1
|
Glycobiology of cellular expiry: Decrypting the role of glycan-lectin regulatory complex and therapeutic strategies focusing on cancer. Biochem Pharmacol 2023; 207:115367. [PMID: 36481348 DOI: 10.1016/j.bcp.2022.115367] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Often the outer leaflets of living cells bear a coat of glycosylated proteins, which primarily regulates cellular processes. Glycosylation of such proteins occurs as part of their post-translational modification. Within the endoplasmic reticulum, glycosylation enables the attachment of specific oligosaccharide moieties such as, 'glycan' to the transmembrane receptor proteins which confers precise biological information for governing the cell fate. The nature and degree of glycosylation of cell surface receptors are regulated by a bunch of glycosyl transferases and glycosidases which fine-tune attachment or detachment of glycan moieties. In classical death receptors, upregulation of glycosylation by glycosyl transferases is capable of inducing cell death in T cells, tumor cells, etc. Thus, any deregulated alternation at surface glycosylation of these death receptors can result in life-threatening disorder like cancer. In addition, transmembrane glycoproteins and lectin receptors can transduce intracellular signals for cell death execution. Exogenous interaction of lectins with glycan containing death receptors signals for cell death initiation by modulating downstream signalings. Subsequently, endogenous glycan-lectin interplay aids in the customization and implementation of the cell death program. Lastly, the glycan-lectin recognition system dictates the removal of apoptotic cells by sending accurate signals to the extracellular milieu. Since glycosylation has proven to be a biomarker of cellular death and disease progression; glycans serve as specific therapeutic targets of cancers. In this context, we are reviewing the molecular mechanisms of the glycan-lectin regulatory network as an integral part of cell death machinery in cancer to target them for successful therapeutic and clinical approaches.
Collapse
|
2
|
Jino Blessy J, Siva Shanmugam NR, Veluraja K, Michael Gromiha M. Investigations on the binding specificity of β-galactoside analogues with human galectin-1 using molecular dynamics simulations. J Biomol Struct Dyn 2022; 40:10094-10105. [PMID: 34219624 DOI: 10.1080/07391102.2021.1939788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Galectin-1 (Gal-1) is the first member of galectin family, which has a carbohydrate recognition domain, specifically binds towards β-galactoside containing oligosaccharides. Owing its association with carbohydrates, Gal-1 is involved in many biological processes such as cell signaling, adhesion and pathological pathways such as metastasis, apoptosis and increased tumour cell survival. The development of β-galactoside based inhibitors would help to control the Gal-1 expression. In the current study, we carried out molecular dynamics (MD) simulations to examine the structural and dynamic behaviour Gal-1-thiodigalactoside (TDG), Gal-1-lactobionic acid (LBA) and Gal-1-beta-(1→6)-galactobiose (G16G) complexes. The analysis of glycosidic torsional angles revealed that β-galactoside analogues TDG and LBA have a single binding mode (BM1) whereas G16G has two binding modes (BM1 and BM2) for interacting with Gal-1 protein. We have computed the binding free energies for the complexes Gal-1-TDG, Gal-1-LBA and Gal-1-G16G using MM/PBSA and are -6.45, -6.22 and -3.08 kcal/mol, respectively. This trend agrees well with experiments that the binding of Gal-1 with TDG is stronger than LBA. Further analysis revealed that the interactions due to direct and water-mediated hydrogen bonds play a significant role to the structural stability of the complexes. The result obtained from this study is useful to formulate a set of rules and derive pharmacophore-based features for designing inhibitors against galectin-1.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- J Jino Blessy
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - N R Siva Shanmugam
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - K Veluraja
- PSN college of Engineering and Technology, Tirunelveli, Tamilnadu, India
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
3
|
van Klaveren S, Dernovšek J, Jakopin Ž, Anderluh M, Leffler H, Nilsson UJ, Tomašič T. Design and synthesis of novel 3-triazolyl-1-thiogalactosides as galectin-1, -3 and -8 inhibitors. RSC Adv 2022; 12:18973-18984. [PMID: 35873334 PMCID: PMC9245910 DOI: 10.1039/d2ra03163a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/23/2022] [Indexed: 11/04/2022] Open
Abstract
Galectins are galactoside-binding proteins that play a role in various pathophysiological conditions, making them attractive targets in drug discovery. We have designed and synthesised a focused library of aromatic 3-triazolyl-1-thiogalactosides targeting their core site for binding of galactose and a subsite on its non-reducing side. Evaluation of their binding affinities for galectin-1, -3, and -8N identified acetamide-based compound 36 as a suitable compound for further affinity enhancement by adding groups at the reducing side of the galactose. Synthesis of its dichlorothiophenyl analogue 59 and the thiodigalactoside analogue 62 yielded promising pan-galectin inhibitors.
