1
|
Shree M, Vaishnav J, Gurudayal, Ampapathi RS. In-silico assessment of novel peptidomimetics inhibitor targeting STAT3 and STAT4 N-terminal domain dimerization: A comprehensive study using molecular docking, molecular dynamics simulation, and binding free energy analysis. Biochem Biophys Res Commun 2024; 733:150584. [PMID: 39208642 DOI: 10.1016/j.bbrc.2024.150584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/04/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Dysregulation in Janus kinase-Signal Transducer and Activation of Transcription (JAK-STAT) pathway is closely linked to various cancer types. The N-terminal domain (NTD) of STAT proteins, upon dimerization, assumes a multifaceted role with remarkable adaptability in mediating interactions between proteins. Consequently, the strategic targeting of the N-terminal domain of STATs has emerged as a promising tactic for disrupting dimerization and impeding the translocation of STAT proteins. In this study, we have deployed an integrated in-silico methodology to rationally design Peptidomimetic foldamers as inhibitors of the N-terminal domains of STAT3 and STAT4, with the objective of disrupting protein dimerization. Consequently, we have judiciously designed a series of peptidomimetics that encompass β3-amino acids, bearing side chains that mimic the residues within interface II of the dimeric structures of the NTDs. Employing molecular docking techniques; we have assessed the binding affinity of these designed peptidomimetics toward both the NTDs. Furthermore, we have conducted an evaluation of the stability and conformational alterations within the docked complexes over an extensive Molecular Dynamics, subsequently computing the binding free energy utilizing MM/PBSA calculations. Our findings unequivocally demonstrate that the peptidomimetic foldamers we have devised (Peptide-A, Peptide-B, and Peptide-C) exhibit a propensity to bind to and impede the dimerization process of the NTDs of both STAT3 and STAT4. These outcomes serve to underscore the potential of these meticulously designed peptidomimetics as potential candidates meriting further exploration in the realm of cancer prevention and management.
Collapse
Affiliation(s)
- Megha Shree
- Sophisticated Analytical Instrumentation Facility & Research (SAIF-R), CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India
| | - Jayanti Vaishnav
- Sophisticated Analytical Instrumentation Facility & Research (SAIF-R), CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India
| | - Gurudayal
- Sophisticated Analytical Instrumentation Facility & Research (SAIF-R), CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India
| | - Ravi Sankar Ampapathi
- Sophisticated Analytical Instrumentation Facility & Research (SAIF-R), CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
2
|
Kazantsev K, Toukach P. Remediation of the NMR data of natural glycans. Int J Biol Macromol 2024; 282:137042. [PMID: 39521218 DOI: 10.1016/j.ijbiomac.2024.137042] [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: 07/10/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Primary structure elucidation in glycobiology is strongly affected by published structure-reporting NMR signals, especially on the 13C nucleus. The glycan NMR simulation accuracy and machine learning outcome depend on the quality of the NMR signal assignment in glycan databases. Within our work on improving the data quality in the Carbohydrate Structure Database (CSDB), we have applied a systematic search for inconsistencies in the published NMR data. The search was based on a bulk comparison between the experimental and simulated 13C NMR chemical shifts and manual analysis of the mismatches. On the basis of this analysis, CSDB was remediated by marking and correcting the NMR errors found in 272 structure elucidation reports published over the past 40 years.
Collapse
Affiliation(s)
- Kirill Kazantsev
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russia
| | - Philip Toukach
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russia; National Research University Higher School of Economics, Faculty of Chemistry, Vavilova 7, 117312 Moscow, Russia.
| |
Collapse
|
3
|
Grabarics M, Mallada B, Edalatmanesh S, Jiménez-Martín A, Pykal M, Ondráček M, Kührová P, Struwe WB, Banáš P, Rauschenbach S, Jelínek P, de la Torre B. Atomically resolved imaging of the conformations and adsorption geometries of individual β-cyclodextrins with non-contact AFM. Nat Commun 2024; 15:9482. [PMID: 39488518 PMCID: PMC11531514 DOI: 10.1038/s41467-024-53555-0] [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: 06/22/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024] Open
Abstract
Glycans, consisting of covalently linked sugar units, are a major class of biopolymers essential to all known living organisms. To better understand their biological functions and further applications in fields from biomedicine to materials science, detailed knowledge of their structure is essential. However, due to the extraordinary complexity and conformational flexibility of glycans, state-of-the-art glycan analysis methods often fail to provide structural information with atomic precision. Here, we combine electrospray deposition in ultra-high vacuum with non-contact atomic force microscopy and theoretical calculations to unravel the structure of β-cyclodextrin, a cyclic glucose oligomer, with atomic-scale detail. Our results, established on the single-molecule level, reveal the different adsorption geometries and conformations of β-cyclodextrin. The position of individual hydroxy groups and the location of the stabilizing intramolecular H-bonds are deduced from atomically resolved images, enabling the unambiguous assignment of the molecular structure and demonstrating the potential of the method for glycan analysis.
Collapse
Affiliation(s)
- Márkó Grabarics
- Department of Chemistry, University of Oxford, OX1 3QU, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, OX1 3QU, Oxford, UK
| | - Benjamín Mallada
- Institute of Physics, Czech Academy of Sciences, 16200, Prague, Czech Republic
- Czech Advanced Technology and Research Institute, Palacký University Olomouc, 78371, Olomouc, Czech Republic
| | - Shayan Edalatmanesh
- Institute of Physics, Czech Academy of Sciences, 16200, Prague, Czech Republic
- Czech Advanced Technology and Research Institute, Palacký University Olomouc, 78371, Olomouc, Czech Republic
| | - Alejandro Jiménez-Martín
- Institute of Physics, Czech Academy of Sciences, 16200, Prague, Czech Republic
- Czech Advanced Technology and Research Institute, Palacký University Olomouc, 78371, Olomouc, Czech Republic
- Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, 115 19, Prague, Czech Republic
| | - Martin Pykal
- Czech Advanced Technology and Research Institute, Palacký University Olomouc, 78371, Olomouc, Czech Republic
| | - Martin Ondráček
- Institute of Physics, Czech Academy of Sciences, 16200, Prague, Czech Republic
| | - Petra Kührová
- Czech Advanced Technology and Research Institute, Palacký University Olomouc, 78371, Olomouc, Czech Republic
| | - Weston B Struwe
- Kavli Institute for Nanoscience Discovery, University of Oxford, OX1 3QU, Oxford, UK
- Department of Biochemistry, University of Oxford, OX1 3QU, Oxford, UK
| | - Pavel Banáš
- Czech Advanced Technology and Research Institute, Palacký University Olomouc, 78371, Olomouc, Czech Republic.
| | - Stephan Rauschenbach
- Department of Chemistry, University of Oxford, OX1 3QU, Oxford, UK.
- Kavli Institute for Nanoscience Discovery, University of Oxford, OX1 3QU, Oxford, UK.
| | - Pavel Jelínek
- Institute of Physics, Czech Academy of Sciences, 16200, Prague, Czech Republic.
- Czech Advanced Technology and Research Institute, Palacký University Olomouc, 78371, Olomouc, Czech Republic.
| | - Bruno de la Torre
- Czech Advanced Technology and Research Institute, Palacký University Olomouc, 78371, Olomouc, Czech Republic.
- Nanomaterials and Nanotechnology Research Center, CSIC-UNIOVI-PA, 33940, El Entrego, Spain.
| |
Collapse
|
4
|
D’Orazio G, La Ferla B. Synthesis of a Small Library of Glycoderivative Putative Ligands of SGLT1 and Preliminary Biological Evaluation. Molecules 2024; 29:5067. [PMID: 39519708 PMCID: PMC11547630 DOI: 10.3390/molecules29215067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Sodium-glucose co-transporter 1 (SGLT1) is primarily expressed on the membrane of enterocytes, a type of epithelial cell found in the intestines, where it mediates the unidirectional absorption of glucose and galactose. Beyond its well-established role in nutrient absorption, SGLT1 also plays a protective role in maintaining the integrity of the intestinal barrier. Specifically, the natural ligand of SGLT1 (d-glucose) and a synthetic C-glucoside developed by our group can induce a protective anti-inflammatory effect on the intestinal epithelium. In this paper, we report the creation of a small library of C-glycoside, putative ligands for SGLT1, to gain further insights into its unclear mechanism of action. Preliminary biological experiments performed on an in vitro model of doxorubicin-induced mucositis, a severe intestinal inflammatory condition, indicate that the aromatic moiety present in all the compounds of the library is crucial for biological activity, while the sugar component appears to have less influence. These findings will be exploited to develop new, more potent anti-inflammatory compounds and to better understand and rationalize the protective mechanism of action.
Collapse
Affiliation(s)
- Giuseppe D’Orazio
- Department of Chemistry, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy
| | - Barbara La Ferla
- Department of Earth and Environmental Sciences DISAT, Università degli Studi di Milano-Bicocca, Piazza della Scienza 1, 20126 Milan, Italy
| |
Collapse
|
5
|
Depienne S, Fontenelle C, Light ME, Hecke KV, Linclau B. De Novo Enantioselective Synthesis of Hexafluorinated d-Glucose. J Org Chem 2024; 89:14291-14304. [PMID: 39269924 PMCID: PMC11460824 DOI: 10.1021/acs.joc.4c01724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024]
Abstract
We report a de novo enantioselective synthesis of 2,3,4-trideoxy-2,2,3,3,4,4-hexafluoro-d-glycero-hexopyranose (hexafluorinated d-glucose), an iconic polar hydrophobic glycomimetic. The 12-step synthesis features robust and reproducible chemistry and was achieved by incorporating an asymmetric dihydroxylation step to install the stereogenic center with excellent enantioselectivity (95:5 er). Virtual enantiopurity (>99.5% ee) was further reached using a simple crystallization procedure and the absolute confirmation was ascertained by X-ray analysis. The synthetic route also allowed access to the novel hexafluorinated heptose derivative 2,3,4-trideoxy-2,2,3,3,4,4-hexafluoro-l-threo-heptopyranose.
Collapse
Affiliation(s)
- Sébastien Depienne
- Department
of Organic and Macromolecular Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S4, 9000 Ghent, Belgium
| | - Clément
Q. Fontenelle
- School
of Chemistry, University of Southampton, Highfield, Southampton SO17 1BJ, U.K.
| | - Mark E. Light
- School
of Chemistry, University of Southampton, Highfield, Southampton SO17 1BJ, U.K.
| | - Kristof Van Hecke
- Department
of Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S3, 9000 Ghent, Belgium
| | - Bruno Linclau
- Department
of Organic and Macromolecular Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S4, 9000 Ghent, Belgium
- School
of Chemistry, University of Southampton, Highfield, Southampton SO17 1BJ, U.K.
| |
Collapse
|
6
|
Murphy PV, Dhara A, Fitzgerald LS, Hever E, Konda S, Mandal K. Small lectin ligands as a basis for applications in glycoscience and glycomedicine. Chem Soc Rev 2024; 53:9428-9445. [PMID: 39162695 DOI: 10.1039/d4cs00642a] [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: 08/21/2024]
Abstract
Glycan recognition by lectins mediates important biological events. This Tutorial Review aims to introduce lectin-ligand interactions and show how these molecular recognition events inspire innovations such as: (i) glycomimetic ligands; (ii) multivalent ligand agonists/antagonists; (iii) ligands for precision delivery of therapies to cells, where therapies include vaccines, siRNA and LYTACs (iv) development of diagnostics. A small number of case studies are selected to demonstrate principles for development of new ligands for applications inspired by knowledge of natural glycan ligand structure and function.
