1
|
Artola M, Aerts JMFG, van der Marel GA, Rovira C, Codée JDC, Davies GJ, Overkleeft HS. From Mechanism-Based Retaining Glycosidase Inhibitors to Activity-Based Glycosidase Profiling. J Am Chem Soc 2024; 146:24729-24741. [PMID: 39213505 PMCID: PMC11403624 DOI: 10.1021/jacs.4c08840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Activity-based protein profiling (ABPP) is an effective technology for the identification and functional annotation of enzymes in complex biological samples. ABP designs are normally directed to an enzyme active site nucleophile, and within the field of Carbohydrate-Active Enzymes (CAZymes), ABPP has been most successful for those enzymes that feature such a residue: retaining glycosidases (GHs). Several mechanism-based covalent and irreversible retaining GH inhibitors have emerged over the past sixty years. ABP designs based on these inhibitor chemistries appeared since the turn of the millennium, and we contributed to the field by designing a suite of retaining GH ABPs modeled on the structure and mode of action of the natural product, cyclophellitol. These ABPs enable the study of both exo- and endo-acting retaining GHs in human health and disease, for instance in genetic metabolic disorders in which retaining GHs are deficient. They are also finding increasing use in the study of GHs in gut microbiota and environmental microorganisms, both in the context of drug (de)toxification in the gut and that of biomass polysaccharide processing for future sustainable energy and chemistries. This account comprises the authors' view on the history of mechanism-based retaining GH inhibitor design and discovery, on how these inhibitors served as blueprints for retaining GH ABP design, and on some current and future developments on how cyclophellitol-based ABPs may drive the discovery of retaining GHs and their inhibitors.
Collapse
Affiliation(s)
- Marta Artola
- Leiden Institute of Chemistry, Leiden University, 2300 RA, Leiden, The Netherlands
| | - Johannes M F G Aerts
- Leiden Institute of Chemistry, Leiden University, 2300 RA, Leiden, The Netherlands
| | | | - Carme Rovira
- Departament de Química Inorgànica I Orgànica & IQTCUB, Universitat de Barcelona, Barcelona 08028, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08020, Spain
| | - Jeroen D C Codée
- Leiden Institute of Chemistry, Leiden University, 2300 RA, Leiden, The Netherlands
| | - Gideon J Davies
- Department of Chemistry, The University York, Heslington, York YO10 5DD, United Kingdom
| | - Herman S Overkleeft
- Leiden Institute of Chemistry, Leiden University, 2300 RA, Leiden, The Netherlands
| |
Collapse
|
2
|
Jiang J, Czuchry D, Ru Y, Peng H, Shen J, Wang T, Zhao W, Chen W, Sui SF, Li Y, Li N. Activity-based metaproteomics driven discovery and enzymological characterization of potential α-galactosidases in the mouse gut microbiome. Commun Chem 2024; 7:184. [PMID: 39152233 PMCID: PMC11329505 DOI: 10.1038/s42004-024-01273-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024] Open
Abstract
The gut microbiota offers an extensive resource of enzymes, but many remain uncharacterized. To distinguish the activities of similar annotated proteins and mine the potentially applicable ones in the microbiome, we applied an effective Activity-Based Metaproteomics (ABMP) strategy using a specific activity-based probe (ABP) to screen the entire gut microbiome for directly discovering active enzymes and their potential applications, not for exploring host-microbiome interactions. By using an activity-based cyclophellitol aziridine probe specific to α-galactosidases (AGAL), we successfully identified and characterized several gut microbiota enzymes possessing AGAL activities. Cryo-electron microscopy analysis of a newly characterized enzyme (AGLA5) revealed the covalent binding conformations between the AGAL5 active site and the cyclophellitol aziridine ABP, which could provide insights into the enzyme's catalytic mechanism. The four newly characterized AGALs have diverse potential activities, including raffinose family oligosaccharides (RFOs) hydrolysis and enzymatic blood group transformation. Collectively, we present a ABMP platform that facilitates gut microbiota AGALs discovery, biochemical activity annotations and potential industrial or biopharmaceutical applications.
Collapse
Affiliation(s)
- Jianbing Jiang
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Diana Czuchry
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yanxia Ru
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Huipai Peng
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Junfeng Shen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Teng Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Wenjuan Zhao
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Weihua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Sen-Fang Sui
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Cryo-EM Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yaowang Li
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| | - Nan Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Shenzhen Key Laboratory of Genome Manipulation and Biosynthesis, Shenzhen, China.
| |
Collapse
|
3
|
Gilormini PA, Thota VN, Fers-Lidou A, Ashmus RA, Nodwell M, Brockerman J, Kuo CW, Wang Y, Gray TE, Nitin, McDonagh AW, Guu SY, Ertunc N, Yeo D, Zandberg WF, Khoo KH, Britton R, Vocadlo DJ. A metabolic inhibitor blocks cellular fucosylation and enables production of afucosylated antibodies. Proc Natl Acad Sci U S A 2024; 121:e2314026121. [PMID: 38917011 PMCID: PMC11228515 DOI: 10.1073/pnas.2314026121] [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: 08/15/2023] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
The fucosylation of glycoproteins regulates diverse physiological processes. Inhibitors that can control cellular levels of protein fucosylation have consequently emerged as being of high interest. One area where inhibitors of fucosylation have gained significant attention is in the production of afucosylated antibodies, which exhibit superior antibody-dependent cell cytotoxicity as compared to their fucosylated counterparts. Here, we describe β-carbafucose, a fucose derivative in which the endocyclic ring oxygen is replaced by a methylene group, and show that it acts as a potent metabolic inhibitor within cells to antagonize protein fucosylation. β-carbafucose is assimilated by the fucose salvage pathway to form GDP-carbafucose which, due to its being unable to form the oxocarbenium ion-like transition states used by fucosyltransferases, is an incompetent substrate for these enzymes. β-carbafucose treatment of a CHO cell line used for high-level production of the therapeutic antibody Herceptin leads to dose-dependent reductions in core fucosylation without affecting cell growth or antibody production. Mass spectrometry analyses of the intact antibody and N-glycans show that β-carbafucose is not incorporated into the antibody N-glycans at detectable levels. We expect that β-carbafucose will serve as a useful research tool for the community and may find immediate application for the rapid production of afucosylated antibodies for therapeutic purposes.
