protein engineering and dual-GUS combination strategy with novel β-glucuronidase from Aspergillus calidoustus CLH-22. BIORESOURCE TECHNOLOGY 2024; 413:131436. [PMID: 39245064 DOI: 10.1016/j.biortech.2024.131436] [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: 06/12/2024] [Revised: 08/02/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
Glycyrrhetinicacid (GA) is a high-value pentacyclic triterpenoid with broad applications. However, the industrial production of GA is hindered by low yield and the accumulation of the intermediate product GlycyrrhetinicAcid3-O-Mono-β-D-Glucuronide (GAMG). This study first identified a novel β-glucuronidase (AcGUS) from Aspergillus calidoustus CLH-22 through transcriptomic analysis, demonstrating a substrate preference for GAMG. Subsequently, mutant AcGUS3G461C/Q462H/I575K with significantly improved activity (kcat/Km of 11.02-fold) was obtained via computer-aided engineering. Furthermore, the dual-GUS combination strategy was employed for the first timeto construct engineered Pichia pastoris for GA production, offering multiple advantages of enhanced conversion efficiency and reduced fermentation viscosity. Finally, under systematically optimized conditions and employing Glycyrrhizin (GL) as the substrate, the final concentration of GA was 48.73 g/L with a conversion of 97.26 % in a 1000-L fermenter, representing the optimal biocatalytic performance reported to date. This study provides new ideas and insights for industrial GA production.
Collapse
Affiliation(s)
- Linhao Chen
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Junjie Lai
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Yan Luo
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Tao Shu
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Bo Lv
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China.
| | - Chun Li
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China; Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
3
|
Tseng YH, Lin HP, Lin SY, Chen BM, Vo TNN, Yang SH, Lin YC, Prijovic Z, Czosseck A, Leu YL, Roffler SR. Engineering stable and non-immunogenic immunoenzymes for cancer therapy via in situ generated prodrugs. J Control Release 2024; 369:179-198. [PMID: 38368947 DOI: 10.1016/j.jconrel.2024.02.026] [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: 11/06/2023] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Engineering human enzymes for therapeutic applications is attractive but introducing new amino acids may adversely affect enzyme stability and immunogenicity. Here we used a mammalian membrane-tethered screening system (ECSTASY) to evolve human lysosomal beta-glucuronidase (hBG) to hydrolyze a glucuronide metabolite (SN-38G) of the anticancer drug irinotecan (CPT-11). Three human beta-glucuronidase variants (hBG3, hBG10 and hBG19) with 3, 10 and 19 amino acid substitutions were identified that display up to 40-fold enhanced enzymatic activity, higher stability than E. coli beta-glucuronidase in human serum, and similar pharmacokinetics in mice as wild-type hBG. The hBG variants were two to three orders of magnitude less immunogenic than E. coli beta-glucuronidase in hBG transgenic mice. Intravenous administration of an immunoenzyme (hcc49-hBG10) targeting a sialyl-Tn tumor-associated antigen to mice bearing human colon xenografts significantly enhanced the anticancer activity of CPT-11 as measured by tumor suppression and mouse survival. Our results suggest that genetically-modified human enzymes represent a good alternative to microbially-derived enzymes for therapeutic applications.
Collapse
Affiliation(s)
- Yi-Han Tseng
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Hsuan-Pei Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Sung-Yao Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | | | - Shih-Hung Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Chen Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Zeljko Prijovic
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11001, Serbia
| | - Andreas Czosseck
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Lin Leu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
4
|
Fernández-Murga ML, Gil-Ortiz F, Serrano-García L, Llombart-Cussac A. A New Paradigm in the Relationship between Gut Microbiota and Breast Cancer: β-glucuronidase Enzyme Identified as Potential Therapeutic Target. Pathogens 2023; 12:1086. [PMID: 37764894 PMCID: PMC10535898 DOI: 10.3390/pathogens12091086] [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: 07/12/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer (BC) is the most frequently occurring malignancy and the second cancer-specific cause of mortality in women in developed countries. Over 70% of the total number of BCs are hormone receptor-positive (HR+), and elevated levels of circulating estrogen (E) in the blood have been shown to be a major risk factor for the development of HR+ BC. This is attributable to estrogen's contribution to increased cancer cell proliferation, stimulation of angiogenesis and metastasis, and resistance to therapy. The E metabolism-gut microbiome axis is functional, with subjacent individual variations in the levels of E. It is conceivable that the estrobolome (bacterial genes whose products metabolize E) may contribute to the risk of malignant neoplasms of hormonal origin, including BC, and may serve as a potential biomarker and target. It has been suggested that β-glucuronidase (GUS) enzymes of the intestinal microbiome participate in the strobolome. In addition, it has been proposed that bacterial GUS enzymes from the gastrointestinal tract participate in hormone BC. In this review, we discuss the latest knowledge about the role of the GUS enzyme in the pathogenesis of BC, focusing on (i) the microbiome and E metabolism; (ii) diet, estrobolome, and BC development; (iii) other activities of the bacterial GUS; and (iv) the new molecular targets for BC therapeutic application.
Collapse
Affiliation(s)
- M. Leonor Fernández-Murga
- Clinical and Molecular Oncology Laboratory, Hospital Arnau de Vilanova-Liria, FISABIO, 46015 Valencia, Spain; (L.S.-G.); (A.L.-C.)
| | | | - Lucía Serrano-García
- Clinical and Molecular Oncology Laboratory, Hospital Arnau de Vilanova-Liria, FISABIO, 46015 Valencia, Spain; (L.S.-G.); (A.L.-C.)
| | - Antonio Llombart-Cussac
- Clinical and Molecular Oncology Laboratory, Hospital Arnau de Vilanova-Liria, FISABIO, 46015 Valencia, Spain; (L.S.-G.); (A.L.-C.)
| |
Collapse
|
5
|
Taha M, Imran S, Rahim F, Uddin N, Iqbal N, Khan KM, Farooq RK, Alomari M, Islam I, Algheribe S. Discovering biological efficacy of new thiadiazole as effective inhibitors of urease, glycation, and (DPPH) scavengers: Biochemical and in silico study. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
6
|
Wang Q, Lu D, Liu H, Li C. Tetramerization of GH2 β-Glucuronidases is Essential for Catalyzing the Hydrolysis of the Large Substrate Glycyrrhizin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:16286-16297. [PMID: 36516995 DOI: 10.1021/acs.jafc.2c06859] [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: 06/17/2023]
Abstract
In this study, structural analysis was employed to identify three hotspot residues that contribute most to the tetramer formation of a glycoside hydrolase family 2 (GH2) β-glucuronidase (GUS) from Aspergillus oryzae Li-3. Single-point mutation at these sites completely disrupted the tetramer structure and abolished the glycyrrhizin (GL)-hydrolyzing activity. Then, the W522A dimer was refactored into a tetramer by disulfide bonding, and partial GL activity was restored. Further saturated mutation showed a strong correlation between the GL activity of the mutants and their tetramer ratios. Molecular simulations were employed to illustrate the critical role of the tetramer interface in maintaining a functional active-site structure. The three highly conserved tetramer-forming residues were finally applied to two other GH2 GUSs for tetramer dissociation and demonstrated the significance of the homotetramerization for GL-hydrolyzing activity of GH2 GUSs. This study lays foundation for engineering GL-hydrolyzing GUSs at the quaternary structure level for function regulations.
Collapse
Affiliation(s)
- Qibin Wang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 10081, PR China
| | - Dingyuan Lu
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 10081, PR China
| | - Hu Liu
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 10081, PR China
| | - Chun Li
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 10081, PR China
- Key Laboratory for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 10084, PR China
- Center for Synthetic & Systems Biology, Tsinghua University, Beijing 10084, PR China
| |
Collapse
|
7
|
Phong NV, Zhao Y, Min BS, Yang SY, Kim JA. Inhibitory Activity of Bioactive Phloroglucinols from the Rhizomes of Dryopteris crassirhizoma on Escherichia coli β-Glucuronidase: Kinetic Analysis and Molecular Docking Studies. Metabolites 2022; 12:metabo12100938. [PMID: 36295840 PMCID: PMC9610990 DOI: 10.3390/metabo12100938] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 12/13/2022] Open
Abstract
Phloroglucinols-one of the major secondary metabolites in Dryopteris crassirhizoma-exhibit various pharmacological effects, such as antiviral, antioxidant, and antidiabetic activities. This study evaluated 30 phloroglucinols isolated from the rhizomes of D. crassirhizoma for their inhibitory activity on β-glucuronidase via in vitro assays. Among them, dimeric phloroglucinols 13-15 moderately inhibited β-glucuronidase, and trimeric phloroglucinols 26-28 showed strong inhibitory effects, with IC50 values ranging from 5.6 to 8.0 μM. Enzyme kinetic analysis confirmed all six active compounds to be in a competitive mode of inhibition. Molecular docking simulations revealed the key binding interactions with the active site of β-glucuronidase protein and the binding mechanisms of these active metabolites. Our results suggest that the rhizomes of D. crassirhizoma and trimeric compounds 26-28 may serve as potential candidates for discovering and developing new β-glucuronidase inhibitors.
Collapse
Affiliation(s)
- Nguyen Viet Phong
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Yan Zhao
- School of Pharmacy, Yantai University, Yantai 264005, China
| | - Byung Sun Min
- Drug Research and Development Center, College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Korea
| | - Seo Young Yang
- Department of Pharmaceutical Engineering, Sangji University, Wonju 26339, Korea
- Correspondence: (S.Y.Y.); (J.A.K.); Tel.: +82-33-738-7921 (S.Y.Y.); +82-53-950-8574 (J.A.K.)
| | - Jeong Ah Kim
- Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
- Correspondence: (S.Y.Y.); (J.A.K.); Tel.: +82-33-738-7921 (S.Y.Y.); +82-53-950-8574 (J.A.K.)
| |
Collapse
|
8
|
de Boer C, Armstrong Z, Lit VAJ, Barash U, Ruijgrok G, Boyango I, Weitzenberg MM, Schröder SP, Sarris AJC, Meeuwenoord NJ, Bule P, Kayal Y, Ilan N, Codée JDC, Vlodavsky I, Overkleeft HS, Davies GJ, Wu L. Mechanism-based heparanase inhibitors reduce cancer metastasis in vivo. Proc Natl Acad Sci U S A 2022; 119:e2203167119. [PMID: 35881786 PMCID: PMC9351465 DOI: 10.1073/pnas.2203167119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/22/2022] [Indexed: 01/30/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) mediate essential interactions throughout the extracellular matrix (ECM), providing signals that regulate cellular growth and development. Altered HSPG composition during tumorigenesis strongly aids cancer progression. Heparanase (HPSE) is the principal enzyme responsible for extracellular heparan sulfate catabolism and is markedly up-regulated in aggressive cancers. HPSE overactivity degrades HSPGs within the ECM, facilitating metastatic dissemination and releasing mitogens that drive cellular proliferation. Reducing extracellular HPSE activity reduces cancer growth, but few effective inhibitors are known, and none are clinically approved. Inspired by the natural glycosidase inhibitor cyclophellitol, we developed nanomolar mechanism-based, irreversible HPSE inhibitors that are effective within physiological environments. Application of cyclophellitol-derived HPSE inhibitors reduces cancer aggression in cellulo and significantly ameliorates murine metastasis. Mechanism-based irreversible HPSE inhibition is an unexplored anticancer strategy. We demonstrate the feasibility of such compounds to control pathological HPSE-driven malignancies.