Collapse
Affiliation(s)
- Paul V Murphy
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, University Road, Galway, H91TK33, Ireland
| | - Ashis Dhara
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
| | - Liam S Fitzgerald
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, University Road, Galway, H91TK33, Ireland
| | - Eoin Hever
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| | - Saidulu Konda
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| | - Kishan Mandal
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| |
Collapse
|
7
|
Hu L, Li R, Liu Y, Zhou J, Sun Z. Photocatalytic Synthesis of α-Ketonyl Glycosyl Compounds from Glycosyl Thiols and Silyl Enol Ethers. Org Lett 2024; 26:8188-8193. [PMID: 39297709 DOI: 10.1021/acs.orglett.4c03124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
The synthesis of C1-ketonyl glycosyl compounds featuring α-selectivity has seldom been reported. We herein devise a glycosyl radical-based approach to facilely access stereoenriched ketonyl glycosyl compounds via an Ir photoredox-catalyzed desulfurative addition to silyl enol ethers, using in situ-generated tetrafluoropyridinyl thioglycosides from glycosyl 1-thiols as radical precursors. This protocol features readily prepared starting materials, mild conditions, excellent functional group tolerance, satisfactory scale-up, and notable amenability to late-stage modification of pharmaceutically relevant complex molecules.
Collapse
Affiliation(s)
- Lifu Hu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Ruining Li
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yunqi Liu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Junliang Zhou
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zhankui Sun
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmaceutical Sciences Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
- Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
8
|
Lang M, Walter S, Hatey D, Blanc A, Compain P, Kern N. Gold(I)-Catalyzed Reactions of exo-Glycals with Propargyl Esters toward C-1 Alkenyl Spirocyclopropyl Carbohydrates. Org Lett 2024; 26:8017-8022. [PMID: 39283904 DOI: 10.1021/acs.orglett.4c02754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
An atom-economic and diazo-free strategy for the construction of novel pseudo anomeric C-1 alkenyl spirocyclopropyl sugars is described. Leveraging the 1,2-migration pathway of propargyl esters under gold(I) catalysis, easily available exo-glycals undergo β-selective alkenylcarbenoid insertion in moderate to excellent yields. Preferential activation of propargyl moieties and concerted [2 + 1] insertion are both favored through ligand choice and electron enrichment of esters. Stereocontrol using conformational bias and rearrangement are also demonstrated.
Collapse
Affiliation(s)
- Mylène Lang
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Université de Strasbourg/Université de Haute-Alsace/CNRS (UMR 7042), Equipe de Synthèse Organique et Molécules Bioactives (SYBIO), ECPM, 25 rue Becquerel, 67087 Strasbourg, France
| | - Sophie Walter
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Université de Strasbourg/Université de Haute-Alsace/CNRS (UMR 7042), Equipe de Synthèse Organique et Molécules Bioactives (SYBIO), ECPM, 25 rue Becquerel, 67087 Strasbourg, France
| | - Delphine Hatey
- Institut de Chimie (UMR 7177), Université de Strasbourg, Laboratoire de Catalyse Organométallique, Synthèse organique et Santé (COSyS), 4 rue Blaise Pascal, CS 90032, 67081 Strasbourg, France
| | - Aurélien Blanc
- Institut de Chimie (UMR 7177), Université de Strasbourg, Laboratoire de Catalyse Organométallique, Synthèse organique et Santé (COSyS), 4 rue Blaise Pascal, CS 90032, 67081 Strasbourg, France
| | - Philippe Compain
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Université de Strasbourg/Université de Haute-Alsace/CNRS (UMR 7042), Equipe de Synthèse Organique et Molécules Bioactives (SYBIO), ECPM, 25 rue Becquerel, 67087 Strasbourg, France
| | - Nicolas Kern
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Université de Strasbourg/Université de Haute-Alsace/CNRS (UMR 7042), Equipe de Synthèse Organique et Molécules Bioactives (SYBIO), ECPM, 25 rue Becquerel, 67087 Strasbourg, France
| |
Collapse
|
9
|
Petronilho EC, de Andrade GC, de Sousa GDS, Almeida FP, Mota MF, Gomes AVDS, Pinheiro CHS, da Silva MC, Arruda HRS, Marques MA, Vieira TCRG, de Oliveira GAP, Silva JL. Oncogenic p53 triggers amyloid aggregation of p63 and p73 liquid droplets. Commun Chem 2024; 7:207. [PMID: 39284933 PMCID: PMC11405828 DOI: 10.1038/s42004-024-01289-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/29/2024] [Indexed: 09/20/2024] Open
Abstract
P53 Phase separation is crucial towards amyloid aggregation and p63 and p73 have enhanced expression in tumors. This study examines the phase behaviors of p53, p63, and p73. Here we show that unlike the DNA-binding domain of p53 (p53C), the p63C and p73C undergo phase separation, but do not form amyloids under physiological temperatures. Wild-type and mutant p53C form droplets at 4°C and aggregates at 37 °C with amyloid properties. Mutant p53C promotes amyloid-like states in p63C and p73C, recruiting them into membraneless organelles. Amyloid conversion is supported by thioflavin T and Congo red binding, increased light scattering, and circular dichroism. Full-length mutant p53 and p63C (or p73C) co-transfection shows reduced fluorescence recovery after photobleaching. Heparin inhibits the prion-like aggregation of p63C and p73C induced by p53C. These findings highlight the role of p53 in initiating amyloid aggregation in p63 and p73, opening avenues for targeting prion-like conversion in cancer therapy.
Collapse
Affiliation(s)
- Elaine C Petronilho
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Guilherme C de Andrade
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gileno Dos S de Sousa
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernando P Almeida
- National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Michelle F Mota
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Vitória Dos S Gomes
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carlos Henrique S Pinheiro
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mylena C da Silva
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Hiam R S Arruda
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mayra A Marques
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Tuane C R G Vieira
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Guilherme A P de Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Jerson L Silva
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
- National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
10
|
Jordan C, Hayashi T, Löbbert A, Fan J, Teschers CS, Siebold K, Aufiero M, Pape F, Campbell E, Axer A, Bussmann K, Bergander K, Köhnke J, Gossert AD, Gilmour R. Probing the Origin of Affinity in the GM1-Cholera Toxin Complex through Site-Selective Editing with Fluorine. ACS CENTRAL SCIENCE 2024; 10:1481-1489. [PMID: 39220706 PMCID: PMC11363330 DOI: 10.1021/acscentsci.4c00622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 09/04/2024]
Abstract
Carbohydrates regulate an inimitable spectrum of biological functions, yet successfully leveraging this therapeutic avenue continues to be frustrated by low affinities with glycan-specific proteins. A conspicuous exception is the interaction of monosialotetrahexosylganglioside (GM1) with the carbohydrate-recognition domain of cholera toxin from Vibrio cholerae: this is one of the strongest protein-carbohydrate interactions known. To establish the importance of a long-discussed key hydrogen bond between C2 of the terminal galactose of GM1 and the B subunit pentamer of cholera toxin (CTB5), the total synthesis of a selectively fluorinated GM1 epitope was conducted in 19 steps. This process of molecular editing (Oδ-H → Fδ-) strategically deletes the hydrogen bond donor while retaining the localized partial charge of the substituent. Comparison of the binding affinity of F-GM1/CTB5 with native GM1, the GM1 carbohydrate epitope, and meta-mononitrophenyl-α-galactoside (MNPG) revealed a trend that fully supports the importance of this key interaction. These NMR data suggest that F-GM1 binds in a closely similar conformation as native GM1. Crystallographic analyses of the complex also confirm that the OH → F bioisosteric exchange at C2 of the terminal galactose induces a ring conformation that eliminates key hydrogen bonds: these interactions are compensated for by inter- and intramolecular fluorine-specific interactions.
Collapse
Affiliation(s)
- Christina Jordan
- Institute
for Organic Chemistry, University of Münster, 48149 Münster, Germany
| | - Taiki Hayashi
- Institute
for Organic Chemistry, University of Münster, 48149 Münster, Germany
| | | | - Jingran Fan
- Institut
für Lebensmittelchemie, Leibniz Universität
Hannover, 30167 Hannover, Germany
| | | | - Kathrin Siebold
- Institute
for Organic Chemistry, University of Münster, 48149 Münster, Germany
| | - Marialuisa Aufiero
- Institute
for Organic Chemistry, University of Münster, 48149 Münster, Germany
| | - Felix Pape
- Institute
for Organic Chemistry, University of Münster, 48149 Münster, Germany
| | - Emma Campbell
- Institute
for Organic Chemistry, University of Münster, 48149 Münster, Germany
| | - Alexander Axer
- Institute
for Organic Chemistry, University of Münster, 48149 Münster, Germany
| | - Kathrin Bussmann
- Institute
for Organic Chemistry, University of Münster, 48149 Münster, Germany
| | - Klaus Bergander
- Institute
for Organic Chemistry, University of Münster, 48149 Münster, Germany
| | - Jesko Köhnke
- Institut
für Lebensmittelchemie, Leibniz Universität
Hannover, 30167 Hannover, Germany
| | | | - Ryan Gilmour
- Institute
for Organic Chemistry, University of Münster, 48149 Münster, Germany
| |
Collapse
|
11
|
Krömer M, Poštová Slavětínská L, Hocek M. Glyco-DNA: Enzymatic Synthesis of Base-Modified and Hypermodified DNA Displaying up to Four Different Monosaccharide Units in the Major Groove. Chemistry 2024; 30:e202402318. [PMID: 38896019 DOI: 10.1002/chem.202402318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/21/2024]
Abstract
A portfolio of six modified 2'-deoxyribonucleoside triphosphate (dNTP) derivatives derived from 5-substituted pyrimidine or 7-substituted 7-deazapurine bearing different carbohydrate units (d-glucose, d-galactose, d-mannose, l-fucose, sialic acid and N-Ac-d-galactosamine) tethered through propargyl-glycoside linker was designed and synthesized via the Sonogashira reactions of halogenated dNTPs with the corresponding propargyl-glycosides. The nucleotides were found to be good substrates for DNA polymerases in enzymatic primer extension and PCR synthesis of modified and hypermodified DNA displaying up to four different sugars. Proof of concept binding study of sugar-modified oligonucleotides with concanavalin A showed positive effect of avidity and sugar units count.
Collapse
Affiliation(s)
- Matouš Krömer
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
- Department of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, 12843, Prague 2, Czech Republic
| | - Lenka Poštová Slavětínská
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
| | - Michal Hocek
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, 16000, Prague 6, Czech Republic
- Department of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, 12843, Prague 2, Czech Republic
| |
Collapse
|
12
|
Diehl R, Chorghade RS, Keys AM, Alam MM, Early SA, Dugan AE, Krupkin M, Ribbeck K, Kulik HJ, Kiessling LL. CH-π Interactions Are Required for Human Galectin-3 Function. JACS AU 2024; 4:3028-3037. [PMID: 39211619 PMCID: PMC11350569 DOI: 10.1021/jacsau.4c00357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 09/04/2024]
Abstract
Glycan-binding proteins, or lectins, recognize distinct structural elements of polysaccharides, to mediate myriad biological functions. Targeting glycan-binding proteins involved in human disease has been challenging due to an incomplete understanding of the molecular mechanisms that govern protein-glycan interactions. Bioinformatics and structural studies of glycan-binding proteins indicate that aromatic residues with the potential for CH-π interactions are prevalent in glycan-binding sites. However, the contributions of these CH-π interactions to glycan binding and their relevance in downstream function remain unclear. An emblematic lectin, human galectin-3, recognizes lactose and N-acetyllactosamine-containing glycans by positioning the electropositive face of a galactose residue over the tryptophan 181 (W181) indole forming a CH-π interaction. We generated a suite of galectin-3 W181 variants to assess the importance of these CH-π interactions to glycan binding and function. As determined experimentally and further validated with computational modeling, variants with smaller or less electron-rich aromatic side chains (W181Y, W181F, W181H) or sterically similar but nonaromatic residues (W181M, W181R) showed poor or undetectable binding to lactose and attenuated ability to bind mucins or agglutinate red blood cells. The latter functions depend on multivalent binding, highlighting that weakened CH-π interactions cannot be overcome by avidity. Two galectin-3 variants with disrupted hydrogen bonding interactions (H158A and E184A) showed similarly impaired lactose binding. Molecular simulations demonstrate that all variants have decreased binding orientation stability relative to native galectin-3. Thus, W181 collaborates with the endogenous hydrogen bonding network to enhance binding affinity for lactose, and abrogation of these CH-π interactions is as deleterious as eliminating key hydrogen bonding interactions. These findings underscore the critical roles of CH-π interactions in carbohydrate binding and lectin function and will aid the development of novel lectin inhibitors.