Collapse
Affiliation(s)
| | | | - Anthony Fers-Lidou
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Roger A Ashmus
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Matthew Nodwell
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Jacob Brockerman
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Chu-Wei Kuo
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Yang Wang
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Taylor E Gray
- Department of Chemistry, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| | - Nitin
- Department of Chemistry, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| | - Anthony W McDonagh
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Shih-Yun Guu
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Nursah Ertunc
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | | | - Wesley F Zandberg
- Department of Chemistry, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Robert Britton
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - David J Vocadlo
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
4
|
Christin O, Roulland E. Advancements in Enacyloxins Total Synthesis: Access to the Chlorinated Polyunsaturated Chain Peculiar to this Promising Family of Antibiotics. Org Lett 2023; 25:6869-6874. [PMID: 37676860 DOI: 10.1021/acs.orglett.3c02477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
The first synthesis of the protected chain specific to the enacyloxin antibiotic family is reported. The noticeable features are (a) the construction of the chlorinated undecapentaenoic moiety implementing the sequence Tsuji's alkyne syn allyl-chlorination, E-selective Pd/Cu-catalyzed allene-alkyne coupling, Horner-Wadsworth-Emmons olefination, dehydration; (b) control of the C18 chlorinated stereogenic center by organo-catalyzed aldehyde α-chlorination; and (c) the assemblage of this aldehyde with the C1-C16 ketone using a highly diastereoselective Mukaiyama aldol.
Collapse
Affiliation(s)
- Orane Christin
- CiTCoM, UMR 8038, CNRS-Université Paris Cité, Faculté de Pharmacie, 4, avenue de l'Observatoire, 75006 Paris, France
| | - Emmanuel Roulland
- CiTCoM, UMR 8038, CNRS-Université Paris Cité, Faculté de Pharmacie, 4, avenue de l'Observatoire, 75006 Paris, France
| |
Collapse
|
5
|
Vinaykumar A, Surender B, Rao BV. Chemoselective Nozaki-Hiyama-Takai-Kishi and Grignard reaction: short synthesis of some carbahexopyranoses. RSC Adv 2023; 13:22824-22830. [PMID: 37520087 PMCID: PMC10375257 DOI: 10.1039/d3ra03704e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/20/2023] [Indexed: 08/01/2023] Open
Abstract
A common, divergent, efficient, stereoselective and short approach for the total syntheses of some carbahexopyranoses namely, MK7607, (-)-gabosine A, (-)-conduritol E, (-)-conduritol F, 6a-carba-β-d-fructopyranose and other carbasugars using chemoselective Grignard or Nozaki-Hiyama-Takai-Kishi (NHTK) reactions and RCM. Herein, the Grignard and NHTK reactions are able to differentiate the reactivity difference between lactol or lactolacetate and aldehyde of 2 & 6 under given conditions to give the desired skeleton chemoselectivity.
Collapse
Affiliation(s)
- Allam Vinaykumar
- Fluoro-Agrochemicals, CSIR-Indian Institute of Chemical Technology Hyderabad India
| | - Banothu Surender
- Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology Hyderabad India
| | - Batchu Venkateswara Rao
- Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology Hyderabad India
| |
Collapse
|
6
|
Hikawa R, Shimogaki M, Kano T. Construction of three contiguous stereocenters through amine-catalyzed asymmetric aldol reactions. Chem Commun (Camb) 2023. [PMID: 37334826 DOI: 10.1039/d3cc01606d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Three contiguous stereocenters were constructed by an amino acid-catalyzed asymmetric aldol reaction of α-siloxyketones with racemizable α-haloaldehydes via dynamic kinetic resolution. One-pot catalytic asymmetric synthesis of the highly functionalized products could also be accomplished by the α-bromination of simple aldehydes and the subsequent asymmetric aldol reaction.
Collapse
Affiliation(s)
- Ryoga Hikawa
- Department of Applied Chemistry, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan.
| | - Mio Shimogaki
- Graduate School of Science, University of Hyogo, Hyogo 678-1297, Japan
| | - Taichi Kano
- Department of Applied Chemistry, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan.
| |
Collapse
|
7
|
Grayfer T, Yamani K, Jung E, Chesnokov GA, Ferrara I, Hsiao CC, Georgiou A, Michel J, Bailly A, Sieber S, Eberl L, Gademann K. Allylic Carbocyclic Inhibitors Covalently Bind Glycoside Hydrolases. JACS AU 2023; 3:1151-1161. [PMID: 37124289 PMCID: PMC10131216 DOI: 10.1021/jacsau.3c00037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 05/03/2023]
Abstract
Allylic cyclitols were investigated as covalent inhibitors of glycoside hydrolases by chemical, enzymatic, proteomic, and computational methods. This approach was inspired by the C7 cyclitol natural product streptol glucoside, which features a potential carbohydrate leaving group in the 4-position (carbohydrate numbering). To test this hypothesis, carbocyclic inhibitors with leaving groups in the 4- and 6- positions were prepared. The results of enzyme kinetics analyses demonstrated that dinitrophenyl ethers covalently inhibit α-glucosidases of the GH13 family without reactivation. The labeled enzyme was studied by proteomics, and the active site residue Asp214 was identified as modified. Additionally, computational studies, including enzyme homology modeling and density functional theory (DFT) calculations, further delineate the electronic and structural requirements for activity. This study demonstrates that previously unexplored 4- and 6-positions can be exploited for successful inhibitor design.