Collapse
Affiliation(s)
- Sjors van Klaveren
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry Aškerčeva cesta 7 1000 Ljubljana Slovenia
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University SE-221 00 Lund Sweden
| | - Jaka Dernovšek
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry Aškerčeva cesta 7 1000 Ljubljana Slovenia
| | - Žiga Jakopin
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry Aškerčeva cesta 7 1000 Ljubljana Slovenia
| | - Marko Anderluh
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry Aškerčeva cesta 7 1000 Ljubljana Slovenia
| | - Hakon Leffler
- Department of Laboratory Medicine, Section MIG, Lund University BMC-C1228b Klinikgatan 28 221 84 Lund Sweden
| | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University SE-221 00 Lund Sweden
| | - Tihomir Tomašič
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry Aškerčeva cesta 7 1000 Ljubljana Slovenia
| |
Collapse
|
4
|
Bum-Erdene K, Collins PM, Hugo MW, Tarighat SS, Fei F, Kishor C, Leffler H, Nilsson UJ, Groffen J, Grice ID, Heisterkamp N, Blanchard H. Novel Selective Galectin-3 Antagonists Are Cytotoxic to Acute Lymphoblastic Leukemia. J Med Chem 2022; 65:5975-5989. [DOI: 10.1021/acs.jmedchem.1c01296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Khuchtumur Bum-Erdene
- Institute for Glycomics, Griffith University, Gold Coast Campus, Queensland 4222, Australia
| | - Patrick M. Collins
- Institute for Glycomics, Griffith University, Gold Coast Campus, Queensland 4222, Australia
| | - Matthew W. Hugo
- Institute for Glycomics, Griffith University, Gold Coast Campus, Queensland 4222, Australia
| | - Somayeh S. Tarighat
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California 90027, United States
| | - Fei Fei
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California 90027, United States
| | - Chandan Kishor
- Institute for Glycomics, Griffith University, Gold Coast Campus, Queensland 4222, Australia
| | - Hakon Leffler
- Department of Laboratory Medicine, Section MIG, Lund University, BMC-C1228b, Klinikgatan 28, 221 84 Lund, Sweden
| | - Ulf. J. Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P.O. Box 124, 221 00 Lund, Sweden
| | - John Groffen
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California 90027, United States
| | - I. Darren Grice
- Institute for Glycomics, Griffith University, Gold Coast Campus, Queensland 4222, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast Campus, Queensland 4222, Australia
| | - Nora Heisterkamp
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California 90027, United States
| | - Helen Blanchard
- Institute for Glycomics, Griffith University, Gold Coast Campus, Queensland 4222, Australia
| |
Collapse
|
5
|
Convenient synthesis of long alkyl-chain triazolylglycosides using ionic liquid as dual promoter-solvent: Readily access to non-ionic triazolylglycoside surfactants for evaluation of cytotoxic activity. Tetrahedron Lett 2021. [DOI: 10.1016/j.tetlet.2021.153325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
6
|
Agrahari AK, Bose P, Jaiswal MK, Rajkhowa S, Singh AS, Hotha S, Mishra N, Tiwari VK. Cu(I)-Catalyzed Click Chemistry in Glycoscience and Their Diverse Applications. Chem Rev 2021; 121:7638-7956. [PMID: 34165284 DOI: 10.1021/acs.chemrev.0c00920] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Copper(I)-catalyzed 1,3-dipolar cycloaddition between organic azides and terminal alkynes, commonly known as CuAAC or click chemistry, has been identified as one of the most successful, versatile, reliable, and modular strategies for the rapid and regioselective construction of 1,4-disubstituted 1,2,3-triazoles as diversely functionalized molecules. Carbohydrates, an integral part of living cells, have several fascinating features, including their structural diversity, biocompatibility, bioavailability, hydrophilicity, and superior ADME properties with minimal toxicity, which support increased demand to explore them as versatile scaffolds for easy access to diverse glycohybrids and well-defined glycoconjugates for complete chemical, biochemical, and pharmacological investigations. This review highlights the successful development of CuAAC or click chemistry in emerging areas of glycoscience, including the synthesis of triazole appended carbohydrate-containing molecular architectures (mainly glycohybrids, glycoconjugates, glycopolymers, glycopeptides, glycoproteins, glycolipids, glycoclusters, and glycodendrimers through regioselective triazole forming modular and bio-orthogonal coupling protocols). It discusses the widespread applications of these glycoproducts as enzyme inhibitors in drug discovery and development, sensing, gelation, chelation, glycosylation, and catalysis. This review also covers the impact of click chemistry and provides future perspectives on its role in various emerging disciplines of science and technology.