Collapse
Affiliation(s)
- Roger
C. Diehl
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Rajeev S. Chorghade
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Allison M. Keys
- Program
in Computational and Systems Biology, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Mohammad Murshid Alam
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Stephen A. Early
- Department
of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Amanda E. Dugan
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Miri Krupkin
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Katharina Ribbeck
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Heather J. Kulik
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Laura L. Kiessling
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
- The Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Koch
Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
13
|
Shi WY, Ma JJ, Li HY, Chen D, Liu XY, Liang YM. Synthesis of C-Alkyl Glycosides from Alkyl Bromides and Glycosyl Carboxylic Acids via Ni/Photoredox Dual Catalysis. J Org Chem 2024; 89:11136-11147. [PMID: 39106492 DOI: 10.1021/acs.joc.4c00533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
Abstract
C-Alkyl glycosides, an important class of C-glycosides, are widely found in various drugs and natural products. The synthesis of C-alkyl glycosides has attracted considerable attention. Herein, we developed a Ni/photoredox catalyzed decarboxylative C(sp3)-C(sp3) coupling reaction of stable glycosylcarboxylic acids with simple aliphatic bromides to generate C-alkyl glycosides. The method successfully linked several functional molecular fragments (natural products or drugs) to a sugar moiety, showing the extensive application prospects of this transformation. Controlled experiments and DFT calculations demonstrated that the reaction pathway contains a free radical process, and a possible mechanism is proposed.
Collapse
Affiliation(s)
- Wei-Yu Shi
- State Key Laboratory of Applied Organic Chemistry, School of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Jia-Jun Ma
- State Key Laboratory of Applied Organic Chemistry, School of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Hu-Yi Li
- State Key Laboratory of Applied Organic Chemistry, School of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Dongping Chen
- College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu 730070, P. R. China
| | - Xue-Yuan Liu
- State Key Laboratory of Applied Organic Chemistry, School of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Yong-Min Liang
- State Key Laboratory of Applied Organic Chemistry, School of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| |
Collapse
|
14
|
Alshehri JA, Jones AM. Chemical approaches to the sulfation of small molecules: current progress and future directions. Essays Biochem 2024:EBC20240001. [PMID: 38958528 DOI: 10.1042/ebc20240001] [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: 03/04/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
Sulfation is one of the most important modifications that occur to a wide range of bioactive small molecules including polysaccharides, proteins, flavonoids, and steroids. In turn, these sulfated molecules have significant biological and pharmacological roles in diverse processes including cell signalling, modulation of immune and inflammation response, anti-coagulation, anti-atherosclerosis, and anti-adhesive properties. This Essay summarises the most encountered chemical sulfation methods of small molecules. Sulfation reactions using sulfur trioxide amine/amide complexes are the most used method for alcohol and phenol groups in carbohydrates, steroids, proteins, and related scaffolds. Despite the effectiveness of these methods, they suffer from issues including multiple-purification steps, toxicity issues (e.g., pyridine contamination), purification challenges, stoichiometric excess of reagents which leads to an increase in reaction cost, and intrinsic stability issues of both the reagent and product. Recent advances including SuFEx, the in situ reagent approach, and TBSAB show the widespread appeal of novel sulfating approaches that will enable a larger exploration of the field in the years to come by simplifying the purification and isolation process to access bespoke sulfated small molecules.
Collapse
Affiliation(s)
- Jaber A Alshehri
- School of Pharmacy, University of Birmingham, Edgbaston, B15 2TT, United Kingdom
| | - Alan M Jones
- School of Pharmacy, University of Birmingham, Edgbaston, B15 2TT, United Kingdom
| |
Collapse
|
15
|
Jiang Y, Wei Y, Zhou QY, Sun GQ, Fu XP, Levin N, Zhang Y, Liu WQ, Song N, Mohammed S, Davis BG, Koh MJ. Direct radical functionalization of native sugars. Nature 2024; 631:319-327. [PMID: 38898275 PMCID: PMC11236704 DOI: 10.1038/s41586-024-07548-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/09/2024] [Indexed: 06/21/2024]
Abstract
Naturally occurring (native) sugars and carbohydrates contain numerous hydroxyl groups of similar reactivity1,2. Chemists, therefore, rely typically on laborious, multi-step protecting-group strategies3 to convert these renewable feedstocks into reagents (glycosyl donors) to make glycans. The direct transformation of native sugars to complex saccharides remains a notable challenge. Here we describe a photoinduced approach to achieve site- and stereoselective chemical glycosylation from widely available native sugar building blocks, which through homolytic (one-electron) chemistry bypasses unnecessary hydroxyl group masking and manipulation. This process is reminiscent of nature in its regiocontrolled generation of a transient glycosyl donor, followed by radical-based cross-coupling with electrophiles on activation with light. Through selective anomeric functionalization of mono- and oligosaccharides, this protecting-group-free 'cap and glycosylate' approach offers straightforward access to a wide array of metabolically robust glycosyl compounds. Owing to its biocompatibility, the method was extended to the direct post-translational glycosylation of proteins.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Chemistry, National University of Singapore, Singapore, Singapore
- The Rosalind Franklin Institute, Harwell Science and Innovation Campus, Didcot, UK
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Yi Wei
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Qian-Yi Zhou
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Guo-Quan Sun
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Xia-Ping Fu
- The Rosalind Franklin Institute, Harwell Science and Innovation Campus, Didcot, UK
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Nikita Levin
- The Rosalind Franklin Institute, Harwell Science and Innovation Campus, Didcot, UK
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Yijun Zhang
- Department of Chemistry, National University of Singapore, Singapore, Singapore
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, China
| | - Wen-Qiang Liu
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - NingXi Song
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Shabaz Mohammed
- The Rosalind Franklin Institute, Harwell Science and Innovation Campus, Didcot, UK
- Department of Chemistry, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Benjamin G Davis
- The Rosalind Franklin Institute, Harwell Science and Innovation Campus, Didcot, UK.
- Department of Pharmacology, University of Oxford, Oxford, UK.
- Department of Chemistry, University of Oxford, Oxford, UK.
| | - Ming Joo Koh
- Department of Chemistry, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
16
|
He X, Zhao L, Tian Y, Li R, Chu Q, Gu Z, Zheng M, Wang Y, Li S, Jiang H, Jiang Y, Wen L, Wang D, Cheng X. Highly accurate carbohydrate-binding site prediction with DeepGlycanSite. Nat Commun 2024; 15:5163. [PMID: 38886381 PMCID: PMC11183243 DOI: 10.1038/s41467-024-49516-2] [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: 11/27/2023] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
As the most abundant organic substances in nature, carbohydrates are essential for life. Understanding how carbohydrates regulate proteins in the physiological and pathological processes presents opportunities to address crucial biological problems and develop new therapeutics. However, the diversity and complexity of carbohydrates pose a challenge in experimentally identifying the sites where carbohydrates bind to and act on proteins. Here, we introduce a deep learning model, DeepGlycanSite, capable of accurately predicting carbohydrate-binding sites on a given protein structure. Incorporating geometric and evolutionary features of proteins into a deep equivariant graph neural network with the transformer architecture, DeepGlycanSite remarkably outperforms previous state-of-the-art methods and effectively predicts binding sites for diverse carbohydrates. Integrating with a mutagenesis study, DeepGlycanSite reveals the guanosine-5'-diphosphate-sugar-recognition site of an important G-protein coupled receptor. These findings demonstrate DeepGlycanSite is invaluable for carbohydrate-binding site prediction and could provide insights into molecular mechanisms underlying carbohydrate-regulation of therapeutically important proteins.
Collapse
Affiliation(s)
- Xinheng He
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lifen Zhao
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yinping Tian
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Rui Li
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qinyu Chu
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, China
| | - Zhiyong Gu
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, China
| | - Mingyue Zheng
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, China
| | - Yusong Wang
- National Key Laboratory of Human-Machine Hybrid Augmented Intelligence, National Engineering Research Center for Visual Information and Applications, and Institute of Artificial Intelligence and Robotics, Xi'an Jiaotong University, Xi'an, China
| | - Shaoning Li
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, China
- Lingang Laboratory, Shanghai, China
| | - Yi Jiang
- Lingang Laboratory, Shanghai, China
| | - Liuqing Wen
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | | | - Xi Cheng
- State Key Laboratory of Drug Research and State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute of Advanced Study, Hangzhou, China.
| |
Collapse
|
17
|
Van Den Bergh A, Bailly B, Guillon P, von Itzstein M, Dirr L. Novel insights into the host cell glycan binding profile of human metapneumovirus. J Virol 2024; 98:e0164123. [PMID: 38690874 PMCID: PMC11237588 DOI: 10.1128/jvi.01641-23] [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: 10/27/2023] [Accepted: 03/28/2024] [Indexed: 05/03/2024] Open
Abstract
Numerous viruses have been found to exploit glycoconjugates expressed on human cells as their initial attachment factor for viral entry and infection. The virus-cell glycointeractome, when characterized, may serve as a template for antiviral drug design. Heparan sulfate proteoglycans extensively decorate the human cell surface and were previously described as a primary receptor for human metapneumovirus (HMPV). After respiratory syncytial virus, HMPV is the second most prevalent respiratory pathogen causing respiratory tract infection in young children. To date, there is neither vaccine nor drug available to prevent or treat HMPV infection. Using a multidisciplinary approach, we report for the first time the glycointeractome of the HMPV fusion (F) protein, a viral surface glycoprotein that is essential for target-cell recognition, attachment, and entry. Our glycan microarray and surface plasmon resonance results suggest that Galβ1-3/4GlcNAc moieties that may be sialylated or fucosylated are readily recognized by HMPV F. The bound motifs are highly similar to the N-linked and O-linked glycans primarily expressed on the human lung epithelium. We demonstrate that the identified glycans have the potential to compete with the cellular receptors used for HMPV entry and consequently block HMPV infection. We found that lacto-N-neotetraose demonstrated the strongest HMPV binding inhibition in a cell infection assay. Our current findings offer an encouraging and novel avenue for the design of anti-HMPV drug candidates using oligosaccharide templates.IMPORTANCEAll cells are decorated with a dense coat of sugars that makes a sugar code. Many respiratory viruses exploit this sugar code by binding to these sugars to cause infection. Human metapneumovirus is a leading cause for acute respiratory tract infections. Despite its medical importance, there is no vaccine or antiviral drug available to prevent or treat human metapneumovirus infection. This study investigates how human metapneumovirus binds to sugars in order to more efficiently infect the human host. We found that human metapneumovirus binds to a diverse range of sugars and demonstrated that these sugars can ultimately block viral infection. Understanding how viruses can take advantage of the sugar code on our cells could identify new intervention and treatment strategies to combat viral disease.
Collapse
Affiliation(s)
| | - Benjamin Bailly
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Patrice Guillon
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Larissa Dirr
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
18
|
Wagner B, Smieško M, Jakob RP, Mühlethaler T, Cramer J, Maier T, Rabbani S, Schwardt O, Ernst B. Analogues of the pan-selectin antagonist rivipansel (GMI-1070). Eur J Med Chem 2024; 272:116455. [PMID: 38728868 DOI: 10.1016/j.ejmech.2024.116455] [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: 03/09/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/12/2024]
Abstract
The selectin family consisting of E-, P- and L-selectin plays dominant roles in atherosclerosis, ischemia-reperfusion injury, inflammatory diseases, and metastatic spreading of some cancers. An early goal in selectin-targeted drug discovery campaigns was to identify ligands binding to all three selectins, so-called pan-selectin antagonists. The physiological epitope, tetrasaccharide sialyl Lewisx (sLex, 1) binds to all selectins, albeit with very different affinities. Whereas P- and L-selectin require additional interactions contributed by sulfate groups for high binding affinity, E-selectin can functionally bind sLex-modified glycolipids and glycoproteins. Rivipansel (3) marked the first pan-selectin antagonist, which simultaneously interacted with both the sLex and the sulfate binding site. The aim of this contribution was to improve the pan-selectin affinity of rivipansel (3) by leveraging a new class of sLex mimetics in combination with an optimized linker length to the sulfate bearing group. As a result, the pan-selectin antagonist 11b exhibits an approximatively 5-fold improved affinity for E-, as well as P-selectin.