Collapse
Affiliation(s)
- Tatyana
D. Grayfer
- Department
of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Khalil Yamani
- Department
of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Erik Jung
- Department
of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Gleb A. Chesnokov
- Department
of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Isabella Ferrara
- Department
of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Chien-Chi Hsiao
- Department
of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Antri Georgiou
- Department
of Plant and Microbial Biology, University
of Zurich, Zollikerstrasse
107, 8008 Zürich, Switzerland
| | - Jeremy Michel
- Department
of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Aurélien Bailly
- Department
of Plant and Microbial Biology, University
of Zurich, Zollikerstrasse
107, 8008 Zürich, Switzerland
| | - Simon Sieber
- Department
of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Leo Eberl
- Department
of Plant and Microbial Biology, University
of Zurich, Zollikerstrasse
107, 8008 Zürich, Switzerland
| | - Karl Gademann
- Department
of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| |
Collapse
|
8
|
Chuankhayan P, Lee RH, Guan HH, Lin CC, Chen NC, Huang YC, Yoshimura M, Nakagawa A, Chen CJ. Structural insight into the hydrolase and synthase activities of an alkaline α-galactosidase from Arabidopsis from complexes with substrate/product. Acta Crystallogr D Struct Biol 2023; 79:154-167. [PMID: 36762861 PMCID: PMC9912918 DOI: 10.1107/s2059798323000037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/03/2023] [Indexed: 01/21/2023] Open
Abstract
The alkaline α-galactosidase AtAkαGal3 from Arabidopsis thaliana catalyzes the hydrolysis of α-D-galactose from galacto-oligosaccharides under alkaline conditions. A phylogenetic analysis based on sequence alignment classifies AtAkαGal3 as more closely related to the raffinose family of oligosaccharide (RFO) synthases than to the acidic α-galactosidases. Here, thin-layer chromatography is used to demonstrate that AtAkαGal3 exhibits a dual function and is capable of synthesizing stachyose using raffinose, instead of galactinol, as the galactose donor. Crystal structures of complexes of AtAkαGal3 and its D383A mutant with various substrates and products, including galactose, galactinol, raffinose, stachyose and sucrose, are reported as the first representative structures of an alkaline α-galactosidase. The structure of AtAkαGal3 comprises three domains: an N-terminal domain with 13 antiparallel β-strands, a catalytic domain with an (α/β)8-barrel fold and a C-terminal domain composed of β-sheets that form two Greek-key motifs. The WW box of the N-terminal domain, which comprises the conserved residues FRSK75XW77W78 in the RFO synthases, contributes Trp77 and Trp78 to the +1 subsite to contribute to the substrate-binding ability together with the (α/β)8 barrel of the catalytic domain. The C-terminal domain is presumably involved in structural stability. Structures of the D383A mutant in complex with various substrates and products, especially the natural substrate/product stachyose, reveal four complete subsites (-1 to +3) at the catalytic site. A functional loop (residues 329-352) that exists in the alkaline α-galactosidase AtAkαGal3 and possibly in RFO synthases, but not in acidic α-galactosidases, stabilizes the stachyose at the +2 and +3 subsites and extends the catalytic pocket for the transferase mechanism. Considering the similarities in amino-acid sequence, catalytic domain and activity between alkaline α-galactosidases and RFO synthases, the structure of AtAkαGal3 might also serve a model for the study of RFO synthases, structures of which are lacking.
Collapse
Affiliation(s)
- Phimonphan Chuankhayan
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Cente, Hsinchu 30076, Taiwan
| | - Ruey-Hua Lee
- Institute of Tropical Plant Sciences and Microbiology, National Cheng Kung University, Tainan City 701, Taiwan
| | - Hong-Hsiang Guan
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Cente, Hsinchu 30076, Taiwan
| | - Chein-Chih Lin
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Cente, Hsinchu 30076, Taiwan
| | - Nai-Chi Chen
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Cente, Hsinchu 30076, Taiwan
| | - Yen-Chieh Huang
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Cente, Hsinchu 30076, Taiwan
| | - Masato Yoshimura
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Cente, Hsinchu 30076, Taiwan
| | - Atsushi Nakagawa
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Chun-Jung Chen
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Cente, Hsinchu 30076, Taiwan,Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan City 701, Taiwan,Department of Physics, National Tsing Hua University, Hsinchu 30013, Taiwan,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan,Correspondence e-mail:
| |
Collapse
|
9
|
Vilen Z, Reeves AE, Huang ML. (Glycan Binding) Activity‐Based Protein Profiling in Cells Enabled by Mass Spectrometry‐Based Proteomics. Isr J Chem 2023; 63. [PMID: 37131487 PMCID: PMC10150848 DOI: 10.1002/ijch.202200097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The presence of glycan modifications at the cell surface and other locales positions them as key regulators of cell recognition and function. However, due to the complexity of glycosylation, the annotation of which proteins bear glycan modifications, which glycan patterns are present, and which proteins are capable of binding glycans is incomplete. Inspired by activity-based protein profiling to enrich for proteins in cells based on select characteristics, these endeavors have been greatly advanced by the development of appropriate glycan-binding and glycan-based probes. Here, we provide context for these three problems and describe how the capability of molecules to interact with glycans has enabled the assignment of proteins with specific glycan modifications or of proteins that bind glycans. Furthermore, we discuss how the integration of these probes with high resolution mass spectrometry-based technologies has greatly advanced glycoscience.