Collapse
Affiliation(s)
- Casper de Boer
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Zachary Armstrong
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
- Current address: Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Vincent A. J. Lit
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Uri Barash
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Gijs Ruijgrok
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Ilanit Boyango
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Merle M. Weitzenberg
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Sybrin P. Schröder
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Alexi J. C. Sarris
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Nico J. Meeuwenoord
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Pedro Bule
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
- Current address: Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal
| | - Yasmine Kayal
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Jeroen D. C. Codée
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Herman S. Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Gideon J. Davies
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| | - Liang Wu
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
- The Rosalind Franklin Institute, Harwell, OX11 0FA, UK
| |
Collapse
|
9
|
Structural and biochemical basis of a marine bacterial glycoside hydrolase family 2 β-glycosidase with broad substrate specificity. Appl Environ Microbiol 2021; 88:e0222621. [PMID: 34818100 DOI: 10.1128/aem.02226-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Uronic acids are commonly found in marine polysaccharides and increase structural complexity sanand intrinsic recalcitrance to enzymatic attack. The glycoside hydrolase family 2 (GH2) include proteins that target sugar conjugates with hexuronates and are involved in the catabolism and cycling of marine polysaccharides. Here, we reported a novel GH2, AqGalA from a marine algae-associated Bacteroidetes with broad-substrate specificity. Biochemical analyses revealed that AqGalA exhibits hydrolyzing activities against β-galacturonide, β-glucuronide, and β-galactopyranoside via retaining mechanisms. We solved the AqGalA crystal structure in complex with galacturonic acid (GalA) and showed (via mutagenesis) that charge characteristics at uronate-binding subsites controlled substrate selectivity for uronide hydrolysis. Additionally, conformational flexibility of the AqGalA active site pocket was proposed as a key component for broad substrate enzyme selectivity. Our AqGalA structural and functional data augments the current understanding of substrate recognition of GH2 enzymes and provided key insights into the bacterial use of uronic acid containing polysaccharides. IMPORTANCE The decomposition of algal glycans driven by marine bacterial communities represents one of the largest heterotrophic transformation of organic matter fueling marine food webs and global carbon cycling. However, our knowledge of the carbohydrate cycling is limited due to structural complexity of marine polysaccharides and the complicated enzymatic machinery of marine microbes. To degrade algal glycan, marine bacteria such as members of Bacteroidetes produce a complex repertoire of carbohydrate-active enzymes (CAZymes) matching the structural specificity of the different carbohydrates. In this study, we investigated an extracellular GH2 β-glycosidase, AqGalA from a marine Bacteroidetes to identify the key components responsible for glycuronides recognition and hydrolysis. The broad substrate specificity of AqGalA against glycosides with diverse stereochemical substitutions indicates its potential in processing complex marine polysaccharides. Our findings promote a better understanding of microbially-driven mechanisms of marine carbohydrate cycling.
Collapse
|
10
|
Sui Y, Wu J, Chen J. The Role of Gut Microbial β-Glucuronidase in Estrogen Reactivation and Breast Cancer. Front Cell Dev Biol 2021; 9:631552. [PMID: 34458248 PMCID: PMC8388929 DOI: 10.3389/fcell.2021.631552] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 07/09/2021] [Indexed: 12/15/2022] Open
Abstract
Over the past decade, the gut microbiota has received considerable attention for its interactions with the host. Microbial β-glucuronidase generated by this community has hence aroused concern for its biotransformation activity to a wide range of exogenous (foreign) and endogenous compounds. Lately, the role of gut microbial β-glucuronidase in the pathogenesis of breast cancer has been proposed for its estrogen reactivation activity. This is plausible considering that estrogen glucuronides are the primary products of estrogens' hepatic phase II metabolism and are subject to β-glucuronidase-catalyzed hydrolysis in the gut via bile excretion. However, research in this field is still at its very preliminary stage. This review outlines the biology of microbial β-glucuronidase in the gastrointestinal tract and elaborates on the clues to the existence of microbial β-glucuronidase-estrogen metabolism-breast cancer axis. The research gaps in this field will be discussed and possible strategies to address these challenges are suggested.
Collapse
Affiliation(s)
- Yue Sui
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Jianming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| |
Collapse
|
11
|
Marchetti M, Faggiano S, Mozzarelli A. Enzyme Replacement Therapy for Genetic Disorders Associated with Enzyme Deficiency. Curr Med Chem 2021; 29:489-525. [PMID: 34042028 DOI: 10.2174/0929867328666210526144654] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/23/2021] [Accepted: 03/17/2021] [Indexed: 11/22/2022]
Abstract
Mutations in human genes might lead to loss of functional proteins, causing diseases. Among these genetic disorders, a large class is associated with the deficiency in metabolic enzymes, resulting in both an increase in the concentration of substrates and a loss in the metabolites produced by the catalyzed reactions. The identification of therapeutic actions based on small molecules represents a challenge to medicinal chemists because the target is missing. Alternative approaches are biology-based, ranging from gene and stem cell therapy, CRISPR/Cas9 technology, distinct types of RNAs, and enzyme replacement therapy (ERT). This review will focus on the latter approach that since the 1990s has been successfully applied to cure many rare diseases, most of them being lysosomal storage diseases or metabolic diseases. So far, a dozen enzymes have been approved by FDA/EMA for lysosome storage disorders and only a few for metabolic diseases. Enzymes for replacement therapy are mainly produced in mammalian cells and some in plant cells and yeasts and are further processed to obtain active, highly bioavailable, less degradable products. Issues still under investigation for the increase in ERT efficacy are the optimization of enzymes interaction with cell membrane and internalization, the reduction in immunogenicity, and the overcoming of blood-brain barrier limitations when neuronal cells need to be targeted. Overall, ERT has demonstrated its efficacy and safety in the treatment of many genetic rare diseases, both saving newborn lives and improving patients' life quality, and represents a very successful example of targeted biologics.
Collapse
Affiliation(s)
- Marialaura Marchetti
- Biopharmanet-TEC Interdepartmental Center, University of Parma, Parco Area delle Scienze, Bldg 33., 43124, Parma, Italy
| | - Serena Faggiano
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 23/A, 43124, Parma, Italy
| | - Andrea Mozzarelli
- Institute of Biophysics, National Research Council, Via Moruzzi 1, 56124, Pisa, Italy
| |
Collapse
|
12
|
Wang P, Jia Y, Wu R, Chen Z, Yan R. Human gut bacterial β-glucuronidase inhibition: An emerging approach to manage medication therapy. Biochem Pharmacol 2021; 190:114566. [PMID: 33865833 DOI: 10.1016/j.bcp.2021.114566] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
Bacterial β-glucuronidase enzymes (BGUSs) are at the interface of host-microbial metabolic symbiosis, playing an important role in health and disease as well as medication outcomes (efficacy or toxicity) by deconjugating a large number of endogenous and exogenous glucuronides. In recent years, BGUSs inhibition has emerged as a new approach to manage diseases and medication therapy and attracted an increasing research interest. However, a growing body of evidence underlines great genetic diversity, functional promiscuity and varied inhibition propensity of BGUSs, which have posed big challenges to identifying BGUSs involved in a specific pathophysiological or pharmacological process and developing effective inhibition. In this article, we offered a general introduction of the function, in particular the physiological, pathological and pharmacological roles, of BGUSs and their taxonomic distribution in human gut microbiota, highlighting the structural features (active sites and adjacent loop structures) that affecting the protein-substrate (inhibitor) interactions. Recent advances in BGUSs-mediated deconjugation of drugs and carcinogens and the discovery and applications of BGUS inhibitors in management of medication therapy, typically, irinotecan-induced diarrhea and non-steroidal anti-inflammatory drugs (NSAIDs)-induced enteropathy, were also reviewed. At the end, we discussed the perspectives and the challenges of tailoring BGUS inhibition towards precision medicine.
Collapse
Affiliation(s)
- Panpan Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Yifei Jia
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Rongrong Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Zhiqiang Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China.
| |
Collapse
|
13
|
Liu M, Yu J, Lv B, Hou Y, Liu X, Feng X, Li C. Improving the activity and thermostability of GH2 β-glucuronidases via domain reassembly. Biotechnol Bioeng 2021; 118:1962-1972. [PMID: 33559890 DOI: 10.1002/bit.27710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 01/31/2021] [Indexed: 11/07/2022]
Abstract
Glycoside hydrolase family 2 (GH2) enzymes are generally composed of three domains: TIM-barrel domain (TIM), immunoglobulin-like β-sandwich domain (ISD), and sugar-binding domain (SBD). The combination of these three domains yields multiple structural combinations with different properties. Theoretically, the drawbacks of a given GH2 fold may be circumvented by efficiently reassembling the three domains. However, very few successful cases have been reported. In this study, we used six GH2 β-glucuronidases (GUSs) from bacteria, fungi, or humans as model enzymes and constructed a series of mutants by reassembling the domains from different GUSs. The mutants PGUS-At, GUS-PAA, and GUS-PAP, with reassembled domains from fungal GUSs, showed improved expression levels, activity, and thermostability, respectively. Specifically, compared to the parental enzyme, the mutant PGUS-At displayed 3.8 times higher expression, the mutant GUS-PAA displayed 1.0 time higher catalytic efficiency (kcat /Km ), and the mutant GUS-PAP displayed 7.5 times higher thermostability at 65°C. Furthermore, two-hybrid mutants, GUS-AEA and GUS-PEP, were constructed with the ISD from a bacterial GUS and SBD and TIM domain from fungal GUSs. GUS-AEA and GUS-PEP showed 30.4% and 23.0% higher thermostability than GUS-PAP, respectively. Finally, molecular dynamics simulations were conducted to uncover the molecular reasons for the increased thermostability of the mutant.