Collapse
Affiliation(s)
- Anand K Agrahari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Priyanka Bose
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Manoj K Jaiswal
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Sanchayita Rajkhowa
- Department of Chemistry, Jorhat Institute of Science and Technology (JIST), Jorhat, Assam 785010, India
| | - Anoop S Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Srinivas Hotha
- Department of Chemistry, Indian Institute of Science and Engineering Research (IISER), Pune, Maharashtra 411021, India
| | - Nidhi Mishra
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Vinod K Tiwari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
7
|
Milenin SA, Drozdov FV, Bezlepkina KA, Majorov VY, Muzafarov AM. Acid-Catalyzed Rearrangement of Azidopropyl-Siloxane Monomers for the Synthesis of Azidopropyl-Polydimethylsiloxane and Their Carboxylic Acid Derivatives. Macromolecules 2021. [DOI: 10.1021/acs.macromol.0c02790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Sergey A. Milenin
- Enikolopov Institute of Synthetic Polymeric Materials, Russian Academy of Sciences, 70 Profsouznaya St., Moscow 117393, Russia
| | - Fedor V. Drozdov
- Enikolopov Institute of Synthetic Polymeric Materials, Russian Academy of Sciences, 70 Profsouznaya St., Moscow 117393, Russia
| | - Kseniya A. Bezlepkina
- Enikolopov Institute of Synthetic Polymeric Materials, Russian Academy of Sciences, 70 Profsouznaya St., Moscow 117393, Russia
| | - Valeriy Yu. Majorov
- Enikolopov Institute of Synthetic Polymeric Materials, Russian Academy of Sciences, 70 Profsouznaya St., Moscow 117393, Russia
| | - Aziz M. Muzafarov
- Enikolopov Institute of Synthetic Polymeric Materials, Russian Academy of Sciences, 70 Profsouznaya St., Moscow 117393, Russia
| |
Collapse
|
8
|
Miller MC, Zheng Y, Suylen D, Ippel H, Cañada FJ, Berbís MA, Jiménez-Barbero J, Tai G, Gabius HJ, Mayo KH. Targeting the CRD F-face of Human Galectin-3 and Allosterically Modulating Glycan Binding by Angiostatic PTX008 and a Structurally Optimized Derivative. ChemMedChem 2020; 16:713-723. [PMID: 33156953 DOI: 10.1002/cmdc.202000742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/31/2020] [Indexed: 12/25/2022]
Abstract
Calix[4]arene PTX008 is an angiostatic agent that inhibits tumor growth in mice by binding to galectin-1, a β-galactoside-binding lectin. To assess the affinity profile of PTX008 for galectins, we used 15 N,1 H HSQC NMR spectroscopy to show that PTX008 also binds to galectin-3 (Gal-3), albeit more weakly. We identified the contact site for PTX008 on the F-face of the Gal-3 carbohydrate recognition domain. STD NMR revealed that the hydrophobic phenyl ring crown of the calixarene is the binding epitope. With this information, we performed molecular modeling of the complex to assist in improving the rather low affinity of PTX008 for Gal-3. By removing the N-dimethyl alkyl chain amide groups, we produced PTX013 whose reduced alkyl chain length and polar character led to an approximately eightfold stronger binding than PTX008. PTX013 also binds Gal-1 more strongly than PTX008, whereas neither interacts strongly, if at all, with Gal-7. In addition, PTX013, like PTX008, is an allosteric inhibitor of galectin binding to the canonical ligand lactose. This study broadens the scope for galectin targeting by calixarene-based compounds and opens the perspective for selective galectin blocking.
Collapse
Affiliation(s)
- Michelle C Miller
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yi Zheng
- School of Life Science, Northeast Normal University, 130024, Changchun, People's Republic of China
| | - Dennis Suylen
- Department of Biochemistry and CARIM, Maastricht University, 6229HX, Maastricht, The Netherlands
| | - Hans Ippel
- Department of Biochemistry and CARIM, Maastricht University, 6229HX, Maastricht, The Netherlands
| | - F Javier Cañada
- NMR and Molecular Recognition Group, Centro de Investigaciones Biológicas Margarita Salas (CSIC), C/Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - M Alvaro Berbís
- NMR and Molecular Recognition Group, Centro de Investigaciones Biológicas Margarita Salas (CSIC), C/Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Jesús Jiménez-Barbero
- NMR and Molecular Recognition Group, Centro de Investigaciones Biológicas Margarita Salas (CSIC), C/Ramiro de Maeztu 9, 28040, Madrid, Spain.,CIC bioGUNE, Bizkaia Technological Park, Building 801 A, 48160, Derio, Spain.,Ikerbasque, Basque Foundation for Science, 28009, Bilbao, Spain
| | - Guihua Tai
- School of Life Science, Northeast Normal University, 130024, Changchun, People's Republic of China
| | - Hans-Joachim Gabius
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximillians-University, 80539, Munich, Germany
| | - Kevin H Mayo
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
9
|
New maleimide 1,2,3-triazole hybrids: design, synthesis, anticancer, and antimicrobial activities. MONATSHEFTE FUR CHEMIE 2020. [DOI: 10.1007/s00706-020-02685-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
10
|
Freichel T, Heine V, Laaf D, Mackintosh EE, Sarafova S, Elling L, Snyder NL, Hartmann L. Sequence-Defined Heteromultivalent Precision Glycomacromolecules Bearing Sulfonated/Sulfated Nonglycosidic Moieties Preferentially Bind Galectin-3 and Delay Wound Healing of a Galectin-3 Positive Tumor Cell Line in an In Vitro Wound Scratch Assay. Macromol Biosci 2020; 20:e2000163. [PMID: 32715650 PMCID: PMC9831253 DOI: 10.1002/mabi.202000163] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/28/2020] [Indexed: 01/12/2023]
Abstract
Within this work, a new class of sequence-defined heteromultivalent glycomacromolecules bearing lactose residues and nonglycosidic motifs for probing glycoconjugate recognition in carbohydrate recognition domain (CRD) of galectin-3 is presented. Galectins, a family of β-galactoside-binding proteins, are known to play crucial roles in different signaling pathways involved in tumor biology. Thus, research has focused on the design and synthesis of galectin-targeting ligands for use as diagnostic markers or potential therapeutics. Heteromultivalent precision glycomacromolecules have the potential to serve as ligands for galectins. In this work, multivalency and the introduction of nonglycosidic motifs bearing either neutral, amine, or sulfonated/sulfated groups are used to better understand binding in the galectin-3 CRD. Enzyme-linked immunosorbent assays and surface plasmon resonance studies are performed, revealing a positive impact of the sulfonated/sulfated nonglycosidic motifs on galectin-3 binding but not on galectin-1 binding. Selected compounds are then tested with galectin-3 positive MCF 7 breast cancer cells using an in vitro would scratch assay. Preliminary results demonstrate a differential biological effect on MCF 7 cells with high galectin-3 expression in comparison to an HEK 293 control with low galectin-3 expression, indicating the potential for sulfonated/sulfated heteromultivalent glycomacromolecules to serve as preferential ligands for galectin-3 targeting.