Collapse
Affiliation(s)
- Beatrice Wagner
- University of Basel, Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Martin Smieško
- University of Basel, Department of Pharmaceutical Sciences, Group Computational Pharmacy, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Roman P Jakob
- University of Basel, Department Biozentrum, Structural Area Focal Biology, Spitalstrasse 41, 4056, Basel, Switzerland
| | - Tobias Mühlethaler
- University of Basel, Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Jonathan Cramer
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Tim Maier
- University of Basel, Department Biozentrum, Structural Area Focal Biology, Spitalstrasse 41, 4056, Basel, Switzerland
| | - Said Rabbani
- University of Basel, Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Oliver Schwardt
- University of Basel, Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Beat Ernst
- University of Basel, Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| |
Collapse
|
19
|
Jordan C, Siebold K, Priegue P, Seeberger PH, Gilmour R. A Fluorinated Sialic Acid Vaccine Lead Against Meningitis B and C. J Am Chem Soc 2024; 146:15366-15375. [PMID: 38768956 PMCID: PMC11157539 DOI: 10.1021/jacs.4c03179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024]
Abstract
Inspired by the specificity of α-(2,9)-sialyl epitopes in bacterial capsular polysaccharides (CPS), a doubly fluorinated disaccharide has been validated as a vaccine lead against Neisseria meningitidis serogroups C and/or B. Emulating the importance of fluorine in drug discovery, this molecular editing approach serves a multitude of purposes, which range from controlling α-selective chemical sialylation to mitigating competing elimination. Conjugation of the disialoside with two carrier proteins (CRM197 and PorA) enabled a semisynthetic vaccine to be generated; this was then investigated in six groups of six mice. The individual levels of antibodies formed were compared and classified as highly glycan-specific and protective. All glycoconjugates induced a stable and long-term IgG response and binding to the native CPS epitope was achieved. The generated antibodies were protective against MenC and/or MenB; this was validated in vitro by SBA and OPKA assays. By merging the fluorinated glycan epitope of MenC with an outer cell membrane protein of MenB, a bivalent vaccine against both serogroups was created. It is envisaged that validation of this synthetic, fluorinated disialoside bioisostere as a potent antigen will open new therapeutic avenues.
Collapse
Affiliation(s)
- Christina Jordan
- Institute
for Organic Chemistry, University of Münster, Corrensstraße 36, Münster 48149, Germany
| | - Kathrin Siebold
- Institute
for Organic Chemistry, University of Münster, Corrensstraße 36, Münster 48149, Germany
| | - Patricia Priegue
- Department
of Biomolecular Systems, Max Planck Institute
for Colloids and Interfaces, Am Mühlenberg 1, Potsdam 14476, Germany
- Freie
Universität Berlin, Institute of
Chemistry and Biochemistry, Arnimallee 22, Berlin 14195, Germany
| | - Peter H. Seeberger
- Department
of Biomolecular Systems, Max Planck Institute
for Colloids and Interfaces, Am Mühlenberg 1, Potsdam 14476, Germany
- Freie
Universität Berlin, Institute of
Chemistry and Biochemistry, Arnimallee 22, Berlin 14195, Germany
| | - Ryan Gilmour
- Institute
for Organic Chemistry, University of Münster, Corrensstraße 36, Münster 48149, Germany
| |
Collapse
|
20
|
Kizer ME, R. Dwyer J. Editors' Choice-Perspective-Deciphering the Glycan Kryptos by Solid-State Nanopore Single-Molecule Sensing: A Call for Integrated Advancements Across Glyco- and Nanopore Science. ECS SENSORS PLUS 2024; 3:020604. [PMID: 38799647 PMCID: PMC11125560 DOI: 10.1149/2754-2726/ad49b0] [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: 01/31/2024] [Revised: 05/07/2024] [Indexed: 05/29/2024]
Abstract
Glycans, or complex carbohydrates, are information-rich biopolymers critical to many biological processes and with considerable importance in pharmaceutical therapeutics. Our understanding, though, is limited compared to other biomolecules such as DNA and proteins. The greater complexity of glycan structure and the limitations of conventional chemical analysis methods hinder glycan studies. Auspiciously, nanopore single-molecule sensors-commercially available for DNA sequencing-hold great promise as a tool for enabling and advancing glycan analysis. We focus on two key areas to advance nanopore glycan characterization: molecular surface coatings to enhance nanopore performance including by molecular recognition, and high-quality glycan chemical standards for training.
Collapse
Affiliation(s)
- Megan E. Kizer
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States of America
| | - Jason R. Dwyer
- Department of Chemistry, University of Rhode Island, Kingston, Rhode Island, 02881, United States of America
| |
Collapse
|
21
|
Lefèbre J, Falk T, Ning Y, Rademacher C. Secondary Sites of the C-type Lectin-Like Fold. Chemistry 2024; 30:e202400660. [PMID: 38527187 DOI: 10.1002/chem.202400660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 03/27/2024]
Abstract
C-type lectins are a large superfamily of proteins involved in a multitude of biological processes. In particular, their involvement in immunity and homeostasis has rendered them attractive targets for diverse therapeutic interventions. They share a characteristic C-type lectin-like domain whose adaptability enables them to bind a broad spectrum of ligands beyond the originally defined canonical Ca2+-dependent carbohydrate binding. Together with variable domain architecture and high-level conformational plasticity, this enables C-type lectins to meet diverse functional demands. Secondary sites provide another layer of regulation and are often intricately linked to functional diversity. Located remote from the canonical primary binding site, secondary sites can accommodate ligands with other physicochemical properties and alter protein dynamics, thus enhancing selectivity and enabling fine-tuning of the biological response. In this review, we outline the structural determinants allowing C-type lectins to perform a large variety of tasks and to accommodate the ligands associated with it. Using the six well-characterized Ca2+-dependent and Ca2+-independent C-type lectin receptors DC-SIGN, langerin, MGL, dectin-1, CLEC-2 and NKG2D as examples, we focus on the characteristics of non-canonical interactions and secondary sites and their potential use in drug discovery endeavors.
Collapse
Affiliation(s)
- Jonathan Lefèbre
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Torben Falk
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Yunzhan Ning
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| |
Collapse
|
22
|
Taj S, Ashfaq UA, Ahmad M, Noor H, Ikram A, Ahmed R, Tariq M, Masoud MS, Hasan A. The antihyperglycemic potential of pyrazolobenzothiazine 1, 1-dioxide novel derivative in mice using integrated molecular pharmacological approach. Sci Rep 2024; 14:7746. [PMID: 38565861 PMCID: PMC10987501 DOI: 10.1038/s41598-023-49932-2] [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: 07/11/2023] [Accepted: 12/13/2023] [Indexed: 04/04/2024] Open
Abstract
Diabetes Mellitus is a metabolic disease characterized by elevated blood sugar levels caused by inadequate insulin production, which subsequently leads to hyperglycemia. This study was aimed to investigate the antidiabetic potential of pyrazolobenzothiazine derivatives in silico, in vitro, and in vivo. Molecular docking of pyrazolobenzothiazine derivatives was performed against α-glucosidase and α-amylase and compounds were selected based on docking score, bonding interactions and low root mean square deviation (RMSD). Enzyme inhibition assay against α-glucosidase and α-amylase was performed in vitro using p-nitrophenyl-α-D-glucopyranoside (PNPG) and starch substrate. Synthetic compound pyrazolobenzothiazine (S1) exhibited minimal conformational changes during the 100 ns MD simulation run. S1 also revealed effective IC50 values for α-glucosidase (3.91 µM) and α-amylase (8.89 µM) and an enzyme kinetic study showed low ki (- 0.186 µM, - 1.267 µM) and ki' (- 0.691 µM, - 1.78 µM) values with the competitive type of inhibition for both enzymes α-glucosidase and α-amylase, respectively. Moreover, studies were conducted to check the effect of the synthetic compound in a mouse model. A low necrosis rate was observed in the liver, kidney, and pancreas through histology analysis performed on mice. Compound S1 also exhibited a good biochemical profile with lower sugar level (110-115 mg/dL), increased insulin level (25-30 μM/L), and low level of cholesterol (85 mg/dL) and creatinine (0.6 mg/dL) in blood. The treated mice group also exhibited a low % of glycated haemoglobin (3%). This study concludes that S1 is a new antidiabetic-agent that helps lower blood glucose levels and minimizes the complications associated with type-II diabetes.
Collapse
Affiliation(s)
- Saman Taj
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Matloob Ahmad
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Hasnat Noor
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Ayesha Ikram
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Rashid Ahmed
- Department of Biotechnology, Faculty of Natural and Applied Sciences, Mirpur University of Science and Technology, New Mirpur City, 10250, Pakistan
| | - Muhammad Tariq
- Department of Biotechnology, Faculty of Natural and Applied Sciences, Mirpur University of Science and Technology, New Mirpur City, 10250, Pakistan
| | - Muhammad Shareef Masoud
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan.
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, 2713, Doha, Qatar.
- Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| |
Collapse
|
23
|
Mishra VK, Khanna A, Tiwari G, Tyagi R, Sagar R. Recent developments on the synthesis of biologically active glycohybrids. Bioorg Chem 2024; 145:107172. [PMID: 38340475 DOI: 10.1016/j.bioorg.2024.107172] [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: 11/29/2023] [Revised: 01/11/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
The exploration of hybridization emerges as a potent tool in advancing drug discovery research, with a significant emphasis on carbohydrate-containing hybrid scaffolds. Evidence indicates that linking carbohydrate molecules to privileged bioactive scaffolds enhances the bioactivity of drug molecules. This synergy results in a diverse range of activities, making carbohydrate scaffolds pivotal for synthesizing compound libraries with significant functional and structural diversity. Beyond their synthesis utility, these scaffolds offer applications in screening bioactive molecules, presenting alternative avenues for drug development. This comprehensive review spanning 2015 to 2023 focuses on synthesized glycohybrid molecules, revealing their bioactivity in areas such as anti-microbial, anti-cancer, anti-diabetic, anti-inflammatory activities, enzyme inhibition and pesticides. Numerous novel glycohybrids surpass positive control drugs in biological activity. This focused study not only highlights the diverse bioactivities of glycohybrids but also underscores their promising role in innovative drug development strategies.
Collapse
Affiliation(s)
- Vinay Kumar Mishra
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005
| | - Ashish Khanna
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005
| | - Ghanshyam Tiwari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005
| | - Rajdeep Tyagi
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, 110067 New Delhi
| | - Ram Sagar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005; Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, 110067 New Delhi.
| |
Collapse
|
24
|
Cramer J, Pero B, Jiang X, Bosko C, Silbermann M, Rabbani S, Wilke S, Nemli DD, Ernst B, Peczuh MW. Does size matter? - Comparing pyranoses with septanoses as ligands of the bacterial lectin FimH. Eur J Med Chem 2024; 268:116225. [PMID: 38367495 PMCID: PMC10964925 DOI: 10.1016/j.ejmech.2024.116225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/27/2024] [Accepted: 02/07/2024] [Indexed: 02/19/2024]
Abstract
The pharmacological modulation of disease-relevant carbohydrate-protein interactions represents an underexplored area of medicinal chemistry. One particular challenge in the design of glycomimetic compounds is the inherent instability of the glycosidic bond toward enzymatic cleavage. This problem has traditionally been approached by employing S-, N-, or C-glycosides with reduced susceptibility toward glycosidases. The application of ring-extended glycomimetics is an innovative approach to circumvent this issue. On the example of the bacterial adhesin FimH, it was explored how design principles from pyranose glycomimetics transfer to analogous septanose structures. A series of ring-extended FimH antagonists exhibiting the well-proven pharmacophore necessary for targeting the tyrosine-gate of FimH was synthesized. The resulting septanoses were evaluated for their affinity to the conformationally rigid isolated lectin domain of FimH (FimHLD), as well as a structurally flexible full-length FimH (FimHFL) construct. Some elements of potent mannoside-based FimH antagonists could be successfully transferred to septanose-based ligands, ultimately resulting in a 32-fold increase in binding affinity. Interestingly, the canonical ca. 100-fold loss of binding affinity between FimHLD and FimHFL is partly mitigated by the more flexible septanose antagonists, hinting at potentially differing interaction features of the flexible glycomimetics with intermediately populated states during the conformational transition of FimHFL.