Collapse
Affiliation(s)
- Mingzhu Liu
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, PR China
| | - Jing Yu
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, PR China
| | - Bo Lv
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, PR China
| | - Yuhui Hou
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, PR China
| | - Xinhe Liu
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, PR China
| | - Xudong Feng
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, PR China
| | - Chun Li
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Institute of Biochemical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, PR China.,Key Laboratory for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, PR China.,Center for Synthetic & Systems Biology, Tsinghua University, Beijing, PR China
| |
Collapse
|
14
|
Taha M, Uddin N, Ali M, Anouar EH, Rahim F, Khan G, Farooq RK, Gollapalli M, Iqbal N, Farooq M, Khan KM. Inhibition potential of phenyl linked benzimidazole-triazolothiadiazole modular hybrids against β-glucuronidase and their interactions thereof. Int J Biol Macromol 2020; 161:355-363. [DOI: 10.1016/j.ijbiomac.2020.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022]
|
15
|
Besse HC, Chen Y, Scheeren HW, Metselaar JM, Lammers T, Moonen CTW, Hennink WE, Deckers R. A Doxorubicin-Glucuronide Prodrug Released from Nanogels Activated by High-Intensity Focused Ultrasound Liberated β-Glucuronidase. Pharmaceutics 2020; 12:E536. [PMID: 32532061 PMCID: PMC7355552 DOI: 10.3390/pharmaceutics12060536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 11/16/2022] Open
Abstract
The poor pharmacokinetics and selectivity of low-molecular-weight anticancer drugs contribute to the relatively low effectiveness of chemotherapy treatments. To improve the pharmacokinetics and selectivity of these treatments, the combination of a doxorubicin-glucuronide prodrug (DOX-propGA3) nanogel formulation and the liberation of endogenous β-glucuronidase from cells exposed to high-intensity focused ultrasound (HIFU) were investigated in vitro. First, a DOX-propGA3-polymer was synthesized. Subsequently, DOX-propGA3-nanogels were formed from this polymer dissolved in water using inverse mini-emulsion photopolymerization. In the presence of bovine β-glucuronidase, the DOX-propGA3 in the nanogels was quantitatively converted into the chemotherapeutic drug doxorubicin. Exposure of cells to HIFU efficiently induced liberation of endogenous β-glucuronidase, which in turn converted the prodrug released from the DOX-propGA3-nanogels into doxorubicin. β-glucuronidase liberated from cells exposed to HIFU increased the cytotoxicity of DOX-propGA3-nanogels to a similar extend as bovine β-glucuronidase, whereas in the absence of either bovine β-glucuronidase or β-glucuronidase liberated from cells exposed to HIFU, the DOX-propGA3-nanogels hardly showed cytotoxicity. Overall, DOX-propGA3-nanogels systems might help to further improve the outcome of HIFU-related anticancer therapy.
Collapse
Affiliation(s)
- Helena C. Besse
- Division of Imaging and Oncology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (H.C.B.); (C.T.W.M.)
| | - Yinan Chen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (Y.C.); (T.L.); (W.E.H.)
| | - Hans W. Scheeren
- Cluster for Molecular Chemistry, Radboud University, 6525 XZ Nijmegen, The Netherlands;
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, 52074 Aachen, Germany;
| | - Josbert M. Metselaar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, 52074 Aachen, Germany;
- Department of Targeted Therapeutics, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, 7500 AE Enschede, The Netherlands
| | - Twan Lammers
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (Y.C.); (T.L.); (W.E.H.)
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, 52074 Aachen, Germany;
- Department of Targeted Therapeutics, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, 7500 AE Enschede, The Netherlands
| | - Chrit T. W. Moonen
- Division of Imaging and Oncology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (H.C.B.); (C.T.W.M.)
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (Y.C.); (T.L.); (W.E.H.)
| | - Roel Deckers
- Division of Imaging and Oncology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (H.C.B.); (C.T.W.M.)
| |
Collapse
|
16
|
Konada RSR, Venugopal A, Nadimpalli SK. Purification, biochemical and biophysical characterization of lysosomal β-D-glucuronidase from an edible freshwater mussel, Lamellidens corrianus. Int J Biol Macromol 2020; 152:465-472. [PMID: 32084490 DOI: 10.1016/j.ijbiomac.2020.02.190] [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] [Received: 12/03/2019] [Revised: 02/15/2020] [Accepted: 02/17/2020] [Indexed: 10/25/2022]
Abstract
A lysosomal glycosidase, β-glucuronidase, has been purified to homogeneity, from the soluble extracts of a freshwater mussel, L. corrianus, by a series of chromatography techniques involving phenyl-Sepharose, ion exchange, affinity and gel filtration chromatography. In native PAGE, β-glucuronidase resolved into a single band and the molecular mass determined by gel filtration chromatography was found to be 250 kDa. Zymogram analysis with 4-methyl umbelliferyl β-glucuronide substrate validated the purified enzyme as β-glucuronidase. In SDS-PAGE, the purified enzyme was resolved into four sub-units with molecular weights around 90, 75, 65, and 50 kDa, respectively, and two of the subunits (90 and 50 kDa) cross-reacted with human β-glucuronidase antiserum. The optimum pH and temperature of the purified glycosidase were 5.0 and 70 °C, respectively. The enzyme kinetics parameters, substrate affinity (KM) and maximum velocity (Vmax) of the purified protein estimated with p-nitrophenyl β-D-glucuronide were 0.457 mM and 0.11867 μmol-1 min-1 mL-1, respectively. The secondary structure of β-glucuronidase was determined in the far-UV range (190 nm to 230 nm) using CD spectroscopy. Heat denaturation plots determined by CD spectroscopy showed that the purified enzyme was stable up to 70 °C.
Collapse
Affiliation(s)
- Rohit Sai Reddy Konada
- Laboratory for Protein Biochemistry and Glycobiology, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Prof CR Rao Road, Gachibowli, Hyderabad 500046, India.
| | - A Venugopal
- Laboratory for Protein Biochemistry and Glycobiology, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Prof CR Rao Road, Gachibowli, Hyderabad 500046, India.
| | - Siva Kumar Nadimpalli
- Laboratory for Protein Biochemistry and Glycobiology, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Prof CR Rao Road, Gachibowli, Hyderabad 500046, India.
| |
Collapse
|
17
|
Dashnyam P, Lin HY, Chen CY, Gao S, Yeh LF, Hsieh WC, Tu Z, Lin CH. Substituent Position of Iminocyclitols Determines the Potency and Selectivity for Gut Microbial Xenobiotic-Reactivating Enzymes. J Med Chem 2020; 63:4617-4627. [PMID: 32105467 DOI: 10.1021/acs.jmedchem.9b01918] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Selective inhibitors of gut bacterial β-glucuronidases (GUSs) are of particular interest in the prevention of xenobiotic-induced toxicities. This study reports the first structure-activity relationships on potency and selectivity of several iminocyclitols (2-7) for the GUSs. Complex structures of Ruminococcus gnavus GUS with 2-7 explained how charge, conformation, and substituent of iminocyclitols affect their potency and selectivity. N1 of uronic isofagomine (2) made strong electrostatic interactions with two catalytic glutamates of GUSs, resulting in the most potent inhibition (Ki ≥ 11 nM). C6-propyl analogue of 2 (6) displayed 700-fold selectivity for opportunistic bacterial GUSs (Ki = 74 nM for E. coli GUS and 51.8 μM for RgGUS). In comparison with 2, there was 200-fold enhancement in the selectivity, which was attributed to differential interactions between the propyl group and loop 5 residues of the GUSs. The results provide useful insights to develop potent and selective inhibitors for undesired GUSs.
Collapse
Affiliation(s)
- Punsaldulam Dashnyam
- Institute of Biological Chemistry, Academia Sinica, No 128, Academia Road, Taipei 11529, Taiwan.,Molecular and Biological Agricultural Sciences, Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan.,National Chung-Hsing University, Taichung 40227, Taiwan.,Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung 40227, Taiwan
| | - Hsien-Ya Lin
- Institute of Biological Chemistry, Academia Sinica, No 128, Academia Road, Taipei 11529, Taiwan
| | - Chia-Yu Chen
- Institute of Biological Chemistry, Academia Sinica, No 128, Academia Road, Taipei 11529, Taiwan
| | - Shijay Gao
- Institute of Biological Chemistry, Academia Sinica, No 128, Academia Road, Taipei 11529, Taiwan
| | - Lun-Fu Yeh
- Institute of Biological Chemistry, Academia Sinica, No 128, Academia Road, Taipei 11529, Taiwan
| | - Wei-Che Hsieh
- Institute of Biological Chemistry, Academia Sinica, No 128, Academia Road, Taipei 11529, Taiwan
| | - Zhijay Tu
- Institute of Biological Chemistry, Academia Sinica, No 128, Academia Road, Taipei 11529, Taiwan
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, No 128, Academia Road, Taipei 11529, Taiwan.,Molecular and Biological Agricultural Sciences, Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan.,National Chung-Hsing University, Taichung 40227, Taiwan.,Biotechnology Center, National Chung-Hsing University, Taichung 40227, Taiwan.,Department of Chemistry and Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
18
|
Abid O, Imran S, Taha M, Ismail NH, Jamil W, Kashif SM, Khan KM, Yusoff J. Synthesis, β-glucuronidase inhibition and molecular docking studies of cyano-substituted bisindole hydrazone hybrids. Mol Divers 2020; 25:995-1009. [PMID: 32301032 DOI: 10.1007/s11030-020-10084-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/03/2020] [Indexed: 11/30/2022]
Abstract
The β-glucuronidase, a lysosomal enzyme, catalyzes the cleavage of glucuronosyl-O-bonds. Its inhibitors play a significant role in different medicinal therapies as they cause a decrease in carcinogen-induced colonic tumors by reducing the level of toxic substances present in the intestine. Among those inhibitors, bisindole derivatives had displayed promising β-glucuronidase inhibition activity. In the current study, hydrazone derivatives of bisindolymethane (1-30) were synthesized and evaluated for in vitro β-glucuronidase inhibitory activity. Twenty-eight analogs demonstrated better activity (IC50 = 0.50-46.5 µM) than standard D-saccharic acid 1,4-lactone (IC50 = 48.4 ± 1.25 µM). Compounds with hydroxyl group like 6 (0.60 ± 0.01 µM), 20 (1.50 ± 0.10 µM) and 25 (0.50 ± 0.01 µM) exhibited the most potent inhibitory activity, followed by analogs with fluorine 21 (3.50 ± 0.10 µM) and chlorine 23 (8.20 ± 0.20 µM) substituents. The presence of hydroxyl group at the aromatic side chain was observed as the main contributing factor in the inhibitory potential. From the docking studies, it was predicted that the active compounds can fit properly in the binding groove of the β-glucuronidase and displayed significant binding interactions with essential residues.
Collapse
Affiliation(s)
- Obaidurahman Abid
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, 42300, Bandar Puncak Alam, Selangor, Malaysia.,Faculty of Applied Science, Universiti Teknologi MARA (UiTM), 40450, Shah Alam, Selangor, Malaysia.,Department of Chemistry, Faculty of Science, Nangarhar University, Jalalabad, Nangarhar, 2601, Afghanistan
| | - Syahrul Imran
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, 42300, Bandar Puncak Alam, Selangor, Malaysia. .,Faculty of Applied Science, Universiti Teknologi MARA (UiTM), 40450, Shah Alam, Selangor, Malaysia.