Collapse
Affiliation(s)
- Tanja Freichel
- Institute of Organic and Macromolecular Chemistry, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - Viktoria Heine
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstr. 20, Aachen 52074, Germany
| | - Dominic Laaf
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstr. 20, Aachen 52074, Germany
| | | | - Sophia Sarafova
- Department of Biology, Davidson College, Box 7188, Davidson, NC 28035, USA
| | - Lothar Elling
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstr. 20, Aachen 52074, Germany
| | - Nicole L. Snyder
- Department of Chemistry, Davidson College, Box 7120, Davidson, NC 28035, USA
| | - Laura Hartmann
- Institute of Organic and Macromolecular Chemistry Heinrich-Heine University Düsseldorf Universitätsstraße 1, Düsseldorf 40225, Germany
| |
Collapse
|
11
|
Sethi A, Sanam S, Munagalasetty S, Jayanthi S, Alvala M. Understanding the role of galectin inhibitors as potential candidates for SARS-CoV-2 spike protein: in silico studies. RSC Adv 2020; 10:29873-29884. [PMID: 35518264 PMCID: PMC9056307 DOI: 10.1039/d0ra04795c] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/05/2020] [Indexed: 12/21/2022] Open
Abstract
Galectin 3 have the potential to inhibit the SARS-CoV-2 spike protein. We validated the studies by docking, MD and MM/GBSA calculations.
Collapse
Affiliation(s)
- Aaftaab Sethi
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education & Research-Hyderabad
- Balanagar
- India
| | - Swetha Sanam
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education & Research-Hyderabad
- Balanagar
- India
| | - Sharon Munagalasetty
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education & Research-Hyderabad
- Balanagar
- India
| | - Sivaraman Jayanthi
- Computational Drug Design Lab
- School of Bio Sciences and Technology
- Vellore Institute of Technology
- Vellore
- India
| | - Mallika Alvala
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education & Research-Hyderabad
- Balanagar
- India
- MARS Training Academy
| |
Collapse
|
12
|
New clues arising from hunt of saccharides binding to galectin 3 via 3D QSAR and docking studies. INFORMATICS IN MEDICINE UNLOCKED 2020. [DOI: 10.1016/j.imu.2020.100411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
13
|
Goud NS, Soukya PSL, Ghouse M, Komal D, Alvala R, Alvala M. Human Galectin-1 and Its Inhibitors: Privileged Target for Cancer and HIV. Mini Rev Med Chem 2019; 19:1369-1378. [PMID: 30834831 DOI: 10.2174/1389557519666190304120821] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/07/2018] [Accepted: 02/22/2019] [Indexed: 01/18/2023]
Abstract
Galectin 1(Gal-1), a β-galactoside binding mammalian lectin of 14KDa, is implicated in many signalling pathways, immune responses associated with cancer progression and immune disorders. Inhibition of human Gal-1 has been regarded as one of the potential therapeutic approaches for the treatment of cancer, as it plays a major role in tumour development and metastasis by modulating various biological functions viz. apoptosis, angiogenesis, migration, cell immune escape. Gal-1 is considered as a biomarker in diagnosis, prognosis and treatment condition. The overexpression of Gal-1 is well established and seen in many types of cancer progression like osteosarcoma, breast, lung, prostate, melanoma, etc. Gal-1 greatly accelerates the binding kinetics of HIV-1 to susceptible cells, leading to faster viral entry and a more robust viral replication by specific binding of CD4 cells. Hence, the Gal-1 is considered a promising molecular target for the development of new therapeutic drugs for cancer and HIV. The present review laid emphasis on structural insights and functional role of Gal-1 in the disease, current Gal-1 inhibitors and future prospects in the design of specific Gal-1 inhibitors.
Collapse
Affiliation(s)
- Narella Sridhar Goud
- Department of Medicinal Chemistry, National Institute of pharmaceutical Education and Research (NIPER) - Hyderabad, Balanagar, 500037, India
| | - P S Lakshmi Soukya
- Department of Medicinal Chemistry, National Institute of pharmaceutical Education and Research (NIPER) - Hyderabad, Balanagar, 500037, India
| | - Mahammad Ghouse
- Department of Medicinal Chemistry, National Institute of pharmaceutical Education and Research (NIPER) - Hyderabad, Balanagar, 500037, India
| | - Daipule Komal
- Department of Medicinal Chemistry, National Institute of pharmaceutical Education and Research (NIPER) - Hyderabad, Balanagar, 500037, India
| | - Ravi Alvala
- G. Pulla Reddy College of pharmacy, Hyderabad, 500028, India
| | - Mallika Alvala
- Department of Medicinal Chemistry, National Institute of pharmaceutical Education and Research (NIPER) - Hyderabad, Balanagar, 500037, India
| |
Collapse
|
14
|
Dahlqvist A, Furevi A, Warlin N, Leffler H, Nilsson UJ. Stereo- and regioselective hydroboration of 1- exo-methylene pyranoses: discovery of aryltriazolylmethyl C-galactopyranosides as selective galectin-1 inhibitors. Beilstein J Org Chem 2019; 15:1046-1060. [PMID: 31164942 PMCID: PMC6541369 DOI: 10.3762/bjoc.15.102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/21/2019] [Indexed: 12/16/2022] Open
Abstract
Galectins are carbohydrate recognition proteins that bind carbohydrates containing galactose and are involved in cell signaling and cellular interactions, involving them in several diseases. We present the synthesis of (aryltriazolyl)methyl galactopyranoside galectin inhibitors using a highly diastereoselective hydroboration of C1-exo-methylene pyranosides giving inhibitors with fourfold or better selectivity for galectin-1 over galectin-3, -4C (C-terminal CRD), -4N (N-terminal CRD), -7, -8C, -8N, -9C, and -9N and dissociation constants down to 170 µM.