Collapse
Affiliation(s)
- Jonathan Cramer
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland; Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Bryant Pero
- Department of Chemistry, University of Connecticut, 55 North Eagleville Road, U3060, Storrs, CT, 06269, USA
| | - Xiaohua Jiang
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Cristin Bosko
- Department of Chemistry, University of Connecticut, 55 North Eagleville Road, U3060, Storrs, CT, 06269, USA
| | - Marleen Silbermann
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Said Rabbani
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Sebastian Wilke
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Dilara D Nemli
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Beat Ernst
- Molecular Pharmacy Group, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Mark W Peczuh
- Department of Chemistry, University of Connecticut, 55 North Eagleville Road, U3060, Storrs, CT, 06269, USA.
| |
Collapse
|
25
|
Bourgery C, Mendoza DJ, Garnier G, Mouterde LMM, Allais F. Immobilization of Adenosine Derivatives onto Cellulose Nanocrystals via Click Chemistry for Biocatalysis Applications. ACS APPLIED MATERIALS & INTERFACES 2024; 16:11315-11323. [PMID: 38394235 DOI: 10.1021/acsami.3c19025] [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: 02/25/2024]
Abstract
Adenosine triphosphate (ATP) is a central molecule of organisms and is involved in many biological processes. It is also widely used in biocatalytic processes, especially as a substrate and precursor of many cofactors─such as nicotinamide adenine dinucleotide phosphate (NADP(H)), coenzyme A (CoA), and S-adenosylmethionine (SAM). Despite its great scientific interest and pivotal role, its use in industrial processes is impeded by its prohibitory cost. To overcome this limitation, we developed a greener synthesis of adenosine derivatives and efficiently selectively grafted them onto organic nanoparticles. In this study, cellulose nanocrystals were used as a model combined with click chemistry via a copper-catalyzed azide/alkyne cycloaddition reaction (CuAAC). The grafted adenosine triphosphate derivative fully retains its biocatalytic capability, enabling heterobiocatalysis for modern biochemical processes.
Collapse
Affiliation(s)
- Célestin Bourgery
- URD Agro-Biotechnologies Industrielles (ABI), CEBB, AgroParisTech, Pomacle 51110, France
| | - David Joram Mendoza
- Bioresource Processing Research Institute of Australia (BioPRIA), Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Gil Garnier
- URD Agro-Biotechnologies Industrielles (ABI), CEBB, AgroParisTech, Pomacle 51110, France
- Bioresource Processing Research Institute of Australia (BioPRIA), Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Louis M M Mouterde
- URD Agro-Biotechnologies Industrielles (ABI), CEBB, AgroParisTech, Pomacle 51110, France
| | - Florent Allais
- URD Agro-Biotechnologies Industrielles (ABI), CEBB, AgroParisTech, Pomacle 51110, France
- Bioresource Processing Research Institute of Australia (BioPRIA), Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
26
|
Bretagne D, Pâris A, Matthews D, Fougère L, Burrini N, Wagner GK, Daniellou R, Lafite P. "Mix and match" auto-assembly of glycosyltransferase domains delivers biocatalysts with improved substrate promiscuity. J Biol Chem 2024; 300:105747. [PMID: 38354783 PMCID: PMC10937113 DOI: 10.1016/j.jbc.2024.105747] [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: 07/21/2023] [Revised: 01/25/2024] [Accepted: 02/08/2024] [Indexed: 02/16/2024] Open
Abstract
Glycosyltransferases (GT) catalyze the glycosylation of bioactive natural products, including peptides and proteins, flavonoids, and sterols, and have been extensively used as biocatalysts to generate glycosides. However, the often narrow substrate specificity of wild-type GTs requires engineering strategies to expand it. The GT-B structural family is constituted by GTs that share a highly conserved tertiary structure in which the sugar donor and acceptor substrates bind in dedicated domains. Here, we have used this selective binding feature to design an engineering process to generate chimeric glycosyltransferases that combine auto-assembled domains from two different GT-B enzymes. Our approach enabled the generation of a stable dimer with broader substrate promiscuity than the parent enzymes that were related to relaxed interactions between domains in the dimeric GT-B. Our findings provide a basis for the development of a novel class of heterodimeric GTs with improved substrate promiscuity for applications in biotechnology and natural product synthesis.
Collapse
Affiliation(s)
- Damien Bretagne
- Institut de Chimie Organique et Analytique (ICOA), UMR 7311 CNRS-Université d'Orléans, Université d'Orléans, Orléans Cedex 2, France; School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast, United Kingdom
| | - Arnaud Pâris
- Institut de Chimie Organique et Analytique (ICOA), UMR 7311 CNRS-Université d'Orléans, Université d'Orléans, Orléans Cedex 2, France
| | - David Matthews
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast, United Kingdom
| | - Laëtitia Fougère
- Institut de Chimie Organique et Analytique (ICOA), UMR 7311 CNRS-Université d'Orléans, Université d'Orléans, Orléans Cedex 2, France
| | - Nastassja Burrini
- Institut de Chimie Organique et Analytique (ICOA), UMR 7311 CNRS-Université d'Orléans, Université d'Orléans, Orléans Cedex 2, France
| | - Gerd K Wagner
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast, United Kingdom
| | - Richard Daniellou
- Institut de Chimie Organique et Analytique (ICOA), UMR 7311 CNRS-Université d'Orléans, Université d'Orléans, Orléans Cedex 2, France; Chaire de Cosmétologie, AgroParisTech, Orléans, France; Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
| | - Pierre Lafite
- Institut de Chimie Organique et Analytique (ICOA), UMR 7311 CNRS-Université d'Orléans, Université d'Orléans, Orléans Cedex 2, France.
| |
Collapse
|
27
|
Yue S, Ding G, Zheng Y, Song C, Xu P, Yu B, Li J. Dimethyl sulfate and diisopropyl sulfate as practical and versatile O-sulfation reagents. Nat Commun 2024; 15:1861. [PMID: 38424087 PMCID: PMC10904734 DOI: 10.1038/s41467-024-46214-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/19/2024] [Indexed: 03/02/2024] Open
Abstract
O-Sulfation is a vital post-translational modification in bioactive molecules, yet there are significant challenges with their synthesis. Dialkyl sulfates, such as dimethyl sulfate and diisopropyl sulfate are commonly used as alkylation agents in alkaline conditions, and result in the formation of sulfate byproducts. We report herein a general and robust approach to O-sulfation by harnessing the tunable reactivity of dimethyl sulfate or diisopropyl sulfate under tetrabutylammonium bisulfate activation. The versatility of this O-sulfation protocol is interrogated with a diverse range of alcohols, phenols and N-OH compounds, including carbohydrates, amino acids and natural products. The enhanced electrophilicity of the sulfur atom in dialkyl sulfates, facilitated by the interaction with bisulfate anion (HSO4-), accounts for this pioneering chemical reactivity. We envision that our method will be useful for application in the comprehension of biological functions and discovery of drugs.
Collapse
Affiliation(s)
- Shuaishuai Yue
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Guoping Ding
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, P. R. China
- Key Laboratory of Structure-based Drug Design & Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Ye Zheng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Chunlan Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China.
| | - Peng Xu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, P. R. China.
| | - Biao Yu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, P. R. China
| | - Jiakun Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China.
| |
Collapse
|
28
|
Zhou X, Wang Z, Yuan K. The effect of diet and nutrition on T cell function in cancer. Int J Cancer 2023; 153:1954-1966. [PMID: 37504380 DOI: 10.1002/ijc.34668] [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: 04/27/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023]
Abstract
Cancer can be considered one of the most threatening diseases to human health, and immunotherapy, especially T-cell immunotherapy, is the most promising treatment for cancers. Diet therapy is widely concerned in cancer because of its safety and fewer side effects. Many studies have shown that both the function of T cells and the progression of cancer can be affected by nutrients in the diet. In fact, it is challenging for T cells to infiltrate and eliminate cancer cells in tumor microenvironment, because of the harsh metabolic condition. The intake of different nutrients has a great influence on the proliferation, activation, differentiation and exhaustion of T cells. In this review, we summarize the effects of typical amino acids, lipids, carbohydrates and other nutritional factors on T cell functions and provide future perspectives for dietary treatment of cancer based on modifications of T cell functions.
Collapse
Affiliation(s)
- Xinyi Zhou
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen Wang
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kefei Yuan
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Wang X, Xiao G. Recent chemical synthesis of plant polysaccharides. Curr Opin Chem Biol 2023; 77:102387. [PMID: 37716049 DOI: 10.1016/j.cbpa.2023.102387] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/18/2023]
Abstract
Here, chemical syntheses of long, branched and complex glycans over 10-mer from plants are summarized, which highlights amylopectin 20-mer from starch, 17-mer from carthamus tinctorius, α-glucan 30-mer from Longan, 19-mer from psidium guajava and 11-mer from dendrobium huoshanense. The glycans assembly strategies, protecting groups utilization and glycosylation methods discussed here will inspire the efficient synthesis of diverse complex glycans with many 1,2-cis glycosidic linkages.
Collapse
Affiliation(s)
- Xiufang Wang
- Department of Chemistry, Kunming University, 2 Puxing Road, Kunming 650214, China
| | - Guozhi Xiao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 132 Lanhei Road, Kunming 650201, China.
| |
Collapse
|
30
|
Xie J, Wu S, Liao W, Ning J, Ding K. Src is a target molecule of mannose against pancreatic cancer cells growth in vitro & in vivo. Glycobiology 2023; 33:766-783. [PMID: 37658770 DOI: 10.1093/glycob/cwad070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 09/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant cancer with limited treatment options. Mannose, a common monosaccharide taken up by cells through the same transporters as glucose, has been shown to induce growth retardation and enhance cell death in response to chemotherapy in several cancers, including PDAC. However, the molecular targets and mechanisms underlying mannose's action against PDAC are not well understood. In this study, we used an integrative approach of network pharmacology, bioinformatics analysis, and experimental verification to investigate the pharmacological targets and mechanisms of mannose against PDAC. Our results showed that the protein Src is a key target of mannose in PDAC. Additionally, computational analysis revealed that mannose is a highly soluble compound that meets Lipinski's rule of five and that the expression of its target molecules is correlated with survival rates and prognosis in PDAC patients. Finally, we validated our findings through in vitro and in vivo experiments. In conclusion, our study provides evidence that mannose plays a critical role in inhibiting PDAC growth by targeting Src, suggesting that it may be a promising therapeutic candidate for PDAC.
Collapse
Affiliation(s)
- Jianhao Xie
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Pudong New district, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
| | - Shengjie Wu
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Pudong New district, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
| | - Wenfeng Liao
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Pudong New district, Shanghai 201203, China
| | - Jingru Ning
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Pudong New district, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Qixia District, Nanjing 210023, China
| | - Kan Ding
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Pudong New district, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| |
Collapse
|
31
|
Li C, Chen Z, Guo XY, Wen LR, Li M, Zhang LB. SO 42- ions as a nucleophilic reagent: straightforward electrochemical access to organosulfates. Chem Commun (Camb) 2023; 59:12164-12167. [PMID: 37743839 DOI: 10.1039/d3cc03960a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
An electrooxidation direct difunctionalization of alkynes with sulfonyl hydrazides has been developed for the construction of sulfonyl alkenyl sulfates in the absence of metal catalysts and a stoichiometric amount of oxidants. Notably, it is the first example to verify that SO42- ions can act as a nucleophilic reagent for the preparation of organosulfates.