Collapse
Affiliation(s)
- Zak Vilen
- Skaggs Graduate School of Chemical and Biological Sciences Scripps Research 10550 N. Torrey Pines Rd. La Jolla CA 92037 USA
- Department of Molecular Medicine Scripps Research 10550 N. Torrey Pines Rd. La Jolla CA 92037, USA
| | - Abigail E. Reeves
- Skaggs Graduate School of Chemical and Biological Sciences Scripps Research 10550 N. Torrey Pines Rd. La Jolla CA 92037 USA
- Department of Molecular Medicine Scripps Research 10550 N. Torrey Pines Rd. La Jolla CA 92037, USA
| | - Mia L. Huang
- Skaggs Graduate School of Chemical and Biological Sciences Scripps Research 10550 N. Torrey Pines Rd. La Jolla CA 92037 USA
- Department of Molecular Medicine Scripps Research 10550 N. Torrey Pines Rd. La Jolla CA 92037, USA
| |
Collapse
|
10
|
Akintola O, Farren-Dai M, Ren W, Bhosale S, Britton R, Świderek K, Moliner V, Bennet AJ. Glycoside Hydrolase Catalysis: Do Substrates and Mechanism-Based Covalent Inhibitors React via Matching Transition States? ACS Catal 2022. [DOI: 10.1021/acscatal.2c04027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Oluwafemi Akintola
- Department of Chemistry, Simon Fraser University, Burnaby, British ColumbiaV5A 1S6, Canada
| | - Marco Farren-Dai
- Department of Chemistry, Simon Fraser University, Burnaby, British ColumbiaV5A 1S6, Canada
| | - Weiwu Ren
- Department of Chemistry, Simon Fraser University, Burnaby, British ColumbiaV5A 1S6, Canada
| | - Sandeep Bhosale
- Department of Chemistry, Simon Fraser University, Burnaby, British ColumbiaV5A 1S6, Canada
| | - Robert Britton
- Department of Chemistry, Simon Fraser University, Burnaby, British ColumbiaV5A 1S6, Canada
| | - Katarzyna Świderek
- BioComp Group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071Castellón, Spain
| | - Vicent Moliner
- BioComp Group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071Castellón, Spain
| | - Andrew J. Bennet
- Department of Chemistry, Simon Fraser University, Burnaby, British ColumbiaV5A 1S6, Canada
| |
Collapse
|
11
|
Riester O, Burkhardtsmaier P, Gurung Y, Laufer S, Deigner HP, Schmidt MS. Synergy of R-(-)carvone and cyclohexenone-based carbasugar precursors with antibiotics to enhance antibiotic potency and inhibit biofilm formation. Sci Rep 2022; 12:18019. [PMID: 36289389 PMCID: PMC9606123 DOI: 10.1038/s41598-022-22807-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/19/2022] [Indexed: 01/24/2023] Open
Abstract
The widespread use of antibiotics in recent decades has been a major factor in the emergence of antibiotic resistances. Antibiotic-resistant pathogens pose increasing challenges to healthcare systems in both developing and developed countries. To counteract this, the development of new antibiotics or adjuvants to combat existing resistance to antibiotics is crucial. Glycomimetics, for example carbasugars, offer high potential as adjuvants, as they can inhibit metabolic pathways or biofilm formation due to their similarity to natural substrates. Here, we demonstrate the synthesis of carbasugar precursors (CSPs) and their application as biofilm inhibitors for E. coli and MRSA, as well as their synergistic effect in combination with antibiotics to circumvent biofilm-induced antibiotic resistances. This results in a biofilm reduction of up to 70% for the CSP rac-7 and a reduction in bacterial viability of MRSA by approximately 45% when combined with the otherwise ineffective antibiotic mixture of penicillin and streptomycin.
Collapse
Affiliation(s)
- Oliver Riester
- grid.21051.370000 0001 0601 6589Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Strasse 17, 78054 Villingen-Schwenningen, Germany ,grid.10392.390000 0001 2190 1447Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard-Karls-University Tuebingen, Auf Der Morgenstelle 8, 72076 Tübingen, Germany
| | - Pia Burkhardtsmaier
- grid.21051.370000 0001 0601 6589Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Strasse 17, 78054 Villingen-Schwenningen, Germany
| | - Yuna Gurung
- grid.21051.370000 0001 0601 6589Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Strasse 17, 78054 Villingen-Schwenningen, Germany
| | - Stefan Laufer
- grid.10392.390000 0001 2190 1447Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard-Karls-University Tuebingen, Auf Der Morgenstelle 8, 72076 Tübingen, Germany ,Tuebingen Center for Academic Drug Discovery and Development (TüCAD2), 72076 Tübingen, Germany
| | - Hans-Peter Deigner
- grid.21051.370000 0001 0601 6589Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Strasse 17, 78054 Villingen-Schwenningen, Germany ,grid.10392.390000 0001 2190 1447Faculty of Science, Eberhard-Karls-University Tuebingen, Auf Der Morgenstelle 8, 72076 Tübingen, Germany ,grid.418008.50000 0004 0494 3022EXIM Department, Fraunhofer Institute IZI (Leipzig), Schillingallee 68, 18057 Rostock, Germany
| | - Magnus S. Schmidt
- grid.21051.370000 0001 0601 6589Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Strasse 17, 78054 Villingen-Schwenningen, Germany
| |
Collapse
|
12
|
Thanvi R, Jayasinghe TD, Kapil S, Obadawo BS, Ronning DR, Sucheck SJ. Synthesis of C7/C8-cyclitols and C7N-aminocyclitols from maltose and X-ray crystal structure of Streptomyces coelicolor GlgEI V279S in a complex with an amylostatin GXG–like derivative. Front Chem 2022; 10:950433. [PMID: 36157042 PMCID: PMC9501709 DOI: 10.3389/fchem.2022.950433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/26/2022] [Indexed: 11/30/2022] Open
Abstract
C7/C8-cyclitols and C7N-aminocyclitols find applications in the pharmaceutical sector as α-glucosidase inhibitors and in the agricultural sector as fungicides and insecticides. In this study, we identified C7/C8-cyclitols and C7N-aminocyclitols as potential inhibitors of Streptomyces coelicolor (Sco) GlgEI-V279S based on the docking scores. The protein and the ligand (targets 11, 12, and 13) were prepared, the states were generated at pH 7.0 ± 2.0, and the ligands were docked into the active sites of the receptor via Glide™. The synthetic route to these targets was similar to our previously reported route used to obtain 4-⍺-glucoside of valienamine (AGV), except the protecting group for target 12 was a p-bromobenzyl (PBB) ether to preserve the alkene upon deprotection. While compounds 11–13 did not inhibit Sco GlgEI-V279S at the concentrations evaluated, an X-ray crystal structure of the Sco GlgE1-V279S/13 complex was solved to a resolution of 2.73 Å. This structure allowed assessment differences and commonality with our previously reported inhibitors and was useful for identifying enzyme–compound interactions that may be important for future inhibitor development. The Asp 394 nucleophile formed a bidentate hydrogen bond interaction with the exocyclic oxygen atoms (C(3)-OH and C(7)-OH) similar to the observed interactions with the Sco GlgEI-V279S in a complex with AGV (PDB:7MGY). In addition, the data suggest replacing the cyclohexyl group with more isosteric and hydrogen bond–donating groups to increase binding interactions in the + 1 binding site.