Collapse
Affiliation(s)
- Alexander Dahlqvist
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Box 124, SE-221 00 LUND, Sweden
| | - Axel Furevi
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Box 124, SE-221 00 LUND, Sweden
| | - Niklas Warlin
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Box 124, SE-221 00 LUND, Sweden
| | - Hakon Leffler
- Division of Microbiology, Immunology and Glycobiology, Lund University, BMC C12, SE-221 84 LUND, Sweden
| | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Box 124, SE-221 00 LUND, Sweden
| |
Collapse
|
15
|
Decoding the sweet regulation of apoptosis: the role of glycosylation and galectins in apoptotic signaling pathways. Cell Death Differ 2019; 26:981-993. [PMID: 30903104 DOI: 10.1038/s41418-019-0317-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/02/2019] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
Glycosylation and glycan-binding proteins such as galectins play an important role in the control of cell death signaling. Strikingly, very little attention has been given so far to the understanding of the molecular details behind this key regulatory network. Glycans attached to the death receptors such as CD95 and TRAIL-Rs, either alone or in a complex with galectins, might promote or inhibit apoptotic signals. However, we have just started to decode the functions of galectins in the modulation of extrinsic and intrinsic apoptosis. In this work, we have discussed the current understanding of the glycosylation-galectin regulatory network in CD95- as well as TRAIL-R-induced apoptosis and therapeutic strategies based on targeting galectins in cancer.
Collapse
|
16
|
Restuccia A, Fettis MM, Farhadi SA, Molinaro MD, Kane B, Hudalla GA. Evaluation of Self-Assembled Glycopeptide Nanofibers Modified with N,N′-Diacetyllactosamine for Selective Galectin-3 Recognition and Inhibition. ACS Biomater Sci Eng 2018. [DOI: 10.1021/acsbiomaterials.8b00611] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Antonietta Restuccia
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, PO Box 116131, Biomedical Sciences Building JG56, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Margaret M. Fettis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, PO Box 116131, Biomedical Sciences Building JG56, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Shaheen A. Farhadi
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, PO Box 116131, Biomedical Sciences Building JG56, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Matthew D. Molinaro
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, PO Box 116131, Biomedical Sciences Building JG56, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Bryant Kane
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, Florida 32611, United States
| | - Gregory A. Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, PO Box 116131, Biomedical Sciences Building JG56, 1275 Center Drive, Gainesville, Florida 32611, United States
| |
Collapse
|
17
|
Dings RPM, Miller MC, Griffin RJ, Mayo KH. Galectins as Molecular Targets for Therapeutic Intervention. Int J Mol Sci 2018; 19:ijms19030905. [PMID: 29562695 PMCID: PMC5877766 DOI: 10.3390/ijms19030905] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 02/06/2023] Open
Abstract
Galectins are a family of small, highly conserved, molecular effectors that mediate various biological processes, including chemotaxis and angiogenesis, and that function by interacting with various cell surface glycoconjugates, usually targeting β-galactoside epitopes. Because of their significant involvement in various biological functions and pathologies, galectins have become a focus of therapeutic discovery for clinical intervention against cancer, among other pathological disorders. In this review, we focus on understanding galectin structure-function relationships, their mechanisms of action on the molecular level, and targeting them for therapeutic intervention against cancer.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Michelle C Miller
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Kevin H Mayo
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
18
|
Zhang H, Laaf D, Elling L, Pieters RJ. Thiodigalactoside-Bovine Serum Albumin Conjugates as High-Potency Inhibitors of Galectin-3: An Outstanding Example of Multivalent Presentation of Small Molecule Inhibitors. Bioconjug Chem 2018; 29:1266-1275. [PMID: 29474087 PMCID: PMC5909177 DOI: 10.1021/acs.bioconjchem.8b00047] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
![]()
Galectin inhibitors
are urgently needed to understand the mode
of action and druggability of different galectins, but potent and
selective agents still evade researchers. Small-sized inhibitors based
on thiodigalactoside (TDG) have shown their potential while modifications
at their C3 position indicated a strategy to improve selectivity and
potency. Considering the role of galectins as glycoprotein traffic
police, involved in multivalent bridging interactions, we aimed to
create multivalent versions of the potent TDG inhibitors. We herein
present for the first time the multivalent attachment of a TDG derivative
using bovine serum albumin (BSA) as the scaffold. An efficient synthetic
method is presented to obtain a novel type of neoglycosylated proteins
loaded with different numbers of TDG moieties. A polyethylene glycol
(PEG)-spacer is introduced between the TDG and the protein scaffold
maintaining appropriate accessibility for an adequate galectin interaction.