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 31441, Dammam, Saudi Arabia.
| | - Nor Hadiani Ismail
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, 42300, Bandar Puncak Alam, Selangor, Malaysia.,Faculty of Applied Science, Universiti Teknologi MARA (UiTM), 40450, Shah Alam, Selangor, Malaysia
| | - Waqas Jamil
- Institute of Advanced Research Studies in Chemical Sciences, University of Sindh Jamshoro, Hyderabad, 76080, Pakistan
| | - Syed Muhammad Kashif
- Institute of Advanced Research Studies in Chemical Sciences, University of Sindh Jamshoro, Hyderabad, 76080, Pakistan
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Juliana Yusoff
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, 42300, Bandar Puncak Alam, Selangor, Malaysia
| |
Collapse
|
19
|
Awolade P, Cele N, Kerru N, Gummidi L, Oluwakemi E, Singh P. Therapeutic significance of β-glucuronidase activity and its inhibitors: A review. Eur J Med Chem 2020; 187:111921. [PMID: 31835168 PMCID: PMC7111419 DOI: 10.1016/j.ejmech.2019.111921] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 01/02/2023]
Abstract
The emergence of disease and dearth of effective pharmacological agents on most therapeutic fronts, constitutes a major threat to global public health and man's existence. Consequently, this has created an exigency in the search for new drugs with improved clinical utility or means of potentiating available ones. To this end, accumulating empirical evidence supports molecular target therapy as a plausible egress and, β-glucuronidase (βGLU) - a lysosomal acid hydrolase responsible for the catalytic deconjugation of β-d-glucuronides has emerged as a viable molecular target for several therapeutic applications. The enzyme's activity level in body fluids is also deemed a potential biomarker for the diagnosis of some pathological conditions. Moreover, due to its role in colon carcinogenesis and certain drug-induced dose-limiting toxicities, the development of potent inhibitors of βGLU in human intestinal microbiota has aroused increased attention over the years. Nevertheless, although our literature survey revealed both natural products and synthetic scaffolds as potential inhibitors of the enzyme, only few of these have found clinical utility, albeit with moderate to poor pharmacokinetic profile. Hence, in this review we present a compendium of exploits in the present millennium directed towards the inhibition of βGLU. The aim is to proffer a platform on which new scaffolds can be modelled for improved βGLU inhibitory potency and the development of new therapeutic agents in consequential.
Collapse
Affiliation(s)
- Paul Awolade
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Nosipho Cele
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Nagaraju Kerru
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Lalitha Gummidi
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Ebenezer Oluwakemi
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Parvesh Singh
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa.
| |
Collapse
|
20
|
Kurdziel M, Kopeć M, Pâris A, Lewiński K, Lafite P, Daniellou R. Thioglycoligation of aromatic thiols using a natural glucuronide donor. Org Biomol Chem 2020; 18:5582-5585. [DOI: 10.1039/d0ob00226g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This is the first example of a thioglycoligase that is able to catalyse the formation of S-glucuronides using aromatic thiols and a natural glucuronide donor.
Collapse
Affiliation(s)
- Martyna Kurdziel
- Institut de Chimie Organique et Analytique (ICOA)
- Université d'Orléans/CNRS
- UMR 7311
- Orléans Cedex 2
- France
| | - Magdalena Kopeć
- Institut de Chimie Organique et Analytique (ICOA)
- Université d'Orléans/CNRS
- UMR 7311
- Orléans Cedex 2
- France
| | - Arnaud Pâris
- Institut de Chimie Organique et Analytique (ICOA)
- Université d'Orléans/CNRS
- UMR 7311
- Orléans Cedex 2
- France
| | - Krzysztof Lewiński
- Jagiellonian University
- Faculty of Chemistry
- Department of Crystal Chemistry and Crystal Physics
- Gronostajowa 2
- Poland
| | - Pierre Lafite
- Institut de Chimie Organique et Analytique (ICOA)
- Université d'Orléans/CNRS
- UMR 7311
- Orléans Cedex 2
- France
| | - Richard Daniellou
- Institut de Chimie Organique et Analytique (ICOA)
- Université d'Orléans/CNRS
- UMR 7311
- Orléans Cedex 2
- France
| |
Collapse
|
21
|
Transmembrane signaling on a protocell: Creation of receptor-enzyme chimeras for immunodetection of specific antibodies and antigens. Sci Rep 2019; 9:18189. [PMID: 31796769 PMCID: PMC6890649 DOI: 10.1038/s41598-019-54539-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/13/2019] [Indexed: 01/22/2023] Open
Abstract
It is known that digital counting of fluorescent signals generated in many small compartments can significantly improve the detection sensitivity of the enzyme-linked immunosorbent assay (ELISA). However, the reported digital ELISA systems need extensive washing steps to remove background signal, which hampers their performance. To tackle this problem, we developed a vesicle (Protocell) array wherein binding of an external protein analyte is coupled to signal amplification and intra-vesicular fluorescence readout. We chose β-glucuronidase (GUS) as a reporter enzyme as its function requires assembly of four subunits through dimerization of a pair of dimers that can be inhibited by a set of interface mutations. Using a thermostabilized GUS mutant IV-5, we screened out an interface mutant (M516K, F517W) to create IV5m - a mutant with high thermostability and activity conditional on induced dimerization. After tethering a short N-terminal tag and transmembrane (TM) sequences, the fusion protein was expressed by cell-free protein synthesis inside protocells. When a corresponding tag-specific antibody was applied outside of the protocells, a clear increase in GUS activity was observed inside vesicles by adding fluorescent substrate, probably due to spontaneous integration of the tagged TM protein into the vesicles and dimerization by the antibody bound to the displayed tag. Furthermore, using flow cytometry, quantitative digital read out was obtained by counting fluorescent protocells exposed to varying concentrations of external antibodies that included Trastuzumab. Additionally, through use of an anti-caffeine VHH-SpyCatcher fusion protein, caffeine could be detected using SpyTag-fused TM-IV5m protein expressed in protocells, suggesting utility of this platform for detection of diverse antigen types.
Collapse
|
22
|
Creation of stable and strictly regulated enzyme switch for signal-on immunodetection of various small antigens. J Biosci Bioeng 2019; 128:677-682. [DOI: 10.1016/j.jbiosc.2019.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/15/2019] [Accepted: 05/27/2019] [Indexed: 12/11/2022]
|
23
|
Almandil NB, Taha M, Gollapalli M, Rahim F, Ibrahim M, Mosaddik A, Anouar EH. Indole bearing thiadiazole analogs: synthesis, β-glucuronidase inhibition and molecular docking study. BMC Chem 2019; 13:14. [PMID: 31384763 PMCID: PMC6661955 DOI: 10.1186/s13065-019-0522-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/16/2019] [Indexed: 12/05/2022] Open
Abstract
Indole based thiadiazole derivatives (1-22) have synthesized, characterized by NMR and HREI-MS and evaluated for β-Glucuronidase inhibition. All compounds showed outstanding β-glucuronidase activity with IC50 values ranging between 0.5 ± 0.08 to 38.9 ± 0.8 µM when compared with standard d-saccharic acid 1,4 lactone (IC50 value of 48.1 ± 1.2 µM). The compound 6, a 2,3-dihydroxy analog was found the most potent among the series with IC50 value 0.5 ± 0.08 µM. Structure activity relationship has been established for all compounds. To confirm the binding interactions of these newly synthesized compounds, molecular docking study have been carried out which reveal that these compounds established stronger hydrogen bonding networks with active site residues.
Collapse
Affiliation(s)
- Noor Barak Almandil
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam, 31441 Saudi Arabia
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam, 31441 Saudi Arabia
| | - Mohammed Gollapalli
- Department of Computer Information Systems, College of Computer Science & Information Technology, Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam, 31441 Saudi Arabia
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra, 21300 Khyber Pakhtunkhwa Pakistan
| | - Mohamed Ibrahim
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam, 31441 Saudi Arabia
| | - Ashik Mosaddik
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam, 31441 Saudi Arabia
| | - El Hassane Anouar
- Department of Chemistry, College of Sciences and Humanities, Prince Sattam Bin Abdulaziz University, P.O. Box 83, Al-Kharj, 11942 Saudi Arabia
| |
Collapse
|
24
|
Afzal A, Thayyil MS, Shariq M, Mary YS, Resmi KS, Thomas R, Islam N, Abinu AJ. Anti‐Cancerous Brucine and Colchicine: Experimental and Theoretical Characterization. ChemistrySelect 2019. [DOI: 10.1002/slct.201902698] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Aboothahir Afzal
- Department of PhysicsCalicut University, Malappuram district Kerala
- Department of PhysicsGovt. Arts and Science college Calicut Kerala
| | | | - Mohammad Shariq
- Department of PhysicsFaculty of ScienceJazan University, Jazan Saudi Arabia
| | - Yohannan Sheena Mary
- Department of PhysicsFatima Mata National College(Autonomous), Kollam, Kerala India
| | | | - Renjith Thomas
- Department of ChemistrySt. Berchmans College (Autonomous), Changanasserry, Kerala India
| | - Nasarul Islam
- Department of ChemistryGovt. Degree College, Sopore, J&K-193201 India
| | | |
Collapse
|
25
|
Zhang H, He YB, Wu PF, Zhang SF, Xie ZX, Li DX, Lin L, Chen F, Wang DZ. Functional Differences in the Blooming Phytoplankton Heterosigma akashiwo and Prorocentrum donghaiense Revealed by Comparative Metaproteomics. Appl Environ Microbiol 2019; 85:e01425-19. [PMID: 31375486 PMCID: PMC6752027 DOI: 10.1128/aem.01425-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/07/2019] [Indexed: 12/21/2022] Open
Abstract
Phytoplankton blooms are natural phenomena in the ocean, which are the results of rapid cell growth of some phytoplankton species in a unique environment. However, little is known about the molecular events occurring during the bloom. Here, we compared metaproteomes of two phytoplankton Heterosigma akashiwo and Prorocentrum donghaiense in the coastal East China Sea. H. akashiwo and P. donghaiense accounted for 7.82% and 4.74% of the phytoplankton community protein abundances in the nonbloom sample, whereas they contributed to 60.13% and 78.09%, respectively, in their individual blooming samples. Compared with P. donghaiense, H. akashiwo possessed a significantly higher abundance of light-harvesting complex proteins, carbonic anhydrasem and RuBisCO. The blooming H. akashiwo cells expressed more proteins related to external nutrient acquisition, such as bicarbonate transporter SLC4, ammonium transporter, nitrite transporter, and alkaline phosphatase, while the blooming P. donghaiense cells highly expressed proteins related to extra- and intracellular organic nutrient utilization, such as amino acid transporter, 5'-nucleotidase, acid phosphatase, and tripeptidyl-peptidase. The strong capabilities of light harvesting, as well as acquisition and assimilation of inorganic carbon, nitrogen, and phosphorus, facilitated the formation of the H. akashiwo bloom under the high turbidity and inorganic nutrient-sufficient condition, whereas the competitive advantages in organic nutrient acquisition and reallocation guaranteed the occurrence of the P. donghaiense bloom under the inorganic nutrient-insufficient condition. This study highlights the power of metaproteomics for revealing the underlying molecular behaviors of different coexisting phytoplankton species and advances our knowledge on the formation of phytoplankton blooms.IMPORTANCE A deep understanding of the mechanisms driving bloom formation is a prerequisite for effective bloom management. Metaproteomics was applied in this study to reveal the adaptive and responsive strategies of two coexisting phytoplankton species, H. akashiwo and P. donghaiense, during their bloom periods. Metabolic features and niche divergence in light harvesting, as well as carbon, nitrogen, and phosphorus acquisition and assimilation likely promoted the bloom occurrence under different environments. The molecular behaviors of coexisting bloom-causing species will give clues for bloom monitoring and management in the oceans.