Collapse
Affiliation(s)
- Radhika Thanvi
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH, United States
| | - Thilina D. Jayasinghe
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sunayana Kapil
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH, United States
| | | | - Donald R. Ronning
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Donald R. Ronning, ; Steven J. Sucheck,
| | - Steven J. Sucheck
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH, United States
- *Correspondence: Donald R. Ronning, ; Steven J. Sucheck,
| |
Collapse
|
13
|
Miyazaki T, Ikegaya M, Alonso-Gil S. Structural and mechanistic insights into the substrate specificity and hydrolysis of GH31 α-N-acetylgalactosaminidase. Biochimie 2021; 195:90-99. [PMID: 34826537 DOI: 10.1016/j.biochi.2021.11.007] [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: 09/08/2021] [Revised: 11/15/2021] [Accepted: 11/22/2021] [Indexed: 11/02/2022]
Abstract
Glycoside hydrolase family 31 (GH31) is a diversified family of anomer-retaining α-glycoside hydrolases, such as α-glucosidase and α-xylosidase, among others. Recently, GH31 α-N-acetylgalactosaminidases (Nag31s) have been identified to hydrolyze the core of mucin-type O-glycans and the crystal structure of a gut bacterium Enterococcus faecalis Nag31 has been reported. However, the mechanisms of substrate specificity and hydrolysis of Nag31s are not well investigated. Herein, we show that E. faecalis Nag31 has the ability to release N-acetylgalactosamine (GalNAc) from O-glycoproteins, such as fetuin and mucin, but has low activity against Tn antigen. Mutational analysis and crystal structures of the Michaelis complexes reveal that residues of the active site work in concert with their conformational changes to act on only α-N-acetylgalactosaminides. Docking simulations using GalNAc-attached peptides suggest that the enzyme mainly recognizes GalNAc and side chains of Ser/Thr, but not strictly other peptide residues. Moreover, quantum mechanics calculations indicate that the enzyme preferred p-nitrophenyl α-N-acetylgalactosaminide to Tn antigen and that the hydrolysis progresses through a conformational itinerary, 4C1 → 1S3 → 4C1, in GalNAc of substrates. Our results provide novel insights into the diversification of the sugar recognition and hydrolytic mechanisms of GH31 enzymes.
Collapse
Affiliation(s)
- Takatsugu Miyazaki
- Research Institute of Green Science and Technology, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529, Japan; Department of Bioscience, Graduate School of Science and Technology, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529, Japan.
| | - Marina Ikegaya
- Department of Bioscience, Graduate School of Science and Technology, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529, Japan
| | - Santiago Alonso-Gil
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria.
| |
Collapse
|
14
|
Nejatie A, Steves E, Gauthier N, Baker J, Nesbitt J, McMahon SA, Oehler V, Thornton NJ, Noyovitz B, Khazaei K, Byers BW, Zandberg WF, Gloster TM, Moore MM, Bennet AJ. Kinetic and Structural Characterization of Sialidases (Kdnases) from Ascomycete Fungal Pathogens. ACS Chem Biol 2021; 16:2632-2640. [PMID: 34724608 DOI: 10.1021/acschembio.1c00666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sialidases catalyze the release of sialic acid from the terminus of glycan chains. We previously characterized the sialidase from the opportunistic fungal pathogen, Aspergillus fumigatus, and showed that it is a Kdnase. That is, this enzyme prefers 3-deoxy-d-glycero-d-galacto-non-2-ulosonates (Kdn glycosides) as the substrate compared to N-acetylneuraminides (Neu5Ac). Here, we report characterization and crystal structures of putative sialidases from two other ascomycete fungal pathogens, Aspergillus terreus (AtS) and Trichophyton rubrum (TrS). Unlike A. fumigatus Kdnase (AfS), hydrolysis with the Neu5Ac substrates was negligible for TrS and AtS; thus, TrS and AtS are selective Kdnases. The second-order rate constant for hydrolysis of aryl Kdn glycosides by AtS is similar to that by AfS but 30-fold higher by TrS. The structures of these glycoside hydrolase family 33 (GH33) enzymes in complex with a range of ligands for both AtS and TrS show subtle changes in ring conformation that mimic the Michaelis complex, transition state, and covalent intermediate formed during catalysis. In addition, they can aid identification of important residues for distinguishing between Kdn and Neu5Ac substrates. When A. fumigatus, A. terreus, and T. rubrum were grown in chemically defined media, Kdn was detected in mycelial extracts, but Neu5Ac was only observed in A. terreus or T. rubrum extracts. The C8 monosaccharide 3-deoxy-d-manno-oct-2-ulosonic acid (Kdo) was also identified in A. fumigatus and T. rubrum samples. A fluorescent Kdn probe was synthesized and revealed the localization of AfS in vesicles at the cell surface.