The novel conjugates were evaluated in galectin binding and inhibition
studies in vitro. The conjugate with a moderate density
of 19 conjugated TDGs was identified as one of the most potent multivalent
Gal-3 inhibitors so far, with a clear demonstration of the benefit
of a multivalent ligand presentation. The described method may facilitate
the development of specific galectin inhibitors and their application
in biomedical research.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences , Utrecht University , Universiteitsweg 99 , 3584 CG Utrecht , The Netherlands
| | - Dominic Laaf
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering , RWTH Aachen University , Pauwelsstrasse 20 , 52074 Aachen , Germany
| | - Lothar Elling
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering , RWTH Aachen University , Pauwelsstrasse 20 , 52074 Aachen , Germany
| | - Roland J Pieters
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences , Utrecht University , Universiteitsweg 99 , 3584 CG Utrecht , The Netherlands
| |
Collapse
|
19
|
Peterson K, Kumar R, Stenström O, Verma P, Verma PR, Håkansson M, Kahl-Knutsson B, Zetterberg F, Leffler H, Akke M, Logan DT, Nilsson UJ. Systematic Tuning of Fluoro-galectin-3 Interactions Provides Thiodigalactoside Derivatives with Single-Digit nM Affinity and High Selectivity. J Med Chem 2018; 61:1164-1175. [PMID: 29284090 DOI: 10.1021/acs.jmedchem.7b01626] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Symmetrical and asymmetrical fluorinated phenyltriazolyl-thiodigalactoside derivatives have been synthesized and evaluated as inhibitors of galectin-1 and galectin-3. Systematic tuning of the phenyltriazolyl-thiodigalactosides' fluoro-interactions with galectin-3 led to the discovery of inhibitors with exceptional affinities (Kd down to 1-2 nM) in symmetrically substituted thiodigalactosides as well as unsurpassed combination of high affinity (Kd 7.5 nM) and selectivity (46-fold) over galectin-1 for asymmetrical thiodigalactosides by carrying one trifluorphenyltriazole and one coumaryl moiety. Studies of the inhibitor-galectin complexes with isothermal titration calorimetry and X-ray crystallography revealed the importance of fluoro-amide interaction for affinity and for selectivity. Finally, the high affinity of the discovered inhibitors required two competitive titration assay tools to be developed: a new high affinity fluorescent probe for competitive fluorescent polarization and a competitive ligand optimal for analyzing high affinity galectin-3 inhibitors with competitive isothermal titration calorimetry.
Collapse
Affiliation(s)
- Kristoffer Peterson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University , Box 124, SE-221 00 Lund, Sweden
| | - Rohit Kumar
- Biochemistry and Structural Biology, Center for Molecular Protein Science, Department of Chemistry, Lund University , Box 124, SE-221 00 Lund, Sweden
| | - Olof Stenström
- Biophysical Chemistry, Center for Molecular Protein Science, Department of Chemistry, Lund University , Box 124, SE-221 00 Lund, Sweden
| | - Priya Verma
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University , Box 124, SE-221 00 Lund, Sweden
| | - Prashant R Verma
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University , Box 124, SE-221 00 Lund, Sweden
| | - Maria Håkansson
- SARomics Biostructures AB , Medicon Village, SE-223 63 Lund, Sweden
| | - Barbro Kahl-Knutsson
- Department of Laboratory Medicine, Section MIG, Lund University BMC-C1228b , Klinikgatan 28, 221 84 Lund, Sweden
| | - Fredrik Zetterberg
- Galecto Biotech AB , Sahlgrenska Science Park, Medicinaregatan 8 A, SE-413 46 Gothenburg, Sweden
| | - Hakon Leffler
- Department of Laboratory Medicine, Section MIG, Lund University BMC-C1228b , Klinikgatan 28, 221 84 Lund, Sweden
| | - Mikael Akke
- Biophysical Chemistry, Center for Molecular Protein Science, Department of Chemistry, Lund University , Box 124, SE-221 00 Lund, Sweden
| | - Derek T Logan
- Biochemistry and Structural Biology, Center for Molecular Protein Science, Department of Chemistry, Lund University , Box 124, SE-221 00 Lund, Sweden.,SARomics Biostructures AB , Medicon Village, SE-223 63 Lund, Sweden
| | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University , Box 124, SE-221 00 Lund, Sweden
| |
Collapse
|
20
|
Koonce NA, Griffin RJ, Dings RPM. Galectin-1 Inhibitor OTX008 Induces Tumor Vessel Normalization and Tumor Growth Inhibition in Human Head and Neck Squamous Cell Carcinoma Models. Int J Mol Sci 2017; 18:ijms18122671. [PMID: 29232825 PMCID: PMC5751273 DOI: 10.3390/ijms18122671] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/01/2017] [Accepted: 12/05/2017] [Indexed: 12/23/2022] Open
Abstract
Galectin-1 is a hypoxia-regulated protein and a prognostic marker in head and neck squamous cell carcinomas (HNSCC). Here we assessed the ability of non-peptidic galectin-1 inhibitor OTX008 to improve tumor oxygenation levels via tumor vessel normalization as well as tumor growth inhibition in two human HNSCC tumor models, the human laryngeal squamous carcinoma SQ20B and the human epithelial type 2 HEp-2. Tumor-bearing mice were treated with OTX008, Anginex, or Avastin and oxygen levels were determined by fiber-optics and molecular marker pimonidazole binding. Immuno-fluorescence was used to determine vessel normalization status. Continued OTX008 treatment caused a transient reoxygenation in SQ20B tumors peaking on day 14, while a steady increase in tumor oxygenation was observed over 21 days in the HEp-2 model. A >50% decrease in immunohistochemical staining for tumor hypoxia verified the oxygenation data measured using a partial pressure of oxygen (pO2) probe. Additionally, OTX008 induced tumor vessel normalization as tumor pericyte coverage increased by approximately 40% without inducing any toxicity. Moreover, OTX008 inhibited tumor growth as effectively as Anginex and Avastin, except in the HEp-2 model where Avastin was found to suspend tumor growth. Galectin-1 inhibitor OTX008 transiently increased overall tumor oxygenation via vessel normalization to various degrees in both HNSCC models. These findings suggest that targeting galectin-1—e.g., by OTX008—may be an effective approach to treat cancer patients as stand-alone therapy or in combination with other standards of care.