Collapse
Affiliation(s)
- Hao Zhang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen, China
| | - Yan-Bin He
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Peng-Fei Wu
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen, China
| | - Shu-Feng Zhang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen, China
| | - Zhang-Xian Xie
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen, China
| | - Dong-Xu Li
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen, China
| | - Lin Lin
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen, China
| | - Feng Chen
- Institute of Marine and Environmental Technology, University of Maryland Center for Environmental Science, Baltimore, Maryland, USA
| | - Da-Zhi Wang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen, China
| |
Collapse
|
26
|
Synthesis of oxadiazole-coupled-thiadiazole derivatives as a potent β-glucuronidase inhibitors and their molecular docking study. Bioorg Med Chem 2019; 27:3145-3155. [DOI: 10.1016/j.bmc.2019.05.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/16/2019] [Accepted: 05/31/2019] [Indexed: 11/19/2022]
|
27
|
Chamseddine AN, Ducreux M, Armand JP, Paoletti X, Satar T, Paci A, Mir O. Intestinal bacterial β-glucuronidase as a possible predictive biomarker of irinotecan-induced diarrhea severity. Pharmacol Ther 2019; 199:1-15. [DOI: 10.1016/j.pharmthera.2019.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
28
|
Anouar EH, Moustapha ME, Taha M, Geesi MH, Farag ZR, Rahim F, Almandil NB, Farooq RK, Nawaz M, Mosaddik A. Synthesis, Molecular Docking and β-Glucuronidase Inhibitory Potential of Indole Base Oxadiazole Derivatives. Molecules 2019; 24:molecules24050963. [PMID: 30857263 PMCID: PMC6429331 DOI: 10.3390/molecules24050963] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 12/31/2022] Open
Abstract
β-glucuronidase is a lysosomal glycosidase enzyme which catalyzes the extracellular matrix of cancer and normal cells and the glycosaminoglycans of the cell membrane, which is important for cancer cell proliferation, invasion, and metastasis. Liver cancer, colon carcinoma, and neoplasm bladder are triggered by the increase of the level of β-glucuronidase activity. The most valuable structures are indole and oxadiazole which has gain immense attention because of its pharmacological behavior and display many biological properties. Twenty-two (1⁻22) analogs of indole based oxadiazole were synthesized and screened for their inhibitory potential against β-glucuronidase. Majority of the compounds showed potent inhibitory potential with IC50 values ranging between 0.9 ± 0.01 to 46.4 ± 0.9 µM, under positive control of standard drug d-saccharic acid 1,4 lactone (IC50 = 48.1 ± 1.2 µM). Structural activity relationship (SAR) has been established for all synthesized compounds. To shed light on molecular interactions between the synthesized compounds and β-glucuronidase, 1, 4, and 6 compounds were docked into the active binding site of β-glucuronidase. The obtained results showed that this binding is thermodynamically favorable and β-glucuronidase inhibition of the selected compounds increases with the number of hydrogen bonding established in selected compound-β-glucuronidase complexes.
Collapse
Affiliation(s)
- El Hassane Anouar
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, P.O. Box 83, 11942 Al Kharj, Saudi Arabia.
| | - Moustapha Eid Moustapha
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, P.O. Box 83, 11942 Al Kharj, Saudi Arabia.
- University Central Laboratory, College of Science and Humanities, Prince Sattam bin Abdulaziz University, P.O. Box 83, 11942 Al Kharj, Saudi Arabia.
| | - Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441 Dammam, Saudi Arabia.
| | - Mohammed H Geesi
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, P.O. Box 83, 11942 Al Kharj, Saudi Arabia.
| | - Zeinab R Farag
- Chemistry Department, Faculty of Science, Fayoum University, 63514 Fayoum, Egypt.
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra-21300, Khyber Pakhtunkhwa 21300, Pakistan.
| | - Noor Barak Almandil
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441 Dammam, Saudi Arabia.
| | - Rai Khalid Farooq
- Department of Neuroscience Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441 Dammam, Saudi Arabia.
| | - Muhammad Nawaz
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441 Dammam, Saudi Arabia.
| | - Ashik Mosaddik
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441 Dammam, Saudi Arabia.
| |
Collapse
|
29
|
Chen J, Guo X, Zhu M, Chen C, Li D. Polysaccharide monooxygenase-catalyzed oxidation of cellulose to glucuronic acid-containing cello-oligosaccharides. BIOTECHNOLOGY FOR BIOFUELS 2019; 12:42. [PMID: 30858879 PMCID: PMC6391835 DOI: 10.1186/s13068-019-1384-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/20/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Polysaccharide monooxygenases (PMOs) play an important role in the enzymatic degradation of cellulose. They have been demonstrated to able to C6-oxidize cellulose to produce C6-hexodialdoses. However, the biological function of C6 oxidation of PMOs remains unknown. In particular, it is unclear whether C6-hexodialdoses can be further oxidized to uronic acid (glucuronic acid-containing oligosaccharides). RESULTS A PMO gene, Hipmo1, was isolated from Humicola insolens and expressed in Pichia pastoris. This PMO (HiPMO1), belonging to the auxiliary activity 9 (AA9) family, was shown to able to cleave cellulose to yield non-oxidized and oxidized cello-oligosaccharides. The enzyme oxidizes C6 positions in cellulose to form glucuronic acid-containing cello-oligosaccharides, followed by hydrolysis with beta-glucosidase and beta-glucuronidase to yield glucose, glucuronic acid, and saccharic acid. This indicates that HiPMO1 can catalyze C6 oxidation of hydroxyl groups of cellulose to carboxylic groups. CONCLUSIONS HiPMO1 oxidizes C6 of cellulose to form glucuronic acid-containing cello-oligosaccharides followed by hydrolysis with beta-glucosidase and beta-glucuronidase to yield glucose, glucuronic acid, and saccharic acid, and even possibly by beta-eliminative cleavage to produce unsaturated cello-oligosaccharides. This study provides a new mechanism for cellulose cleavage by C6 oxidation of HiPMO1.
Collapse
Affiliation(s)
- Jinyin Chen
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| | - Xiuna Guo
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| | - Min Zhu
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| | - Chen Chen
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| | - Duochuan Li
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| |
Collapse
|
30
|
Kantaputra PN, Smith LJ, Casal ML, Kuptanon C, Chang YC, Nampoothiri S, Paiyarom A, Veerasakulwong T, Trachoo O, Ketudat Cairns JR, Chinadet W, Tanpaiboon P. Oral manifestations in patients and dogs with mucopolysaccharidosis Type VII. Am J Med Genet A 2019; 179:486-493. [PMID: 30653816 DOI: 10.1002/ajmg.a.61034] [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: 11/12/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 11/07/2022]
Abstract
Mucopolysaccharidosis Type VII (MPS7, also called β-glucuronidase deficiency or Sly syndrome; MIM 253220) is an extremely rare autosomal recessive lysosomal storage disease, caused by mutations in the GUSB gene. β-glucuronidase (GUSB) is a lysosomal hydrolase involved in the stepwise degradation of glucuronic acid-containing glycosaminoglycans (GAGs). Patients affected with MPS VII are not able to completely degrade glucuronic acid-containing GAGs, including chondroitin 4-sulfate, chondroitin 6-sulfate, dermatan sulfate, and heparan sulfate. The accumulation of these GAGs in lysosomes of various tissues leads to cellular and organ dysfunctions. Characteristic features of MPS VII include short stature, macrocephaly, hirsutism, coarse facies, hearing loss, cloudy cornea, short neck, valvular cardiac defects, hepatosplenomegaly, and dysostosis multiplex. Oral manifestations in patients affected with MPS VII have never been reported. Oral manifestations observed in three patients consist of wide root canal spaces, taurodontism, hyperplastic dental follicles, malposition of unerupted permanent molars, and failure of tooth eruption with malformed roots. The unusual skeletal features of the patients include maxillary hypoplasia, hypoplastic midface, long mandibular length, mandibular prognathism, hypoplastic and aplastic mandibular condyles, absence of the dens of the second cervical vertebra, and erosion of the cortex of the lower border of mandibles. Dogs affected with MPS VII had anterior and posterior open bite, maxillary hypoplasia, premolar crowding, and mandibular prognathism. Unlike patients with MPS VII, the dogs had unremarkable mandibular condyles. This is the first report of oral manifestations in patients affected with MPS VII.
Collapse
Affiliation(s)
- Piranit N Kantaputra
- Center of Excellence in Medical Genetics Research, Chiang Mai University, Chiang Mai, Thailand.,Division of Pediatric Dentistry, Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.,Dentaland Clinic, Chiang Mai, Thailand
| | - Lachlan J Smith
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Margret L Casal
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chulaluck Kuptanon
- Department of Pediatrics, College of Medicine, Rangsit University, Bangkok, Thailand.,Division of Genetics, Queen Sirikit National Institute of Child Health, Department of Medical Services, Ministry of Public Health, Bangkok, Thailand
| | - Yu-Cheng Chang
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences & Research Centre, AIMS Ponekkara PO, Cochin, Kerala, India
| | | | | | | | - James R Ketudat Cairns
- School of Chemistry, Institute of Science, and Center for Biomolecular Structure, Function and Application, Suranaree University of Technology, Nakhon Ratchasima, Thailand.,Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand
| | - Wannapa Chinadet
- Center of Excellence in Medical Genetics Research, Chiang Mai University, Chiang Mai, Thailand.,Division of Pediatric Dentistry, Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | | |
Collapse
|
31
|
X-Ray Crystallography in Structure-Function Characterization of Therapeutic Enzymes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1148:81-103. [DOI: 10.1007/978-981-13-7709-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
32
|
Brissonnet Y, Compain G, Renoux B, Krammer EM, Daligault F, Deniaud D, Papot S, Gouin SG. Monitoring glycosidase activity for clustered sugar substrates, a study on β-glucuronidase. RSC Adv 2019; 9:40263-40267. [PMID: 35542663 PMCID: PMC9076263 DOI: 10.1039/c9ra08847d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/18/2019] [Indexed: 12/15/2022] Open
Abstract
Enzymatically-triggered probes to determine glucuronidase hydrolysis kinetics for clustered substrates.