Collapse
Affiliation(s)
- Ali Nejatie
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, British
Columbia, Canada
| | - Elizabeth Steves
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, British
Columbia, Canada
| | - Nick Gauthier
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, British
Columbia, Canada
| | - Jamie Baker
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, British
Columbia, Canada
| | - Jason Nesbitt
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, British
Columbia, Canada
| | - Stephen A. McMahon
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews KY16 9ST, Fife, U.K
| | - Verena Oehler
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews KY16 9ST, Fife, U.K
| | - Nicholas J. Thornton
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews KY16 9ST, Fife, U.K
| | - Benjamin Noyovitz
- Department of Chemistry, I. K. Barber Faculty of Science, University of British Columbia, 3247 University Way, Kelowna V1V 1V7, British Columbia, Canada
| | - Kobra Khazaei
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, British
Columbia, Canada
| | - Brock W. Byers
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, British
Columbia, Canada
| | - Wesley F. Zandberg
- Department of Chemistry, I. K. Barber Faculty of Science, University of British Columbia, 3247 University Way, Kelowna V1V 1V7, British Columbia, Canada
| | - Tracey M. Gloster
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews KY16 9ST, Fife, U.K
| | - Margo M. Moore
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, British
Columbia, Canada
| | - Andrew J. Bennet
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby V5A 1S6, British
Columbia, Canada
| |
Collapse
|
15
|
Jain N, Tamura K, Déjean G, Van Petegem F, Brumer H. Orthogonal Active-Site Labels for Mixed-Linkage endo-β-Glucanases. ACS Chem Biol 2021; 16:1968-1984. [PMID: 33988963 DOI: 10.1021/acschembio.1c00063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Small molecule irreversible inhibitors are valuable tools for determining catalytically important active-site residues and revealing key details of the specificity, structure, and function of glycoside hydrolases (GHs). β-glucans that contain backbone β(1,3) linkages are widespread in nature, e.g., mixed-linkage β(1,3)/β(1,4)-glucans in the cell walls of higher plants and β(1,3)glucans in yeasts and algae. Commensurate with this ubiquity, a large diversity of mixed-linkage endoglucanases (MLGases, EC 3.2.1.73) and endo-β(1,3)-glucanases (laminarinases, EC 3.2.1.39 and EC 3.2.1.6) have evolved to specifically hydrolyze these polysaccharides, respectively, in environmental niches including the human gut. To facilitate biochemical and structural analysis of these GHs, with a focus on MLGases, we present here the facile chemo-enzymatic synthesis of a library of active-site-directed enzyme inhibitors based on mixed-linkage oligosaccharide scaffolds and N-bromoacetylglycosylamine or 2-fluoro-2-deoxyglycoside warheads. The effectiveness and irreversibility of these inhibitors were tested with exemplar MLGases and an endo-β(1,3)-glucanase. Notably, determination of inhibitor-bound crystal structures of a human-gut microbial MLGase from Glycoside Hydrolase Family 16 revealed the orthogonal labeling of the nucleophile and catalytic acid/base residues with homologous 2-fluoro-2-deoxyglycoside and N-bromoacetylglycosylamine inhibitors, respectively. We anticipate that the selectivity of these inhibitors will continue to enable the structural and mechanistic analyses of β-glucanases from diverse sources and protein families.
Collapse
Affiliation(s)
- Namrata Jain
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Kazune Tamura
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Guillaume Déjean
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Harry Brumer
- Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
16
|
Akintola O, Ren W, Adabala PJP, Bhosale S, Wang Y, Ganga-Sah Y, Britton R, Bennet AJ. Intrinsic Nucleophilicity of Inverting and Retaining Glycoside Hydrolases Revealed Using Carbasugar Glyco-Tools. ACS Catal 2021. [DOI: 10.1021/acscatal.1c01634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Oluwafemi Akintola
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Weiwu Ren
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Pal John Pal Adabala
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Sandeep Bhosale
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Yang Wang
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Yumeela Ganga-Sah
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Robert Britton
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Andrew J. Bennet
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
17
|
Abstract
Glycomimetics are structural mimics of naturally occurring carbohydrates and represent important therapeutic leads in several disease treatments. However, the structural and stereochemical complexity inherent to glycomimetics often challenges medicinal chemistry efforts and is incompatible with diversity-oriented synthesis approaches. Here, we describe a one-pot proline-catalyzed aldehyde α-functionalization/aldol reaction that produces an array of stereochemically well-defined glycomimetic building blocks containing fluoro, chloro, bromo, trifluoromethylthio and azodicarboxylate functional groups. Using density functional theory calculations, we demonstrate both steric and electrostatic interactions play key diastereodiscriminating roles in the dynamic kinetic resolution. The utility of this simple process for generating large and diverse libraries of glycomimetics is demonstrated in the rapid production of iminosugars, nucleoside analogues, carbasugars and carbohydrates from common intermediates.
Collapse
|
18
|
Wilson KA, Kung RW, D'souza S, Wetmore SD. Anatomy of noncovalent interactions between the nucleobases or ribose and π-containing amino acids in RNA-protein complexes. Nucleic Acids Res 2021; 49:2213-2225. [PMID: 33544852 PMCID: PMC7913691 DOI: 10.1093/nar/gkab008] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/22/2021] [Indexed: 01/07/2023] Open
Abstract
A set of >300 nonredundant high-resolution RNA–protein complexes were rigorously searched for π-contacts between an amino acid side chain (W, H, F, Y, R, E and D) and an RNA nucleobase (denoted π–π interaction) or ribose moiety (denoted sugar–π). The resulting dataset of >1500 RNA–protein π-contacts were visually inspected and classified based on the interaction type, and amino acids and RNA components involved. More than 80% of structures searched contained at least one RNA–protein π-interaction, with π–π contacts making up 59% of the identified interactions. RNA–protein π–π and sugar–π contacts exhibit a range in the RNA and protein components involved, relative monomer orientations and quantum mechanically predicted binding energies. Interestingly, π–π and sugar–π interactions occur more frequently with RNA (4.8 contacts/structure) than DNA (2.6). Moreover, the maximum stability is greater for RNA–protein contacts than DNA–protein interactions. In addition to highlighting distinct differences between RNA and DNA–protein binding, this work has generated the largest dataset of RNA–protein π-interactions to date, thereby underscoring that RNA–protein π-contacts are ubiquitous in nature, and key to the stability and function of RNA–protein complexes.