Collapse
Affiliation(s)
- Chen Li
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Zhuo Chen
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Xue-Yang Guo
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Li-Rong Wen
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Ming Li
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Lin-Bao Zhang
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| |
Collapse
|
32
|
Fittolani G, Tyrikos-Ergas T, Poveda A, Yu Y, Yadav N, Seeberger PH, Jiménez-Barbero J, Delbianco M. Synthesis of a glycan hairpin. Nat Chem 2023; 15:1461-1469. [PMID: 37400598 PMCID: PMC10533408 DOI: 10.1038/s41557-023-01255-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 05/26/2023] [Indexed: 07/05/2023]
Abstract
The primary sequence of a biopolymer encodes the essential information for folding, permitting to carry out sophisticated functions. Inspired by natural biopolymers, peptide and nucleic acid sequences have been designed to adopt particular three-dimensional (3D) shapes and programmed to exert specific functions. In contrast, synthetic glycans capable of autonomously folding into defined 3D conformations have so far not been explored owing to their structural complexity and lack of design rules. Here we generate a glycan that adopts a stable secondary structure not present in nature, a glycan hairpin, by combining natural glycan motifs, stabilized by a non-conventional hydrogen bond and hydrophobic interactions. Automated glycan assembly enabled rapid access to synthetic analogues, including site-specific 13C-labelled ones, for nuclear magnetic resonance conformational analysis. Long-range inter-residue nuclear Overhauser effects unequivocally confirmed the folded conformation of the synthetic glycan hairpin. The capacity to control the 3D shape across the pool of available monosaccharides has the potential to afford more foldamer scaffolds with programmable properties and functions.
Collapse
Affiliation(s)
- Giulio Fittolani
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Theodore Tyrikos-Ergas
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Ana Poveda
- CICbioGUNE, Basque Research and Technology Alliance, Derio, Spain
| | - Yang Yu
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Simpson Querrey Institute, Northwestern University, Evanston, IL, USA
| | - Nishu Yadav
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Peter H Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Jesús Jiménez-Barbero
- CICbioGUNE, Basque Research and Technology Alliance, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Department of Organic Chemistry II, Faculty of Science and Technology, University of the Basque Country, Leioa, Spain
- Centro de Investigación Biomedica en Red de Enfermedades Respiratorias, Madrid, Spain
| | - Martina Delbianco
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany.
| |
Collapse
|
33
|
Ji H, Yang X, Zhou H, Cui F, Zhou Q. Rapid Evaluation of Antibacterial Carbohydrates on a Microfluidic Chip Integrated with the Impedimetric Neoglycoprotein Biosensor. BIOSENSORS 2023; 13:887. [PMID: 37754121 PMCID: PMC10526297 DOI: 10.3390/bios13090887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023]
Abstract
The colonization of some bacteria to their host cell is mediated by selective adhesion between adhesin and glycan. The evaluation of antiadhesive carbohydrates in vitro has great significance in discovering new antibacterial drugs. In this paper, a microfluidic chip integrated with impedimetric neoglycoprotein biosensors was developed to evaluate the antibacterial effect of carbohydrates. Mannosylated bovine serum albumin (Man-BSA) was taken as the neoglycoprotein and immobilized on the microelectrode-modified gold nanoparticles (Au NPs) to form a bionic glycoprotein nanosensing surface (Man-BSA/Au NPs). Salmonella typhimurium (S. typhimurium) was selected as a bacteria model owing to its selective adhesion to the mannose. Electrochemical impedance spectroscopy (EIS) was used to characterize the adhesion capacity of S. typhimurium to the Man-BSA/Au NPs and evaluate the antiadhesive efficacy of nine different carbohydrates. It was illustrated that the 4-methoxyphenyl-α-D-pyran mannoside (Phenyl-Man) and mannan peptide (Mannatide) showed excellent antiadhesive efficacy, with IC50 values of 0.086 mM and 0.094 mM, respectively. The microfluidic device developed in this study can be tested in multiple channels. Compared with traditional methods for evaluating the antibacterial drug in vitro, it has the advantages of being fast, convenient, and cost-effective.
Collapse
Affiliation(s)
| | | | | | - Feiyun Cui
- The Ministry of Education Key Laboratory of Clinical Diagnostics, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China; (H.J.); (X.Y.); (H.Z.)
| | - Qin Zhou
- The Ministry of Education Key Laboratory of Clinical Diagnostics, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China; (H.J.); (X.Y.); (H.Z.)
| |
Collapse
|
34
|
Wei X, Wang P, Liu F, Ye X, Xiong D. Drug Discovery Based on Fluorine-Containing Glycomimetics. Molecules 2023; 28:6641. [PMID: 37764416 PMCID: PMC10536126 DOI: 10.3390/molecules28186641] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Glycomimetics, which are synthetic molecules designed to mimic the structures and functions of natural carbohydrates, have been developed to overcome the limitations associated with natural carbohydrates. The fluorination of carbohydrates has emerged as a promising solution to dramatically enhance the metabolic stability, bioavailability, and protein-binding affinity of natural carbohydrates. In this review, the fluorination methods used to prepare the fluorinated carbohydrates, the effects of fluorination on the physical, chemical, and biological characteristics of natural sugars, and the biological activities of fluorinated sugars are presented.
Collapse
Affiliation(s)
- Xingxing Wei
- Department of Pharmacy, Changzhi Medical College, No. 161, Jiefang East Street, Changzhi 046012, China
| | - Pengyu Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Rd. No. 38, Beijing 100191, China (F.L.); (X.Y.)
| | - Fen Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Rd. No. 38, Beijing 100191, China (F.L.); (X.Y.)
| | - Xinshan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Rd. No. 38, Beijing 100191, China (F.L.); (X.Y.)
| | - Decai Xiong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Rd. No. 38, Beijing 100191, China (F.L.); (X.Y.)
| |
Collapse
|
35
|
Sladek V, Šmak P, Tvaroška I. How E-, L-, and P-Selectins Bind to sLe x and PSGL-1: A Quantification of Critical Residue Interactions. J Chem Inf Model 2023; 63:5604-5618. [PMID: 37486087 DOI: 10.1021/acs.jcim.3c00704] [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: 07/25/2023]
Abstract
Selectins and their ability to interact with specific ligands are a cornerstone in cell communication. Over the last three decades, a considerable wealth of experimental and molecular modeling insights into their structure and modus operandi were gathered. Nonetheless, explaining the role of individual selectin residues on a quantitative level remained elusive, despite its importance in understanding the structure-function relationship in these molecules and designing their inhibitors. This work explores essential interactions of selectin-ligand binding, employing a multiscale approach that combines molecular dynamics, quantum-chemical calculations, and residue interaction network models. Such an approach successfully reproduces most of the experimental findings. It proves to be helpful, with the potential for becoming an established tool for quantitative predictions of residue contribution to the binding of biomolecular complexes. The results empower us to quantify the importance of particular residues and functional groups in the protein-ligand interface and to pinpoint differences in molecular recognition by the three selectins. We show that mutations in the E-, L-, and P-selectins, e.g., different residues in positions 46, 85, 97, and 107, present a crucial difference in how the ligand is engaged. We assess the role of sulfation of tyrosine residues in PSGL-1 and suggest that TyrSO3- in position 51 interacting with Arg85 in P-selectin is a significant factor in the increased affinity of P-selectin to PSGL-1 compared to E- and L-selectins. We propose an original pharmacophore targeting five essential PSGL-binding sites based on the analysis of the selectin···PSGL-1 interactions.
Collapse
Affiliation(s)
- Vladimir Sladek
- Institute of Chemistry, SAS, Dubravska cesta 9, 84538 Bratislava, Slovakia
| | - Pavel Šmak
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Igor Tvaroška
- Institute of Chemistry, SAS, Dubravska cesta 9, 84538 Bratislava, Slovakia
| |
Collapse
|
36
|
Kumar S, Sahu RK, Kumari P, Maity J, Kumar B, Chhatwal RJ, Singh BK, Prasad AK. Efficient and stereoselective synthesis of sugar fused pyrano[3,2- c]pyranones as anticancer agents. RSC Adv 2023; 13:24604-24616. [PMID: 37601594 PMCID: PMC10436030 DOI: 10.1039/d3ra02371k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
A highly stereoselective, efficient and facile route was achieved for the synthesis of novel and biochemically potent sugar fused pyrano[3,2-c]pyranone derivatives starting from inexpensive, naturally occurring d-galactose and d-glucose. First, β-C-glycopyranosyl aldehydes were synthesized from these d-hexose sugars in six steps, with overall yields 41-55%. Next, two different 1-C-formyl glycals were synthesized from these β-C-glycopyranosyl aldehydes by treatment in basic conditions. The optimization of reaction conditions was carried out following reactions between 1-C-formyl galactal and 4-hydroxycoumarin. Next, 1-C-formyl galactal and 1-C-formyl glucal were treated with nine substituted 4-hydroxy coumarins at room temperature (25 °C) in ethyl acetate for ∼1-2 h in the presence of l-proline to obtain exclusively single diastereomers of pyrano[3,2-c]pyranone derivatives in excellent yields. Four compounds were found to be active for the MCF-7 cancer cell line. The MTT assay, apoptosis assay and migration analysis showed significant death of the cancer cells induced by the synthesized compounds.
Collapse
Affiliation(s)
- Sandeep Kumar
- Bioorganic Laboratory, Department of Chemistry, University of Delhi Delhi-110007 India
- Department of Chemistry, Ramjas College, University of Delhi Delhi-110007 India
| | - Ram Krishna Sahu
- National Institute of Cancer Prevention & Research Noida Uttar Pradesh India
| | - Priti Kumari
- Bioorganic Laboratory, Department of Chemistry, University of Delhi Delhi-110007 India
| | - Jyotirmoy Maity
- Department of Chemistry, St. Stephen College, University of Delhi Delhi-110007 India
| | - Binayak Kumar
- National Institute of Cancer Prevention & Research Noida Uttar Pradesh India
| | | | - Brajendra K Singh
- Bioorganic Laboratory, Department of Chemistry, University of Delhi Delhi-110007 India
| | - Ashok K Prasad
- Bioorganic Laboratory, Department of Chemistry, University of Delhi Delhi-110007 India
| |
Collapse
|
37
|
Simard RD, Joyal M, Beaugrand T, Gauthier J, Hardine E, Desriac A, Buffet CH, Prévost M, Nemer M, Guindon Y. Synthesis of Sialyl Lewis X Mimetics with E- and P-Selectin Binding Properties and Immunosuppressive Activity. J Org Chem 2023; 88:10974-10985. [PMID: 37449872 DOI: 10.1021/acs.joc.3c00956] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
E- and P-selectins are adhesion proteins implicated in immune cell recruitment at sites of infection, making them important drug targets for diseases involving excessive and uncontrolled inflammation. In this study, we developed an efficient strategy to synthesize bicyclic galactopyranosides through a key stereoselective equatorial C4-propiolate addition and TMSCN axial C-glycosidation. The nitrile group can then be converted to the carboxyl and different bioisosteres at a late stage in the synthesis, allowing for various derivatizations to potentially enhance biological activity. The sialyl LewisX glycomimetic featuring this rigidified bicyclic galactopyranoside moiety prevents neutrophil adhesion to endothelial cells in vitro by binding to both E- and P-selectins. We show here that the axial carboxyl analogue blocks immune cell recruitment in vivo, demonstrating its potential as an immunomodulator.