Collapse
Affiliation(s)
- Yoan Brissonnet
- Université de Nantes
- CEISAM, Chimie Et Interdisciplinarité, Synthèse, Analyse, Modélisation
- UMR CNRS 6230
- UFR des Sciences et des Techniques
- 44322 Nantes Cedex 3
| | - Guillaume Compain
- Institut de Chimie des Milieux et des Matériaux de Poitiers
- IC2MP
- Université de Poitiers
- UMR-CNRS 7285
- 86022 Poitiers
| | - Brigitte Renoux
- Institut de Chimie des Milieux et des Matériaux de Poitiers
- IC2MP
- Université de Poitiers
- UMR-CNRS 7285
- 86022 Poitiers
| | - Eva-Maria Krammer
- Structure et Fonction des Membranes Biologiques
- Université Libre de Bruxelles (ULB)
- Brussels
- Belgium
| | - Franck Daligault
- Université de Nantes
- UFIP
- UMR CNRS 6286
- UFR des Sciences et des Techniques
- France
| | - David Deniaud
- Université de Nantes
- CEISAM, Chimie Et Interdisciplinarité, Synthèse, Analyse, Modélisation
- UMR CNRS 6230
- UFR des Sciences et des Techniques
- 44322 Nantes Cedex 3
| | - Sébastien Papot
- Institut de Chimie des Milieux et des Matériaux de Poitiers
- IC2MP
- Université de Poitiers
- UMR-CNRS 7285
- 86022 Poitiers
| | - Sébastien G. Gouin
- Université de Nantes
- CEISAM, Chimie Et Interdisciplinarité, Synthèse, Analyse, Modélisation
- UMR CNRS 6230
- UFR des Sciences et des Techniques
- 44322 Nantes Cedex 3
| |
Collapse
|
33
|
Haq IU, Akram F. Enhanced production, overexpression and characterization of a hyperthermophilic multimodular GH family 2 β‑glucuronidase (TpGUS) cloned from Thermotoga petrophila RKU-1 T in a mesophilic host. Int J Biol Macromol 2018; 123:1132-1142. [PMID: 30465846 DOI: 10.1016/j.ijbiomac.2018.11.189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 11/02/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022]
Abstract
A multimodular hyperthermophilic β‑glucuronidase (TpGUS) from Thermotoga petrophila RKU-1T, belongs to glycoside hydrolase family 2 (GH2), was cloned and overexpressed in Escherichia coli BL21 CodonPlus (DE3)-RIPL. Expression and production of extracellular TpGUS was enhanced through various specific cultivation and induction strategies. Extracellular TpGUS activity was improved by 3.44 and 7 fold in 4 × ZB medium induced with 0.5 mM IPTG and 100 mM lactose, respectively. The enzyme was purified to homogeneity with a single band of 65.6 kDa on SDS-PAGE, using two subsequent steps of anion exchange and hydrophobic interaction chromatography after heat precipitation (70 °C, 1 h). Optimal activity of TpGUS was observed at 95 °C and pH 6.0; and it displayed prodigious thermal stability over a temperature range of 50-85 °C for 12 h at pH 6.0-7.5. Km, Vmax, VmaxKm-1, kcat, and kcatKm-1 were calculated to be 0.7 mM, 227 mmol mg-1 min-1, 324.3 min-1, 164,492.7 s-1 and 234,989.6 mM-1 s-1, respectively using pNPGU as a substrate. Recombinant TpGUS exhibited favorable properties which make this a promising candidate for various biotechnological and pharmacological applications.
Collapse
Affiliation(s)
- Ikram Ul Haq
- Institute of Industrial Biotechnology, GC University, Lahore 54000, Pakistan.
| | - Fatima Akram
- Institute of Industrial Biotechnology, GC University, Lahore 54000, Pakistan
| |
Collapse
|
34
|
Dashnyam P, Mudududdla R, Hsieh TJ, Lin TC, Lin HY, Chen PY, Hsu CY, Lin CH. β-Glucuronidases of opportunistic bacteria are the major contributors to xenobiotic-induced toxicity in the gut. Sci Rep 2018; 8:16372. [PMID: 30401818 PMCID: PMC6219552 DOI: 10.1038/s41598-018-34678-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023] Open
Abstract
Gut bacterial β-D-glucuronidases (GUSs) catalyze the removal of glucuronic acid from liver-produced β-D-glucuronides. These reactions can have deleterious consequences when they reverse xenobiotic metabolism. The human gut contains hundreds of GUSs of variable sequences and structures. To understand how any particular bacterial GUS(s) contributes to global GUS activity and affects human health, the individual substrate preference(s) must be known. Herein, we report that representative GUSs vary in their ability to produce various xenobiotics from their respective glucuronides. To attempt to explain the distinct substrate preference, we solved the structure of a bacterial GUS complexed with coumarin-3-β-D-glucuronide. Comparisons of this structure with other GUS structures identified differences in loop 3 (or the α2-helix loop) and loop 5 at the aglycone-binding site, where differences in their conformations, hydrophobicities and flexibilities appear to underlie the distinct substrate preference(s) of the GUSs. Additional sequence, structural and functional analysis indicated that several groups of functionally related gut bacterial GUSs exist. Our results pinpoint opportunistic gut bacterial GUSs as those that cause xenobiotic-induced toxicity. We propose a structure-activity relationship that should allow both the prediction of the functional roles of GUSs and the design of selective inhibitors.
Collapse
Affiliation(s)
- Punsaldulam Dashnyam
- 0000 0000 9360 4962grid.469086.5Molecular and Biological Agricultural Sciences, Taiwan International Graduate Program, Academia Sinica and National Chung-Hsing University, Taipei, 11529 Taiwan ,0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, 11529 Taiwan ,0000 0004 0532 3749grid.260542.7Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung, 40227 Taiwan
| | - Ramesh Mudududdla
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, 11529 Taiwan
| | - Tung-Ju Hsieh
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, 11529 Taiwan
| | - Ting-Chien Lin
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, 11529 Taiwan
| | - Hsien-Ya Lin
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, 11529 Taiwan
| | - Peng-Yuan Chen
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, 11529 Taiwan
| | - Chia-Yi Hsu
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, 11529 Taiwan
| | - Chun-Hung Lin
- 0000 0000 9360 4962grid.469086.5Molecular and Biological Agricultural Sciences, Taiwan International Graduate Program, Academia Sinica and National Chung-Hsing University, Taipei, 11529 Taiwan ,0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, 11529 Taiwan ,0000 0004 0532 3749grid.260542.7Biotechnology Center, National Chung-Hsing University, Taichung, 40227 Taiwan ,0000 0004 0546 0241grid.19188.39Department of Chemistry and Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617 Taiwan
| |
Collapse
|
35
|
Han B, Hou Y, Jiang T, Lv B, Zhao L, Feng X, Li C. Computation-Aided Rational Deletion of C-Terminal Region Improved the Stability, Activity, and Expression Level of GH2 β-Glucuronidase. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:11380-11389. [PMID: 30296070 DOI: 10.1021/acs.jafc.8b03449] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In this study, computation-aided design on the basis of structural analysis was employed to rationally identify a highly dynamic C-terminal region that regulates the stability, expression level, and activity of a GH2 fungal glucuronidase from Aspergillus oryzae Li-3 (PGUS). Then, four mutants with a precisely truncated C-terminal region in different lengths were constructed; among them, mutant D591-604 with a 3.8-fold increase in half-life at 65 °C and a 6.8 kJ/mol increase in Gibbs free energy showed obviously improved kinetic and thermodynamic stability in comparison to PGUS. Mutants D590-604 and D591-604 both showed approximately 2.4-fold increases in the catalytic efficiency kcat/ Km and 1.8-fold increases in the expression level. Additionally, the expression level of PGUS was doubled through a C-terminal region swap with bacterial GUS from E. coli (EGUS). Finally, the robust PGUS mutants D590-604 and D591-604 were applied in the preparation of glycyrrhetinic acid with 4.0- and 4.4-fold increases in concentration through glycyrrhizin hydrolysis by a fed-batch process.
Collapse
Affiliation(s)
- Beijia Han
- Institute for Synthetic Biosystem/Department of Biochemical Engineering, School of Chemistry and Chemical Engineering , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| | - Yuhui Hou
- Institute for Synthetic Biosystem/Department of Biochemical Engineering, School of Chemistry and Chemical Engineering , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| | - Tian Jiang
- Institute for Synthetic Biosystem/Department of Biochemical Engineering, School of Chemistry and Chemical Engineering , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| | - Bo Lv
- Institute for Synthetic Biosystem/Department of Biochemical Engineering, School of Chemistry and Chemical Engineering , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| | - Lina Zhao
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Xudong Feng
- Institute for Synthetic Biosystem/Department of Biochemical Engineering, School of Chemistry and Chemical Engineering , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| | - Chun Li
- Institute for Synthetic Biosystem/Department of Biochemical Engineering, School of Chemistry and Chemical Engineering , Beijing Institute of Technology , Beijing 100081 , People's Republic of China
| |
Collapse
|
36
|
Pellock SJ, Walton WG, Biernat KA, Torres-Rivera D, Creekmore BC, Xu Y, Liu J, Tripathy A, Stewart LJ, Redinbo MR. Three structurally and functionally distinct β-glucuronidases from the human gut microbe Bacteroides uniformis. J Biol Chem 2018; 293:18559-18573. [PMID: 30301767 DOI: 10.1074/jbc.ra118.005414] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/04/2018] [Indexed: 12/16/2022] Open
Abstract
The glycoside hydrolases encoded by the human gut microbiome play an integral role in processing a variety of exogenous and endogenous glycoconjugates. Here we present three structurally and functionally distinct β-glucuronidase (GUS) glycoside hydrolases from a single human gut commensal microbe, Bacteroides uniformis We show using nine crystal structures, biochemical, and biophysical data that whereas these three proteins share similar overall folds, they exhibit different structural features that create three structurally and functionally unique enzyme active sites. Notably, quaternary structure plays an important role in creating distinct active site features that are hard to predict via structural modeling methods. The enzymes display differential processing capabilities toward glucuronic acid-containing polysaccharides and SN-38-glucuronide, a metabolite of the cancer drug irinotecan. We also demonstrate that GUS-specific and nonselective inhibitors exhibit varying potencies toward each enzyme. Together, these data highlight the diversity of GUS enzymes within a single Bacteroides gut commensal and advance our understanding of how structural details impact the specific roles microbial enzymes play in processing drug-glucuronide and glycan substrates.
Collapse
Affiliation(s)
| | | | | | | | | | - Yongmei Xu
- Chemical Biology and Medicinal Chemistry, and
| | - Jian Liu
- Chemical Biology and Medicinal Chemistry, and
| | | | - Lance J Stewart
- the Department of Biochemistry, Institute for Protein Design, University of Washington, Seattle, Washington 98195
| | - Matthew R Redinbo
- From the Departments of Chemistry, .,Biochemistry and Biophysics, and.,the Departments of Microbiology and Immunology, and Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599 and
| |
Collapse
|
37
|
Guo L, Katiyo W, Lu L, Zhang X, Wang M, Yan J, Ma X, Yang R, Zou L, Zhao W. Glycyrrhetic Acid 3-O-Mono-β-d
-glucuronide (GAMG): An Innovative High-Potency Sweetener with Improved Biological Activities. Compr Rev Food Sci Food Saf 2018; 17:905-919. [DOI: 10.1111/1541-4337.12353] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/17/2018] [Accepted: 03/19/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Lichun Guo
- State Key Laboratory of Food Science and Technology; Jiangnan Univ.; Wuxi Jiangsu 214122 China
| | - Wendy Katiyo
- Dept. of Food Science; Univ. of Pretoria; Hatfield 0028 South Africa
| | - Liushen Lu
- School of Biotechnology; Jiangnan Univ.; Wuxi Jiangsu 214122 China
| | - Xuan Zhang
- State Key Laboratory of Food Science and Technology; Jiangnan Univ.; Wuxi Jiangsu 214122 China
| | - Mingming Wang
- State Key Laboratory of Food Science and Technology; Jiangnan Univ.; Wuxi Jiangsu 214122 China
| | - Jiai Yan
- State Key Laboratory of Food Science and Technology; Jiangnan Univ.; Wuxi Jiangsu 214122 China
| | - Xiaoyun Ma
- School of Foreign Studies; Jiangnan Univ.; Wuxi Jiangsu 214122 China
| | - Ruijin Yang
- State Key Laboratory of Food Science and Technology; Jiangnan Univ.; Wuxi Jiangsu 214122 China
| | - Long Zou
- Bunge Ingredient Innovation Center; 725 North Kinzie Avenue Bradley IL 60915 U.S.A
| | - Wei Zhao
- State Key Laboratory of Food Science and Technology; Jiangnan Univ.; Wuxi Jiangsu 214122 China
| |
Collapse
|
38
|
Increased activity of β -glucuronidase variants produced by site-directed mutagenesis. Enzyme Microb Technol 2018; 109:20-24. [DOI: 10.1016/j.enzmictec.2017.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 09/05/2017] [Accepted: 09/16/2017] [Indexed: 11/22/2022]
|
39
|
Taha M, Arbin M, Ahmat N, Imran S, Rahim F. Synthesis: Small library of hybrid scaffolds of benzothiazole having hydrazone and evaluation of their β-glucuronidase activity. Bioorg Chem 2018; 77:47-55. [PMID: 29331764 DOI: 10.1016/j.bioorg.2018.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/30/2017] [Accepted: 01/02/2018] [Indexed: 10/18/2022]
Abstract
Due to the great biological importance of β-glucuronidase inhibitors, here in this study, we have synthesized a library of novel benzothiazole derivatives (1-30), characterized by different spectroscopic methods and evaluated for β-glucuronidase inhibitory potential. Among the series sixteen compounds i.e.1-6, 8, 9, 11, 14, 15, 20-23 and 26 showed outstanding inhibitory potential with IC50 value ranging in between 16.50 ± 0.26 and 59.45 ± 1.12 when compared with standard d-Saccharic acid 1,4-lactone (48.4 ± 1.25 µM). Except compound 8 and 23 all active analogs showed better potential than the standard. Structure activity relationship has been established.