Collapse
Affiliation(s)
- Katie A Wilson
- Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive West, Lethbridge, Alberta T1K 3M4, Canada
| | - Ryan W Kung
- Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive West, Lethbridge, Alberta T1K 3M4, Canada
| | - Simmone D'souza
- Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive West, Lethbridge, Alberta T1K 3M4, Canada
| | - Stacey D Wetmore
- Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive West, Lethbridge, Alberta T1K 3M4, Canada
| |
Collapse
|
19
|
Sweeney RP, Danby PM, Geissner A, Karimi R, Brask J, Withers SG. Development of an active site titration reagent for α-amylases. Chem Sci 2020; 12:683-687. [PMID: 34163800 PMCID: PMC8178983 DOI: 10.1039/d0sc05380e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/03/2020] [Indexed: 01/12/2023] Open
Abstract
α-Amylases are among the most widely used classes of enzymes in industry and considerable effort has gone into optimising their activities. Efforts to find better amylase mutants, such as through high-throughput screening, would be greatly aided by access to precise and robust active site titrating agents for quantitation of active mutants in crude cell lysates. While active site titration reagents designed for retaining β-glycosidases quantify these enzymes down to nanomolar levels, convenient titrants for α-glycosidases are not available. We designed such a reagent by incorporating a highly reactive fluorogenic leaving group onto unsaturated cyclitol ethers, which have been recently shown to act as slow substrates for retaining glycosidases that operate via a covalent 'glycosyl'-enzyme intermediate. By appending this warhead onto the appropriate oligosaccharide, we developed efficient active site titration reagents for α-amylases that effect quantitation down to low nanomolar levels.
Collapse
Affiliation(s)
- Ryan P Sweeney
- Department of Chemistry, The University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Phillip M Danby
- Department of Chemistry, The University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Andreas Geissner
- Department of Chemistry, The University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Ryan Karimi
- Department of Chemistry, The University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Jesper Brask
- Novozymes Krogshoejvej 36 2880 Bagsvaerd Denmark
| | - Stephen G Withers
- Department of Chemistry, The University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| |
Collapse
|
20
|
Ren W, Farren-Dai M, Sannikova N, Świderek K, Wang Y, Akintola O, Britton R, Moliner V, Bennet AJ. Glycoside hydrolase stabilization of transition state charge: new directions for inhibitor design. Chem Sci 2020; 11:10488-10495. [PMID: 34094307 PMCID: PMC8162432 DOI: 10.1039/d0sc04401f] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Carbasugars are structural mimics of naturally occurring carbohydrates that can interact with and inhibit enzymes involved in carbohydrate processing. In particular, carbasugars have attracted attention as inhibitors of glycoside hydrolases (GHs) and as therapeutic leads in several disease areas. However, it is unclear how the carbasugars are recognized and processed by GHs. Here, we report the synthesis of three carbasugar isotopologues and provide a detailed transition state (TS) analysis for the formation of the initial GH-carbasugar covalent intermediate, as well as for hydrolysis of this intermediate, using a combination of experimentally measured kinetic isotope effects and hybrid QM/MM calculations. We find that the α-galactosidase from Thermotoga maritima effectively stabilizes TS charge development on a remote C5-allylic center acting in concert with the reacting carbasugar, and catalysis proceeds via an exploded, or loose, SN2 transition state with no discrete enzyme-bound cationic intermediate. We conclude that, in complement to what we know about the TS structures of enzyme-natural substrate complexes, knowledge of the TS structures of enzymes reacting with non-natural carbasugar substrates shows that GHs can stabilize a wider range of positively charged TS structures than previously thought. Furthermore, this enhanced understanding will enable the design of new carbasugar GH transition state analogues to be used as, for example, chemical biology tools and pharmaceutical lead compounds. Positive charge stabilized on remote C5-allylic center with catalysis occurring via a loose SN2 transition state.![]()
Collapse
Affiliation(s)
- Weiwu Ren
- Department of Chemistry, Simon Fraser University Burnaby British Columbia V5A 1S6 Canada +1-778-782-8814
| | - Marco Farren-Dai
- Department of Chemistry, Simon Fraser University Burnaby British Columbia V5A 1S6 Canada +1-778-782-8814
| | - Natalia Sannikova
- Department of Chemistry, Simon Fraser University Burnaby British Columbia V5A 1S6 Canada +1-778-782-8814
| | - Katarzyna Świderek
- Departament de Química Física i Analítica, Universitat Jaume I 12560 Castellón Spain
| | - Yang Wang
- Department of Chemistry, Simon Fraser University Burnaby British Columbia V5A 1S6 Canada +1-778-782-8814
| | - Oluwafemi Akintola
- Department of Chemistry, Simon Fraser University Burnaby British Columbia V5A 1S6 Canada +1-778-782-8814
| | - Robert Britton
- Department of Chemistry, Simon Fraser University Burnaby British Columbia V5A 1S6 Canada +1-778-782-8814
| | - Vicent Moliner
- Departament de Química Física i Analítica, Universitat Jaume I 12560 Castellón Spain
| | - Andrew J Bennet
- Department of Chemistry, Simon Fraser University Burnaby British Columbia V5A 1S6 Canada +1-778-782-8814
| |
Collapse
|
21
|
Meanwell M, Silverman SM, Lehmann J, Adluri B, Wang Y, Cohen R, Campeau LC, Britton R. A short de novo synthesis of nucleoside analogs. Science 2020; 369:725-730. [PMID: 32764073 DOI: 10.1126/science.abb3231] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
Abstract
Nucleoside analogs are commonly used in the treatment of cancer and viral infections. Their syntheses benefit from decades of research but are often protracted, unamenable to diversification, and reliant on a limited pool of chiral carbohydrate starting materials. We present a process for rapidly constructing nucleoside analogs from simple achiral materials. Using only proline catalysis, heteroaryl-substituted acetaldehydes are fluorinated and then directly engaged in enantioselective aldol reactions in a one-pot reaction. A subsequent intramolecular fluoride displacement reaction provides a functionalized nucleoside analog. The versatility of this process is highlighted in multigram syntheses of d- or l-nucleoside analogs, locked nucleic acids, iminonucleosides, and C2'- and C4'-modified nucleoside analogs. This de novo synthesis creates opportunities for the preparation of diversity libraries and will support efforts in both drug discovery and development.