Collapse
Affiliation(s)
- Ryan D Simard
- Bioorganic Chemistry Laboratory, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
- Department of Chemistry, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Mathieu Joyal
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Thomas Beaugrand
- Bioorganic Chemistry Laboratory, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Julien Gauthier
- Bioorganic Chemistry Laboratory, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Elodie Hardine
- Bioorganic Chemistry Laboratory, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
- Department of Chemistry, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Axelle Desriac
- Bioorganic Chemistry Laboratory, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Charles-Henri Buffet
- Bioorganic Chemistry Laboratory, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Michel Prévost
- Bioorganic Chemistry Laboratory, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Mona Nemer
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Yvan Guindon
- Bioorganic Chemistry Laboratory, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
38
|
Ye G, Zhang J, Xu X, Zeng C, Ye Q, Wang Z. Comparative Analysis of Water-Soluble Polysaccharides from Dendrobium Second Love 'Tokimeki' and Dendrobium nobile in Structure, Antioxidant, and Anti-Tumor Activity In Vitro. Int J Mol Sci 2023; 24:10361. [PMID: 37373508 DOI: 10.3390/ijms241210361] [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: 05/29/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
With potential anti-tumor and antioxidant properties, the polysaccharide content of D. nobile is relatively lower than that of the other medicinal Dendrobium. To find high-content polysaccharide resources, the polysaccharide (DHPP-Ⅰs) was prepared from D. Second Love 'Tokimeki' (a D. nobile hybrid) and compared with DNPP-Ⅰs from D. nobile. DHPP-Is (Mn 31.09 kDa) and DNPP-Is (Mn 46.65 kDa) were found to be O-acetylated glucomannans (-Glcp-(1,4) and O-acetylated-D-Manp-(1,4) backbones), analogous to other Dendrobium polysaccharides. DHPP-Ⅰs had higher glucose content (31.1%) and a lower degree (0.16) of acetylation than DNPP-Ⅰs (15.8%, 0.28). Meanwhile, DHPP-Ⅰs and DNPP-Ⅰs had the same ability in the radical scavenging assay, which was milder than the control of Vc. Both DHPP-Is and DNPP-Is inhibited SPC-A-1 cell proliferation in vitro, with obvious differences in dose concentrations (0.5-2.0 mg/mL) and treatment times (24-72 h). Therefore, the antioxidant activity of DHPP-Ⅰs and DNPP-Ⅰs is not associated with distinction in anti-proliferative activity. As a glucomannan derived from non-medicinal Dendrobium, DHPP-Ⅰs has similar bioactivity to other medicinal Dendrobium, and this could serve as a starting point for studying the conformational-bioactivity relationship of Dendrobium polysaccharides.
Collapse
Affiliation(s)
- Guangying Ye
- Guangdong Key Lab of Ornamental Plant Germplasm Innovation and Utilization, Institute of Environmental Horticulture, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Jinhui Zhang
- Guangdong Key Lab of Ornamental Plant Germplasm Innovation and Utilization, Institute of Environmental Horticulture, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Xiaoli Xu
- Instrumental Analysis and Research Center, South China Agricultural University, Guangzhou 510642, China
| | - Canbiao Zeng
- Guangdong Key Lab of Ornamental Plant Germplasm Innovation and Utilization, Institute of Environmental Horticulture, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Qingsheng Ye
- Guangdong Province Key Lab for Biotechnology of Plant Development, College of Life Science, South China Normal University, Guangzhou 510631, China
| | - Zaihua Wang
- Guangdong Key Lab of Ornamental Plant Germplasm Innovation and Utilization, Institute of Environmental Horticulture, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| |
Collapse
|
39
|
Canner SW, Shanker S, Gray JJ. Structure-based neural network protein-carbohydrate interaction predictions at the residue level. FRONTIERS IN BIOINFORMATICS 2023; 3:1186531. [PMID: 37409346 PMCID: PMC10318439 DOI: 10.3389/fbinf.2023.1186531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/31/2023] [Indexed: 07/07/2023] Open
Abstract
Carbohydrates dynamically and transiently interact with proteins for cell-cell recognition, cellular differentiation, immune response, and many other cellular processes. Despite the molecular importance of these interactions, there are currently few reliable computational tools to predict potential carbohydrate-binding sites on any given protein. Here, we present two deep learning (DL) models named CArbohydrate-Protein interaction Site IdentiFier (CAPSIF) that predicts non-covalent carbohydrate-binding sites on proteins: (1) a 3D-UNet voxel-based neural network model (CAPSIF:V) and (2) an equivariant graph neural network model (CAPSIF:G). While both models outperform previous surrogate methods used for carbohydrate-binding site prediction, CAPSIF:V performs better than CAPSIF:G, achieving test Dice scores of 0.597 and 0.543 and test set Matthews correlation coefficients (MCCs) of 0.599 and 0.538, respectively. We further tested CAPSIF:V on AlphaFold2-predicted protein structures. CAPSIF:V performed equivalently on both experimentally determined structures and AlphaFold2-predicted structures. Finally, we demonstrate how CAPSIF models can be used in conjunction with local glycan-docking protocols, such as GlycanDock, to predict bound protein-carbohydrate structures.
Collapse
Affiliation(s)
- Samuel W. Canner
- Program in Molecular Biophysics, The Johns Hopkins University, Baltimore, MD, United States
| | - Sudhanshu Shanker
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Jeffrey J. Gray
- Program in Molecular Biophysics, The Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
40
|
Leusmann S, Ménová P, Shanin E, Titz A, Rademacher C. Glycomimetics for the inhibition and modulation of lectins. Chem Soc Rev 2023; 52:3663-3740. [PMID: 37232696 PMCID: PMC10243309 DOI: 10.1039/d2cs00954d] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Indexed: 05/27/2023]
Abstract
Carbohydrates are essential mediators of many processes in health and disease. They regulate self-/non-self- discrimination, are key elements of cellular communication, cancer, infection and inflammation, and determine protein folding, function and life-times. Moreover, they are integral to the cellular envelope for microorganisms and participate in biofilm formation. These diverse functions of carbohydrates are mediated by carbohydrate-binding proteins, lectins, and the more the knowledge about the biology of these proteins is advancing, the more interfering with carbohydrate recognition becomes a viable option for the development of novel therapeutics. In this respect, small molecules mimicking this recognition process become more and more available either as tools for fostering our basic understanding of glycobiology or as therapeutics. In this review, we outline the general design principles of glycomimetic inhibitors (Section 2). This section is then followed by highlighting three approaches to interfere with lectin function, i.e. with carbohydrate-derived glycomimetics (Section 3.1), novel glycomimetic scaffolds (Section 3.2) and allosteric modulators (Section 3.3). We summarize recent advances in design and application of glycomimetics for various classes of lectins of mammalian, viral and bacterial origin. Besides highlighting design principles in general, we showcase defined cases in which glycomimetics have been advanced to clinical trials or marketed. Additionally, emerging applications of glycomimetics for targeted protein degradation and targeted delivery purposes are reviewed in Section 4.
Collapse
Affiliation(s)
- Steffen Leusmann
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Petra Ménová
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
| | - Elena Shanin
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| |
Collapse
|
41
|
Arend LB, Verli H. Revisiting the structural basis for biological activity of GMI-1070, a sialyl Lewis x mimetic. Carbohydr Res 2023; 529:108829. [PMID: 37182470 DOI: 10.1016/j.carres.2023.108829] [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: 10/11/2022] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/16/2023]
Abstract
When it comes to the treatment of pathologies in which aberrant cell adhesion and extravasation from the bloodstream have been implicated, the selectins represent a central therapeutic target. In this context, the present work investigates the conformational landscape of two prototypes for the design of new antineoplasic and anti-inflammatory agents: the natural selectin ligand sialyl Lewisx and its mimetic GMI-1070. Accordingly, a series of unbiased molecular dynamics simulations at the microsecond scale using GROMOS 53A6 (GLYC), CHARMM36m and GLYCAM06 force fields were employed, together with ConfID, an analytical method for the characterization of conformational populations of small molecules. Our results for sialyl Lewisx are in agreement with and expand upon prior work. As for the mimetic, our results indicate that, in spite of its conformational restriction, GMI-1070's behavior in solution deviates from what had been proposed, highlighting thus some features that could be optimized, as the development of sialyl Lewisx mimetics continues, and new candidates emerge.
Collapse
Affiliation(s)
- Laís B Arend
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Av Bento Gonçalves, 9500, CP 15005, Porto Alegre, 91500-970, RS, Brazil
| | - Hugo Verli
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Av Bento Gonçalves, 9500, CP 15005, Porto Alegre, 91500-970, RS, Brazil.
| |
Collapse
|
42
|
Fernandes SRG, Mohajershojai T, Lundsten S, Sarmento B, Tomé JPC, Nestor M, Jha P. Photoactive immunoconjugates for targeted photodynamic therapy of cancer. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 243:112716. [PMID: 37126865 DOI: 10.1016/j.jphotobiol.2023.112716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/26/2023] [Accepted: 04/18/2023] [Indexed: 05/03/2023]
Abstract
Photodynamic therapy (PDT) has been used as an alternative or as a complement of conventional approaches for cancer treatment. In PDT, the reactive oxygen species (ROS) produced from the interaction between the photosensitizer (PS), visible light and molecular oxygen, kill malignant cells by triggering a cascade of cytotoxic reactions. In this process, the PS plays an extremely important role in the effectiveness of the therapy. In the present work, a new photoimmunoconjugate (PIC), based on cetuximab and the known third generation PS-glycophthalocyanine ZnPcGal4, was synthesized via reductive amination. The rationale behind this was the simultaneous cancer-associated specific targeting of PIC and photosensitization of targeted receptor positive cells. Varied reaction parameters and photodynamic conditions, such as PS concentrations and both type and intensities of light, were optimized. ZnPcGal4 showed significant photoactivity against EGFR expressing A431, EGFR-transfected HCT116 and HT29 cells when irradiated with white light of stronger intensity (38 mW/cm2). Similarly, the synthesized PICs-T1 and T2 also demonstrated photoactivity with high intensity white light. The best optimized PIC: sample 28 showed no precipitation and aggregation when inspected visually and analyzed through SE-HPLC. Fluorescence excitation of sample 28 and 125I-sample 28 radioconjugate (125I-PIC, 125I-radiolabeling yield ≥95%, determined with ITLC) at 660 nm showed presence of appended ZnPcGal4. In addition, simultaneous fluorescence and radioactivity detection of the 125I-PIC in serum and PBS (pH 7.4) for the longest incubated time point of 72 h, respectively, and superimposed signals thereof demonstrated ≥99% of loading and/or labeling yield, assuring overall stability of the PIC and corresponding PIC-radioconjugate w.r.t. both the appended ZnPcGal4 and bound-125I. Moreover, real-time binding analyses on EGFR-transfected HCT116 cells showed specific binding of 125I-PIC, suggesting no alternation in the binding kinetics of the mAb after appending it with ZnPcGal4. These results suggest dual potential applications of synthesized PICs both for PDT and radio-immunotherapy of cancer.
Collapse
Affiliation(s)
- Sara R G Fernandes
- Centro de Química Estrutural, Institute of Molecular Sciences & Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 751 85, Sweden
| | - Tabassom Mohajershojai
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 751 85, Sweden
| | - Sara Lundsten
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 751 85, Sweden; Ridgeview Instruments AB, Uppsala University, Uppsala 752 37, Sweden
| | - Bruno Sarmento
- INEB - Instituto Nacional de Engenharia Biomédica, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; CESPU, Instituto Universitário de Ciências da Saúde, Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal.
| | - João P C Tomé
- Centro de Química Estrutural, Institute of Molecular Sciences & Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal.
| | - Marika Nestor
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 751 85, Sweden.
| | - Preeti Jha
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 751 85, Sweden; Department of Medicinal Chemistry, Biomedical Centre, Uppsala University, Uppsala 75123, Sweden; Department of Radiology, University of Texas Southwestern Medical Centre, Dallas, TX 75390, United States.
| |
Collapse
|
43
|
Jdanova S, Taylor MS. Mechanistic Study of the Copper(II)-Mediated Site-Selective O-Arylation of Glycosides with Arylboronic Acids. J Org Chem 2023; 88:3487-3498. [PMID: 36888595 DOI: 10.1021/acs.joc.2c02693] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Glycosides having multiple free OH groups have been shown to undergo site-selective O-arylations in the presence of arylboronic acids and copper(II) acetate. Herein, a mechanistic analysis of these Chan-Evans-Lam-type couplings is presented based on reaction kinetics, mass spectrometric analysis of reaction mixtures, and substituent effect studies. The results establish that the formation of a substrate-derived boronic ester accelerates the rate-determining transmetalation step. Intramolecular transfer of the aryl group from the boronic ester is ruled out in favor of a pathway in which the key pre-transmetalation assembly is generated from a boronic ester, a copper complex, and a second equivalent of arylboronic acid.