Collapse
Affiliation(s)
- Muhammad Taha
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 31441, Dammam, Saudi Arabia.
| | - Mastura Arbin
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Puncak Alam Campus, Malaysia; Faculty of Applied Science Universiti Teknologi MARA, 40450 ShahAlam, Selangor D.E, Malaysia
| | - Norizan Ahmat
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Puncak Alam Campus, Malaysia; Faculty of Applied Science Universiti Teknologi MARA, 40450 ShahAlam, Selangor D.E, Malaysia
| | - Syahrul Imran
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Puncak Alam Campus, Malaysia; Faculty of Applied Science Universiti Teknologi MARA, 40450 ShahAlam, Selangor D.E, Malaysia
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra 21120, Pakistan
| |
Collapse
|
40
|
Su J, Dong J, Kitaguchi T, Ohmuro-Matsuyama Y, Ueda H. Noncompetitive homogeneous immunodetection of small molecules based on beta-glucuronidase complementation. Analyst 2018; 143:2096-2101. [DOI: 10.1039/c8an00074c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Small molecules can be sensitively detected with a positive signal by just mixing and measuring the β-glucuronidase activity within 20 min.
Collapse
Affiliation(s)
- Jiulong Su
- Laboratory for Chemistry and Life Science
- Institute of Innovative Research
- Tokyo Institute of Technology
- Japan
| | - Jinhua Dong
- Laboratory for Chemistry and Life Science
- Institute of Innovative Research
- Tokyo Institute of Technology
- Japan
- Key Laboratory of Biological Medicine in Universities of Shandong Province
| | - Tetsuya Kitaguchi
- Laboratory for Chemistry and Life Science
- Institute of Innovative Research
- Tokyo Institute of Technology
- Japan
| | - Yuki Ohmuro-Matsuyama
- Laboratory for Chemistry and Life Science
- Institute of Innovative Research
- Tokyo Institute of Technology
- Japan
| | - Hiroshi Ueda
- Laboratory for Chemistry and Life Science
- Institute of Innovative Research
- Tokyo Institute of Technology
- Japan
| |
Collapse
|
41
|
Chen C, Chen J, Geng Z, Wang M, Liu N, Li D. Regioselectivity of oxidation by a polysaccharide monooxygenase from Chaetomium thermophilum. BIOTECHNOLOGY FOR BIOFUELS 2018; 11:155. [PMID: 29991963 PMCID: PMC5987470 DOI: 10.1186/s13068-018-1156-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 05/29/2018] [Indexed: 05/15/2023]
Abstract
BACKGROUND Polysaccharide monooxygenases (PMOs) of the auxiliary activity 9 (AA9) family have been reported to oxidize C1, C4, and C6 positions in cellulose. However, currently no direct evidence exists that PMOs oxidize C6 positions in cellulose, and molecular mechanism of C1, C4 and C6 oxidation is unclear. RESULTS In this study, a PMO gene (Ctpmo1) belonging to AA9 was isolated from Chaetomium thermophilum and successfully expressed and correctly processed in Pichia pastoris. A simple and effective chemical method of using Br2 to oxidize CtPMO1 reaction products was developed to directly identify C4- and C6-oxidized products by matrix-assisted laser desorption/ionization-time-of-flight tandem mass spectrometry (MALDI-TOF-MS). The PMO (CtPMO1) cleaves phosphoric acid-swollen cellulose (PASC) and celloheptaose, resulting in the formation of oxidized and nonoxidized oligosaccharides. Product identification shows that the enzyme can oxidize C1, C4, and C6 in PASC and cello-oligosaccharides. Mutagenesis of the aromatic residues Tyr27, His64, His157 and residue Tyr206 on the flat surface of CtPMO1 was carried out using site-directed mutagenesis to form the mutated enzymes Y27A, H64A, H157A, and Y206A. It was demonstrated that Y27A retained complete activity of C1, C4, and C6 oxidation on cellulose; Y206A retained partial activity of C1 and C4 oxidation but completely lost activity of C6 oxidation on cellulose; H64A almost completely lost activity of C1, C4, and C6 oxidation on cellulose; and H157A completely lost activity of C1, C4, and C6 oxidation on cellulose. CONCLUSIONS This finding provides direct and molecular evidence for C1, C4, especially C6 oxidation by lytic polysaccharide monooxygenase. CtPMO1 oxidizes not only C1 and C4 but also C6 positions in cellulose. The aromatic acid residues His64, His157 and residue Tyr206 on CtPMO1 flat surface are involved in activity of C1, C4, C6 oxidation.
Collapse
Affiliation(s)
- Chen Chen
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| | - Jinyin Chen
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| | - Zhigang Geng
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| | - Meixia Wang
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| | - Ning Liu
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| | - Duochuan Li
- Department of Mycology, Shandong Agricultural University, Taian, 271018 Shandong China
| |
Collapse
|
42
|
Lv B, Sun H, Huang S, Feng X, Jiang T, Li C. Structure-guided engineering of the substrate specificity of a fungal β-glucuronidase toward triterpenoid saponins. J Biol Chem 2017; 293:433-443. [PMID: 29146597 DOI: 10.1074/jbc.m117.801910] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 11/06/2017] [Indexed: 01/20/2023] Open
Abstract
Glycoside hydrolases (GHs) have attracted special attention in research aimed at modifying natural products by partial removal of sugar moieties to manipulate their solubility and efficacy. However, these modifications are challenging to control because the low substrate specificity of most GHs often generates undesired by-products. We previously identified a GH2-type fungal β-glucuronidase from Aspergillus oryzae (PGUS) exhibiting promiscuous substrate specificity in hydrolysis of triterpenoid saponins. Here, we present the PGUS structure, representing the first structure of a fungal β-glucuronidase, and that of an inactive PGUS mutant in complex with the native substrate glycyrrhetic acid 3-O-mono-β-glucuronide (GAMG). PGUS displayed a homotetramer structure with each monomer comprising three distinct domains: a sugar-binding, an immunoglobulin-like β-sandwich, and a TIM barrel domain. Two catalytic residues, Glu414 and Glu505, acted as acid/base and nucleophile, respectively. Structural and mutational analyses indicated that the GAMG glycan moiety is recognized by polar interactions with nine residues (Asp162, His332, Asp414, Tyr469, Tyr473, Asp505, Arg563, Asn567, and Lys569) and that the aglycone moiety is recognized by aromatic stacking and by a π interaction with the four aromatic residues Tyr469, Phe470, Trp472, and Tyr473 Finally, structure-guided mutagenesis to precisely manipulate PGUS substrate specificity in the biotransformation of glycyrrhizin into GAMG revealed that two amino acids, Ala365 and Arg563, are critical for substrate specificity. Moreover, we obtained several mutants with dramatically improved GAMG yield (>95%). Structural analysis suggested that modulating the interaction of β-glucuronidase simultaneously toward glycan and aglycone moieties is critical for tuning its substrate specificity toward triterpenoid saponins.
Collapse
Affiliation(s)
- Bo Lv
- From the Department of Biochemical Engineering/Institute for Biotransformation and Synthetic Biosystem, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, 100081 Beijing, China and
| | - Hanli Sun
- Institute of Biophysics, Chinese Academy of Science, 100101 Beijing, China
| | - Shen Huang
- From the Department of Biochemical Engineering/Institute for Biotransformation and Synthetic Biosystem, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, 100081 Beijing, China and
| | - Xudong Feng
- From the Department of Biochemical Engineering/Institute for Biotransformation and Synthetic Biosystem, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, 100081 Beijing, China and
| | - Tao Jiang
- Institute of Biophysics, Chinese Academy of Science, 100101 Beijing, China
| | - Chun Li
- From the Department of Biochemical Engineering/Institute for Biotransformation and Synthetic Biosystem, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, 100081 Beijing, China and
| |
Collapse
|
43
|
Cheng KW, Tseng CH, Yang CN, Tzeng CC, Cheng TC, Leu YL, Chuang YC, Wang JY, Lu YC, Chen YL, Cheng TL. Specific Inhibition of Bacterial β-Glucuronidase by Pyrazolo[4,3-c]quinoline Derivatives via a pH-Dependent Manner To Suppress Chemotherapy-Induced Intestinal Toxicity. J Med Chem 2017; 60:9222-9238. [PMID: 29120626 DOI: 10.1021/acs.jmedchem.7b00963] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Kai-Wen Cheng
- Institute of Biomedical
Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Chih-Hua Tseng
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Fragrance
and Cosmetic Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research
Center for Natural Products and Drug Development, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chia-Ning Yang
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung 811, Taiwan
| | - Cherng-Chyi Tzeng
- Research
Center for Natural Products and Drug Development, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department
of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ta-Chun Cheng
- Center
for Biomarkers and Biotech Drugs, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Lin Leu
- Department
of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City 717, Tainan
| | - Yu-Chung Chuang
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung 811, Taiwan
| | - Jaw-Yuan Wang
- Graduate Institute
of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of Gastroenterology and General Surgery, Department
of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yun-Chi Lu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yeh-Long Chen
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department
of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tian-Lu Cheng
- Institute of Biomedical
Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Center
for Biomarkers and Biotech Drugs, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biomedical and Environmental
Biology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| |
Collapse
|
44
|
Taha M, Ullah H, Al Muqarrabun LMR, Khan MN, Rahim F, Ahmat N, Ali M, Perveen S. Synthesis of bis-indolylmethanes as new potential inhibitors of β-glucuronidase and their molecular docking studies. Eur J Med Chem 2017; 143:1757-1767. [PMID: 29133042 DOI: 10.1016/j.ejmech.2017.10.071] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/01/2017] [Accepted: 10/26/2017] [Indexed: 10/18/2022]
Abstract
Thirty-two (32) bis-indolylmethane-hydrazone hybrids 1-32 were synthesized and characterized by 1HNMR, 13CNNMR and HREI-MS. All compounds were evaluated in vitro for β-glucuronidase inhibitory potential. All analogs showed varying degree of β-glucuronidase inhibitory potential ranging from 0.10 ± 0.01 to 48.50 ± 1.10 μM when compared with the standard drug d-saccharic acid-1,4-lactone (IC50 value 48.30 ± 1.20 μM). Derivatives 1-32 showed the highest β-glucuronidase inhibitory potentials which is many folds better than the standard drug d-saccharic acid-1,4-lactone. Further molecular docking study validated the experimental results. It was proposed that bis-indolylmethane may interact with some amino acid residues located within the active site of β-glucuronidase enzyme. This study has culminated in the identification of a new class of potent β-glucuronidase inhibitors.