Collapse
Affiliation(s)
- Michael Meanwell
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Steven M Silverman
- Department of Process Research and Development, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Johannes Lehmann
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | | | - Yang Wang
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Ryan Cohen
- Department of Process Research and Development, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Louis-Charles Campeau
- Department of Process Research and Development, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Robert Britton
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada.
| |
Collapse
|
22
|
Das P, Almond DW, Tumbelty LN, Austin BE, Moura-Letts G. From Heterocycles to Carbacycles: Synthesis of Carbocyclic Nucleoside Analogues from Enals and Hydroxylamines. Org Lett 2020; 22:5491-5495. [PMID: 32602726 DOI: 10.1021/acs.orglett.0c01846] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Substituted and stereochemically dense carbacycles can be prepared by sequential dipolar cycloaddition and reductive cleavage from enals and hydroxylamines. The reaction sequence proceeds with high efficiency for a wide variety of enals and hydroxylamines. The reaction is regio- and diastereoselective for the initial formation of a bridged bisisoxazolidine intermediate, which then undergoes quantitative double N-O cleavage to produce carbacycles as single diastereomers.
Collapse
Affiliation(s)
- Pulakesh Das
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Road, Glassboro, New Jersey 08028, United States
| | - David W Almond
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Road, Glassboro, New Jersey 08028, United States
| | - Lauren N Tumbelty
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Road, Glassboro, New Jersey 08028, United States
| | - Brooke E Austin
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Road, Glassboro, New Jersey 08028, United States
| | - Gustavo Moura-Letts
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Road, Glassboro, New Jersey 08028, United States
| |
Collapse
|
23
|
Adabala PJP, Shamsi Kazem Abadi S, Akintola O, Bhosale S, Bennet AJ. Conformationally Controlled Reactivity of Carbasugars Uncovers the Choreography of Glycoside Hydrolase Catalysis. J Org Chem 2020; 85:3336-3348. [PMID: 31994882 DOI: 10.1021/acs.joc.9b03152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Glycoside hydrolases (GHs) catalyze hydrolyses of glycoconjugates in which the enzyme choreographs a series of conformational changes during the catalytic cycle. As a result, some GH families, including α-amylases (GH13), have their chemical steps concealed kinetically. To address this issue for a GH13 enzyme, we prepared seven cyclohexenyl-based carbasugars of α-d-glucopyranoside that we show are good covalent inhibitors of a GH13 yeast α-glucosidase. The linear free energy relationships between rate constants and pKa of the leaving group are curved upward, which is indicative of a change in mechanism, with the better leaving groups reacting by an SN1 mechanism, while reaction rates for the worse leaving groups are limited by a conformational change of the Michaelis complex prior to a rapid SN2 reaction with the enzymatic nucleophile. Five bicyclo[4.1.0]heptyl-based carbaglucoses were tested with this enzyme, and our results are consistent with pseudoglycosidic bond cleavage that occurs via SN1 transition states that include nonproductive binding of the leaving group to the enzyme. In total, we show that the conformationally orthogonal reactions of these two carbasugars reveal mechanistic details hidden by conformational changes that the Michaelis complex of the enzyme and natural substrate undergoes which align the nucleophile for efficient catalysis.
Collapse
Affiliation(s)
- Pal John Pal Adabala
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Saeideh Shamsi Kazem Abadi
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Oluwafemi Akintola
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Sandeep Bhosale
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Andrew J Bennet
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
24
|
Abstract
Quantum mechanics (QM) methods provide a fine description of receptor-ligand interactions and of chemical reactions. Their use in drug design and drug discovery is increasing, especially for complex systems including metal ions in the binding sites, for the design of highly selective inhibitors, for the optimization of bi-specific compounds, to understand enzymatic reactions, and for the study of covalent ligands and prodrugs. They are also used for generating molecular descriptors for predictive QSAR/QSPR models and for the parameterization of force fields. Thanks to the continuous increase of computational power offered by GPUs and to the development of sophisticated algorithms, QM methods are becoming part of the standard tools used in computer-aided drug design (CADD). We present the most used QM methods and software packages, and we discuss recent representative applications in drug design and drug discovery.
Collapse
Affiliation(s)
- Martin Kotev
- Global Research Informatics/Cheminformatics and Drug Design, Evotec (France) SAS, Toulouse, France
| | - Laurie Sarrat
- Global Research Informatics/Cheminformatics and Drug Design, Evotec (France) SAS, Toulouse, France
| | | |
Collapse
|
25
|
Tyrikos-Ergas T, Fittolani G, Seeberger PH, Delbianco M. Structural Studies Using Unnatural Oligosaccharides: Toward Sugar Foldamers. Biomacromolecules 2019; 21:18-29. [DOI: 10.1021/acs.biomac.9b01090] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Theodore Tyrikos-Ergas
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Giulio Fittolani
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Peter H. Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Martina Delbianco
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
| |
Collapse
|
26
|
Ray S, Murkin AS. New Electrophiles and Strategies for Mechanism-Based and Targeted Covalent Inhibitor Design. Biochemistry 2019; 58:5234-5244. [PMID: 30990686 DOI: 10.1021/acs.biochem.9b00293] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Covalent inhibitors are experiencing a growing resurgence in drug design and are an increasingly useful tool in molecular biology. The ability to attach inhibitors to their targets by a covalent linkage offers pharmacodynamic and pharmacokinetic advantages, but this can also be a liability if undesired off-target reactions are not mitigated. The discovery of new electrophilic groups that react selectively with specific amino acid residues is therefore highly desirable in the design of targeted covalent inhibitors (TCIs). Additionally, the ability to control the reactivity through exploitation of the target enzyme's machinery, as in mechanism-based inhibitors (MBIs), greatly benefits from the discovery of new strategies. This Perspective showcases recent advances in electrophile development and their application in TCIs and MBIs, exhibiting high selectivity for their targets.
Collapse
Affiliation(s)
- Sneha Ray
- Department of Chemistry , University at Buffalo, The State University of New York , Buffalo , New York 14260-3000 , United States
| | - Andrew S Murkin
- Department of Chemistry , University at Buffalo, The State University of New York , Buffalo , New York 14260-3000 , United States
| |
Collapse
|