Collapse
Affiliation(s)
- Nathan A Koonce
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
21
|
Biotinylated N-Acetyllactosamine- and N,N-Diacetyllactosamine-Based Oligosaccharides as Novel Ligands for Human Galectin-3. Bioengineering (Basel) 2017; 4:bioengineering4020031. [PMID: 28952509 PMCID: PMC5590477 DOI: 10.3390/bioengineering4020031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 03/28/2017] [Accepted: 03/31/2017] [Indexed: 12/19/2022] Open
Abstract
Galectin inhibitor design is an emerging research field due to the involvement of galectins in cancer. Galectin-3, in particular, plays an important role in tumor progression. To generate inhibitors, modifications of the glycan structure can be introduced. Conjugation of hydrophobic compounds to saccharides has proven to be promising as increased binding of galectin-3 can be observed. In the present study, we report on neo-glycans carrying hydrophobic biotin as novel ligands for human galectin-3. We modified N-acetyllactosamine- and N,N-diacetyllactosamine-based tetrasaccharides at the C6-position of the terminal saccharide unit using selective enzymatic oxidation and subsequent chemical conjugation of biotinamidohexanoic acid hydrazide. These neo-glycans were much better bound by galectin-3 than the unmodified counterparts. High selectivity for galectin-3 over galectin-1 was also proven. We generated multivalent neo-glycoproteins by conjugation of neo-glycans to bovine serum albumin showing high affinity for galectin-3. Compared to non-biotinylated neo-glycoproteins, we achieved high binding levels of galectin-3 with a lesser amount of conjugated neo-glycans. Multivalent ligand presentation of neo-glycoproteins significantly increased the inhibitory potency towards galectin-3 binding to asialofetuin when compared to free monovalent glycans. Our findings show the positive impact of 6-biotinylation of tetrasaccharides on galectin-3 binding, which broadens the recent design approaches for producing high-affinity ligands.
Collapse
|
22
|
Silva BL, Alves RJ, Speziali NL. Crystal structure of 4-nitro-phenyl 6- O-ethyl-β-d-galacto-pyran-oside monohydrate. Acta Crystallogr E Crystallogr Commun 2017; 73:598-601. [PMID: 28435729 PMCID: PMC5382630 DOI: 10.1107/s2056989017004595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/22/2017] [Indexed: 11/10/2022]
Abstract
The synthesis and crystal structure of the title compound, C14H19NO8·H2O, prepared in three steps from 6-O-ethyl-1,2;3,4-di-O-iso-propyl-idene-α-d-galacto-pyran-ose using protecting-group strategies employed in carbohydrate chemistry, is reported. The asymmetric unit consists of a single galactoside mol-ecule, in which the pyran-oid ring has a 4C1 conformation and the 4-nitro-phenyl moiety is essentially planar. In the crystal, each carbohydrate is surrounded by other d-galactose residues and water mol-ecules, linked by O-H⋯O hydrogen bonds involving all hy-droxy groups, giving a two-dimensional substructure lying parallel to (100) and extended into three dimensions by C-H⋯O inter-actions.
Collapse
Affiliation(s)
- Bruno Leonardo Silva
- Departamento de Produtos Farmacêuticos – Faculdade de Farmácia – Universidade Federal de Minas Gerais – Avenida Antônio Carlos 6627, Belo Horizonte MG, 31.270-901, Brazil
| | - Ricardo José Alves
- Departamento de Produtos Farmacêuticos – Faculdade de Farmácia – Universidade Federal de Minas Gerais – Avenida Antônio Carlos 6627, Belo Horizonte MG, 31.270-901, Brazil
| | - Nivaldo Lúcio Speziali
- Departamento de Física – Instituto de Ciências Exatas – Universidade Federal de Minas Gerais – Avenida Antônio Carlos 6627, Belo Horizonte MG, 31.270-901, Brazil
| |
Collapse
|
23
|
Dheer D, Singh V, Shankar R. Medicinal attributes of 1,2,3-triazoles: Current developments. Bioorg Chem 2017; 71:30-54. [PMID: 28126288 DOI: 10.1016/j.bioorg.2017.01.010] [Citation(s) in RCA: 542] [Impact Index Per Article: 77.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/06/2016] [Accepted: 01/15/2017] [Indexed: 02/01/2023]
Abstract
1,2,3-Triazoles are important five-membered heterocyclic scaffold due to their extensive biological activities. This framework can be readily obtained in good to excellent yields on the multigram scale through click chemistry via reaction of aryl/alkyl halides, alkynes and NaN3 under ambient conditions. It has been an emerging area of interest for many researchers throughout the globe owing to its immense pharmacological scope. The present work aims to summarize the current approaches adopted for the synthesis of the 1,2,3-triazole and medicinal significance of these architectures as a lead structure for the discovery of drug molecules such as COX-1/COX-2 inhibitors (celecoxib, pyrazofurin), HIV protease inhibitors, CB1 cannabinoid receptor antagonist and much more which are in the pipeline of clinical trials. The emphasis has been given on the major advancements in the medicinal prospectus of this pharmacophore for the period during 2008-2016.