Collapse
Affiliation(s)
- Muhammad Taha
- Department of clinical pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 31441, Dammam, Saudi Arabia.
| | - Hayat Ullah
- Department of Chemistry, Hazara University, Mansehra 21300, Pakistan
| | - Laode Muhammad Ramadhan Al Muqarrabun
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, 42300, Bandar Puncak Alam, Selangor, Malaysia; Faculty of Applied Science Universiti Teknologi MARA (UiTM), 40450, Shah Alam, Selangor, Malaysia
| | - Muhammad Naseem Khan
- Department of Chemistry, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra 21300, Pakistan
| | - Norizan Ahmat
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, 42300, Bandar Puncak Alam, Selangor, Malaysia; Faculty of Applied Science Universiti Teknologi MARA (UiTM), 40450, Shah Alam, Selangor, Malaysia
| | - Muhammad Ali
- Department of Chemistry, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| | - Shahnaz Perveen
- PCSIR Laboratories Complex, Shahrah-e-Dr. Salimuzzaman Siddiqui, Karachi 75280, Pakistan
| |
Collapse
|
45
|
Bano B, Arshia, Khan KM, Kanwal, Fatima B, Taha M, Ismail NH, Wadood A, Ghufran M, Perveen S. Synthesis, in vitro β -glucuronidase inhibitory potential and molecular docking studies of quinolines. Eur J Med Chem 2017; 139:849-864. [DOI: 10.1016/j.ejmech.2017.08.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 11/26/2022]
|
46
|
Khan SA, Peracha H, Ballhausen D, Wiesbauer A, Rohrbach M, Gautschi M, Mason RW, Giugliani R, Suzuki Y, Orii KE, Orii T, Tomatsu S. Epidemiology of mucopolysaccharidoses. Mol Genet Metab 2017; 121:227-240. [PMID: 28595941 PMCID: PMC5653283 DOI: 10.1016/j.ymgme.2017.05.016] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/22/2017] [Accepted: 05/22/2017] [Indexed: 12/15/2022]
Abstract
The aim of this study was to obtain data about the epidemiology of the different types of mucopolysaccharidoses in Japan and Switzerland and to compare with similar data from other countries. Data for Japan was collected between 1982 and 2009, and 467 cases with MPS were identified. The combined birth prevalence was 1.53 per 100,000 live births. The highest birth prevalence was 0.84 for MPS II, accounting for 55% of all MPS. MPS I, III, and IV accounted for 15, 16, and 10%, respectively. MPS VI and VII were more rare and accounted for 1.7 and 1.3%, respectively. A retrospective epidemiological data collection was performed in Switzerland between 1975 and 2008 (34years), and 41 living MPS patients were identified. The combined birth prevalence was 1.56 per 100,000 live births. The highest birth prevalence was 0.46 for MPS II, accounting for 29% of all MPS. MPS I, III, and IV accounted for 12, 24, and 24%, respectively. As seen in the Japanese population, MPS VI and VII were more rare and accounted for 7.3 and 2.4%, respectively. The high birth prevalence of MPS II in Japan was comparable to that seen in other East Asian countries where this MPS accounted for approximately 50% of all forms of MPS. Birth prevalence was also similar in some European countries (Germany, Northern Ireland, Portugal and the Netherlands) although the prevalence of other forms of MPS is also reported to be higher in these countries. Birth prevalence of MPS II in Switzerland and other European countries is comparatively lower. The birth prevalence of MPS III and IV in Switzerland is higher than in Japan but comparable to that in most other European countries. Moreover, the birth prevalence of MPS VI and VII was very low in both, Switzerland and Japan. Overall, the frequency of MPS varies for each population due to differences in ethnic backgrounds and/or founder effects that affect the birth prevalence of each type of MPS, as seen for other rare genetic diseases. Methods for identification of MPS patients are not uniform across all countries, and consequently, if patients are not identified, recorded prevalence rates will be aberrantly low.
Collapse
Affiliation(s)
- Shaukat A Khan
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Hira Peracha
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Diana Ballhausen
- Centre for Molecular Diseases, Service for Genetic Medicine, University Hospital Lausanne, Switzerland
| | - Alfred Wiesbauer
- Institute of Social and Preventive Medicine, University of Bern, Switzerland
| | - Marianne Rohrbach
- Division of Metabolism and Children's Research Centre (CRC), University Children's Hospital, Zurich, Switzerland
| | - Matthias Gautschi
- Division of Endocrinology, Diabetology and Metabolism, University Children's Hospital, University Institute of Clinical Chemistry, Inselspital, University of Bern, Bern, Switzerland
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Roberto Giugliani
- Medical Genetics Service, HCPA, Dep. Genetics, UFRGS, and INAGEMP, Porto Alegre, Brazil
| | | | - Kenji E Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Tadao Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Pediatrics, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
47
|
Wu L, Jiang J, Jin Y, Kallemeijn WW, Kuo CL, Artola M, Dai W, van Elk C, van Eijk M, van der Marel GA, Codée JDC, Florea BI, Aerts JMFG, Overkleeft HS, Davies GJ. Activity-based probes for functional interrogation of retaining β-glucuronidases. Nat Chem Biol 2017; 13:867-873. [PMID: 28581485 DOI: 10.1038/nchembio.2395] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/10/2017] [Indexed: 02/06/2023]
Abstract
Humans express at least two distinct β-glucuronidase enzymes that are involved in disease: exo-acting β-glucuronidase (GUSB), whose deficiency gives rise to mucopolysaccharidosis type VII, and endo-acting heparanase (HPSE), whose overexpression is implicated in inflammation and cancers. The medical importance of these enzymes necessitates reliable methods to assay their activities in tissues. Herein, we present a set of β-glucuronidase-specific activity-based probes (ABPs) that allow rapid and quantitative visualization of GUSB and HPSE in biological samples, providing a powerful tool for dissecting their activities in normal and disease states. Unexpectedly, we find that the supposedly inactive HPSE proenzyme proHPSE is also labeled by our ABPs, leading to surprising insights regarding structural relationships between proHPSE, mature HPSE, and their bacterial homologs. Our results demonstrate the application of β-glucuronidase ABPs in tracking pathologically relevant enzymes and provide a case study of how ABP-driven approaches can lead to discovery of unanticipated structural and biochemical functionality.
Collapse
Affiliation(s)
- Liang Wu
- York Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York, UK
| | - Jianbing Jiang
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Yi Jin
- York Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York, UK
| | - Wouter W Kallemeijn
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Chi-Lin Kuo
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Marta Artola
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Wei Dai
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Cas van Elk
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Marco van Eijk
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Gijsbert A van der Marel
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Jeroen D C Codée
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Bogdan I Florea
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Johannes M F G Aerts
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Herman S Overkleeft
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Gideon J Davies
- York Structural Biology Laboratory, Department of Chemistry, University of York, Heslington, York, UK
| |
Collapse
|
48
|
Dickson A, Bailey CT, Karanicolas J. Optimal allosteric stabilization sites using contact stabilization analysis. J Comput Chem 2017; 38:1138-1146. [PMID: 27774625 PMCID: PMC5403592 DOI: 10.1002/jcc.24517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/30/2016] [Accepted: 10/01/2016] [Indexed: 11/08/2022]
Abstract
Proteins can be destabilized by a number of environmental factors such as temperature, pH, and mutation. The ability to subsequently restore function under these conditions by adding small molecule stabilizers, or by introducing disulfide bonds, would be a very powerful tool, but the physical principles that drive this stabilization are not well understood. The first problem lies is in choosing an appropriate binding site or disulfide bond location to best confer stability to the active site and restore function. Here, we present a general framework for predicting which allosteric binding sites correlate with stability in the active site. Using the Karanicolas-Brooks Gō-like model, we examine the dynamics of the enzyme β-glucuronidase using an Umbrella Sampling method to thoroughly sample the conformational landscape. Each intramolecular contact is assigned a score termed a "stabilization factor" that measures its correlation with structural changes in the active site. We have carried out this analysis for three different scaling strengths for the intramolecular contacts, and we examine how the calculated stabilization factors depend on the ensemble of destabilized conformations. We further examine a locally destabilized mutant of β-glucuronidase that has been characterized experimentally, and show that this brings about local changes in the stabilization factors. We find that the proximity to the active site is not sufficient to determine which contacts can confer active site stability. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alex Dickson
- Department of Biochemistry & Molecular Biology and the Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, Michigan, 48824
| | - Christopher T Bailey
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan, 48824
| | - John Karanicolas
- Department of Molecular Biosciences and Center for Computational Biology, University of Kansas, Lawrence, Kansas, 66045
| |
Collapse
|
49
|
Baharudin MS, Taha M, Imran S, Ismail NH, Rahim F, Javid MT, Khan KM, Ali M. Synthesis of indole analogs as potent β-glucuronidase inhibitors. Bioorg Chem 2017; 72:323-332. [DOI: 10.1016/j.bioorg.2017.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/15/2017] [Accepted: 05/01/2017] [Indexed: 10/19/2022]
|
50
|
Pellock SJ, Redinbo MR. Glucuronides in the gut: Sugar-driven symbioses between microbe and host. J Biol Chem 2017; 292:8569-8576. [PMID: 28389557 DOI: 10.1074/jbc.r116.767434] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The intestinal milieu is astonishingly complex and home to a constantly changing mixture of small and large molecules, along with an abundance of bacteria, viral particles, and eukaryotic cells. Such complexity makes it difficult to develop testable molecular hypotheses regarding host-microbe interactions. Fortunately, mammals and their associated gastrointestinal (GI) microbes contain complementary systems that are ideally suited for mechanistic studies. Mammalian systems inactivate endobiotic and xenobiotic compounds by linking them to a glucuronic acid sugar for GI excretion. In the GI tract, the microbiota express β-glucuronidase enzymes that remove the glucuronic acid as a carbon source, effectively reversing the actions of mammalian inactivation. Thus, by probing the actions of microbial β-glucuronidases, and by understanding which substrate glucuronides they process, molecular insights into mammalian-microbial symbioses may be revealed amid the complexity of the intestinal tract. Here, we focus on glucuronides in the gut and the microbial proteins that process them.
Collapse
Affiliation(s)
- Samuel J Pellock
- From the Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-3290
| | - Matthew R Redinbo
- From the Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-3290
| |
Collapse
|