Collapse
Affiliation(s)
- Sofia Jdanova
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6 Canada
| | - Mark S Taylor
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6 Canada
| |
Collapse
|
44
|
Canner SW, Shanker S, Gray JJ. Structure-Based Neural Network Protein-Carbohydrate Interaction Predictions at the Residue Level. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.531382. [PMID: 36993750 PMCID: PMC10054975 DOI: 10.1101/2023.03.14.531382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Carbohydrates dynamically and transiently interact with proteins for cell-cell recognition, cellular differentiation, immune response, and many other cellular processes. Despite the molecular importance of these interactions, there are currently few reliable computational tools to predict potential carbohydrate binding sites on any given protein. Here, we present two deep learning models named CArbohydrate-Protein interaction Site IdentiFier (CAPSIF) that predict carbohydrate binding sites on proteins: (1) a 3D-UNet voxel-based neural network model (CAPSIF:V) and (2) an equivariant graph neural network model (CAPSIF:G). While both models outperform previous surrogate methods used for carbohydrate binding site prediction, CAPSIF:V performs better than CAPSIF:G, achieving test Dice scores of 0.597 and 0.543 and test set Matthews correlation coefficients (MCCs) of 0.599 and 0.538, respectively. We further tested CAPSIF:V on AlphaFold2-predicted protein structures. CAPSIF:V performed equivalently on both experimentally determined structures and AlphaFold2 predicted structures. Finally, we demonstrate how CAPSIF models can be used in conjunction with local glycan-docking protocols, such as GlycanDock, to predict bound protein-carbohydrate structures.
Collapse
Affiliation(s)
- Samuel W Canner
- Program in Molecular Biophysics, The Johns Hopkins University, Baltimore, MD, United States of America
| | - Sudhanshu Shanker
- Dept. of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States of America
| | - Jeffrey J Gray
- Program in Molecular Biophysics, The Johns Hopkins University, Baltimore, MD, United States of America
- Dept. of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States of America
| |
Collapse
|
45
|
A Structural-Reporter Group to Determine the Core Conformation of Sialyl Lewisx Mimetics. Molecules 2023; 28:molecules28062595. [PMID: 36985569 PMCID: PMC10054758 DOI: 10.3390/molecules28062595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023] Open
Abstract
The d-GlcNAc moiety in sialyl Lewisx (sLex, 1) acts predominantly as a linker to position the d-Gal and the l-Fuc moieties in the bioactive spatial orientation. The hypothesis has been made that the NHAc group of GlcNAc pushes the fucose underneath the galactose and, thus, contributes to the stabilization of the bioactive conformation of the core of sLex (1). To test this hypothesis, GlcNAc mimetics consisting of (R,R)-1,2-cyclohexanediols substituted with alkyl and aryl substituents adjacent to the linking position of the fucose moiety were synthesized. To explore a broad range of extended and spatially demanding R-groups, an enzymatic approach for the synthesis of 3-alkyl/aryl-1,2-cyclohexanediols (3b-n) was applied. These cyclohexanediol derivatives were incorporated into the sLex mimetics 2b-n. For analyzing the relationship of affinity and core conformation, a 1H NMR structural-reporter-group concept was applied. Thus, the chemical shift of H-C5Fuc proved to be a sensitive indicator for the degree of pre-organization of the core of this class of sLex mimetics and therefore could be used to quantify the contribution of the R-groups.
Collapse
|
46
|
Chemical synthesis of oligosaccharides and their application in new drug research. Eur J Med Chem 2023; 249:115164. [PMID: 36758451 DOI: 10.1016/j.ejmech.2023.115164] [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: 12/16/2022] [Revised: 01/16/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023]
Abstract
Oligosaccharides are the ubiquitous molecules of life. In order to translate human bioglycosylation into clinical applications, homogeneous samples of oligosaccharides and glycoconjugates can be obtained by chemical, enzymatic or other biological methods for systematic studies. However, the structural complexity and diversity of glycans and their conjugates present a major challenge for the synthesis of such molecules. This review summarizes the chemical synthesis methods of oligosaccharides, the application of oligosaccharides in the field of medicinal chemistry according to their related biological activities, and shows the great prospect of oligosaccharides in the field of pharmaceutical chemistry.
Collapse
|
47
|
Glycomimetic Peptides as Therapeutic Tools. Pharmaceutics 2023; 15:pharmaceutics15020688. [PMID: 36840010 PMCID: PMC9966187 DOI: 10.3390/pharmaceutics15020688] [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: 12/23/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
The entry of peptides into glycobiology has led to the development of a unique class of therapeutic tools. Although numerous and well-known peptides are active as endocrine regulatory factors that bind to specific receptors, and peptides have been used extensively as epitopes for vaccine production, the use of peptides that mimic sugars as ligands of lectin-type receptors has opened a unique approach to modulate activity of immune cells. Ground-breaking work that initiated the use of peptides as tools for therapy identified sugar mimetics by screening phage display libraries. The peptides that have been discovered show significant potential as high-avidity, therapeutic tools when synthesized as multivalent structures. Advantages of peptides over sugars as drugs for immune modulation will be illustrated in this review.
Collapse
|
48
|
Joo Y, Sung JY, Shin SM, Park SJ, Kim KS, Park KD, Kim SB, Lee DW. A Retro-Aldol Reaction Prompted the Evolvability of a Phosphotransferase System for the Utilization of a Rare Sugar. Microbiol Spectr 2023; 11:e0366022. [PMID: 36786576 PMCID: PMC10101011 DOI: 10.1128/spectrum.03660-22] [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: 09/09/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
The evolution of the bacterial phosphotransferase system (PTS) linked to glycolysis is dependent on the availability of naturally occurring sugars. Although bacteria exhibit sugar specificities based on carbon catabolite repression, the acquisition and evolvability of the cellular sugar preference under conditions that are suboptimal for growth (e.g., environments rich in a rare sugar) are poorly understood. Here, we generated Escherichia coli mutants via a retro-aldol reaction to obtain progeny that can utilize the rare sugar d-tagatose. We detected a minimal set of adaptive mutations in the d-fructose-specific PTS to render E. coli capable of d-tagatose utilization. These E. coli mutant strains lost the tight regulation of both the d-fructose and N-acetyl-galactosamine PTS following deletions in the binding site of the catabolite repressor/activator protein (Cra) upstream from the fruBKA operon and in the agaR gene, encoding the N-acetylgalactosamine (GalNAc) repressor, respectively. Acquired d-tagatose catabolic pathways then underwent fine-tuned adaptation via an additional mutation in 1-phosphofructose kinase to adjust metabolic fluxes. We determined the evolutionary trajectory at the molecular level, providing insights into the mechanism by which enteric bacteria evolved a substrate preference for the rare sugar d-tagatose. Furthermore, the engineered E. coli mutant strain could serve as an in vivo high-throughput screening platform for engineering non-phosphosugar isomerases to produce rare sugars. IMPORTANCE Microorganisms generate energy through glycolysis, which might have preceded a rapid burst of evolution, including the evolution of cellular respiration in the primordial biosphere. However, little is known about the evolvability of cellular sugar preferences. Here, we generated Escherichia coli mutants via a retro-aldol reaction to obtain progeny that can utilize the rare sugar d-tagatose. Consequently, we identified mutational hot spots and determined the evolutionary trajectory at the molecular level. This provided insights into the mechanism by which enteric bacteria evolved substrate preferences for various sugars, accounting for the widespread occurrence of these taxa. Furthermore, the adaptive laboratory evolution-induced cellular chassis could serve as an in vivo high-throughput screening platform for engineering tailor-made non-phosphorylated sugar isomerases to produce low-calorigenic rare sugars showing antidiabetic, antihyperglycemic, and antitumor activities.
Collapse
Affiliation(s)
- Yunhye Joo
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jae-Yoon Sung
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sun-Mi Shin
- School of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - Sun Jun Park
- Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Kyoung Su Kim
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Ki Duk Park
- Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Seong-Bo Kim
- Bio-Living Engineering Major, Global Leaders College, Yonsei University, Seoul, Republic of Korea
| | - Dong-Woo Lee
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
49
|
Zahorska E, Rosato F, Stober K, Kuhaudomlarp S, Meiers J, Hauck D, Reith D, Gillon E, Rox K, Imberty A, Römer W, Titz A. Neutralizing the Impact of the Virulence Factor LecA from Pseudomonas aeruginosa on Human Cells with New Glycomimetic Inhibitors. Angew Chem Int Ed Engl 2023; 62:e202215535. [PMID: 36398566 PMCID: PMC10107299 DOI: 10.1002/anie.202215535] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022]
Abstract
Bacterial adhesion, biofilm formation and host cell invasion of the ESKAPE pathogen Pseudomonas aeruginosa require the tetravalent lectins LecA and LecB, which are therefore drug targets to fight these infections. Recently, we have reported highly potent divalent galactosides as specific LecA inhibitors. However, they suffered from very low solubility and an intrinsic chemical instability due to two acylhydrazone motifs, which precluded further biological evaluation. Here, we isosterically substituted the acylhydrazones and systematically varied linker identity and length between the two galactosides necessary for LecA binding. The optimized divalent LecA ligands showed improved stability and were up to 1000-fold more soluble. Importantly, these properties now enabled their biological characterization. The lead compound L2 potently inhibited LecA binding to lung epithelial cells, restored wound closure in a scratch assay and reduced the invasiveness of P. aeruginosa into host cells.
Collapse
Affiliation(s)
- Eva Zahorska
- Chemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123, Saarbrücken, Germany.,Department of Chemistry, Saarland University, 66123, Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-, Braunschweig, Germany
| | - Francesca Rosato
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
| | - Kai Stober
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
| | - Sakonwan Kuhaudomlarp
- Université Grenoble Alpes, CNRS, CERMAV, 38000, Grenoble, France.,Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Center for Excellence in Protein and Enzyme Technology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Joscha Meiers
- Chemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123, Saarbrücken, Germany.,Department of Chemistry, Saarland University, 66123, Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-, Braunschweig, Germany
| | - Dirk Hauck
- Chemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123, Saarbrücken, Germany.,Department of Chemistry, Saarland University, 66123, Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-, Braunschweig, Germany
| | - Dorina Reith
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
| | - Emilie Gillon
- Université Grenoble Alpes, CNRS, CERMAV, 38000, Grenoble, France
| | - Katharina Rox
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-, Braunschweig, Germany.,Department of Chemical Biology (CBIO), Helmholtz Centre for Infection Research (HZI), 38124, Braunschweig, Germany
| | - Anne Imberty
- Université Grenoble Alpes, CNRS, CERMAV, 38000, Grenoble, France
| | - Winfried Römer
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany.,Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, 79104, Freiburg, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123, Saarbrücken, Germany.,Department of Chemistry, Saarland University, 66123, Saarbrücken, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-, Braunschweig, Germany
| |
Collapse
|
50
|
Fragment-Based Lead Discovery Strategies in Antimicrobial Drug Discovery. Antibiotics (Basel) 2023; 12:antibiotics12020315. [PMID: 36830226 PMCID: PMC9951956 DOI: 10.3390/antibiotics12020315] [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: 01/09/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Fragment-based lead discovery (FBLD) is a powerful application for developing ligands as modulators of disease targets. This approach strategy involves identification of interactions between low-molecular weight compounds (100-300 Da) and their putative targets, often with low affinity (KD ~0.1-1 mM) interactions. The focus of this screening methodology is to optimize and streamline identification of fragments with higher ligand efficiency (LE) than typical high-throughput screening. The focus of this review is on the last half decade of fragment-based drug discovery strategies that have been used for antimicrobial drug discovery.
Collapse
|