Collapse
Affiliation(s)
- Divya Dheer
- Academy of Scientific and Innovative Research (AcSIR), CSIR-IIIM, Jammu Campus, Jammu 180001, India; Bio-organic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Virender Singh
- Department of Chemistry, National Institute of Technology (NIT), Jalandhar 144011, Punjab, India
| | - Ravi Shankar
- Academy of Scientific and Innovative Research (AcSIR), CSIR-IIIM, Jammu Campus, Jammu 180001, India; Bio-organic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India.
| |
Collapse
|
24
|
Mandal S, Rajput VK, Sundin AP, Leffler H, Mukhopadhyay B, Nilsson UJ. Galactose-amidine derivatives as selective antagonists of galectin-9. CAN J CHEM 2016. [DOI: 10.1139/cjc-2015-0598] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The family of galectin proteins involved in adhesion, growth regulation, immunity, and inflammatory events are important targets for development of small molecule antagonists. Here, N-sulfonyl amidine galactopyranoside derivatives obtained via a multicomponent reaction between galactose alkyne derivatives, sulfonyl azides, and amines were evaluated as antagonists of galectin-1, -2, -3, -4N (N-terminal domain), -4C (C-terminal domain), -8N, -9N, and -9C in a competitive fluorescence polarization assay. Highly selective compounds against galectin-9N with up to 30-fold improved affinity compared to the reference methyl β-d-galactopyranoside were identified. Molecular dynamics simulation suggested that the selectivity and affinity for galectin-9N originate from the N-sulfonyl amidine moieties forming tridentate hydrogen bonds to two asparagine side chains and one phenyl stacking edge-to-face to an arginine side chain. These selective galectin-9N antagonists are of significant value as chemical tools for studying galectin-9 biology and chemistry as well as possible starting structures for the discovery of galectin-9-targeting drugs influencing, e.g., immune regulation.
Collapse
Affiliation(s)
- Santanu Mandal
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, POB124, SE-22100 Lund, Sweden
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, Nadia 741252, India
| | - Vishal K. Rajput
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, POB124, SE-22100 Lund, Sweden
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, Nadia 741252, India
| | - Anders P. Sundin
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, POB124, SE-22100 Lund, Sweden
| | - Hakon Leffler
- Section MIG, Department of Laboratory Medicine, Lund University, BMC-C1228b, Klinikgatan 28, SE-221 84 Lund, Sweden
| | - Balaram Mukhopadhyay
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, Nadia 741252, India
| | - Ulf J. Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, POB124, SE-22100 Lund, Sweden
| |
Collapse
|
25
|
Delaine T, Collins P, MacKinnon A, Sharma G, Stegmayr J, Rajput VK, Mandal S, Cumpstey I, Larumbe A, Salameh BA, Kahl-Knutsson B, van Hattum H, van Scherpenzeel M, Pieters RJ, Sethi T, Schambye H, Oredsson S, Leffler H, Blanchard H, Nilsson UJ. Galectin-3-Binding Glycomimetics that Strongly Reduce Bleomycin-Induced Lung Fibrosis and Modulate Intracellular Glycan Recognition. Chembiochem 2016; 17:1759-70. [PMID: 27356186 DOI: 10.1002/cbic.201600285] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Indexed: 11/12/2022]
Abstract
Discovery of glycan-competitive galectin-3-binding compounds that attenuate lung fibrosis in a murine model and that block intracellular galectin-3 accumulation at damaged vesicles, hence revealing galectin-3-glycan interactions involved in fibrosis progression and in intracellular galectin-3 activities, is reported. 3,3'-Bis-(4-aryltriazol-1-yl)thiodigalactosides were synthesized and evaluated as antagonists of galectin-1, -2, -3, and -4 N-terminal, -4 C-terminal, -7 and -8 N-terminal, -9 N-terminal, and -9 C-terminal domains. Compounds displaying low-nanomolar affinities for galectins-1 and -3 were identified in a competitive fluorescence anisotropy assay. X-ray structural analysis of selected compounds in complex with galectin-3, together with galectin-3 mutant binding experiments, revealed that both the aryltriazolyl moieties and fluoro substituents on the compounds are involved in key interactions responsible for exceptional affinities towards galectin-3. The most potent galectin-3 antagonist was demonstrated to act in an assay monitoring galectin-3 accumulation upon amitriptyline-induced vesicle damage, visualizing a biochemically/medically relevant intracellular lectin-carbohydrate binding event and that it can be blocked by a small molecule. The same antagonist administered intratracheally attenuated bleomycin-induced pulmonary fibrosis in a mouse model with a dose/response profile comparing favorably with that of oral administration of the marketed antifibrotic compound pirfenidone.
Collapse
Affiliation(s)
- Tamara Delaine
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden
| | - Patrick Collins
- Institute for Glycomics, Griffith University, Gold Coast Campus, Queensland, 4222, Australia
| | - Alison MacKinnon
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - G Sharma
- Department of Laboratory Medicine, Section MIG, Lund University, BMC-C1228b, Klinikgatan 28, 221 84, Lund, Sweden
| | - John Stegmayr
- Department of Laboratory Medicine, Section MIG, Lund University, BMC-C1228b, Klinikgatan 28, 221 84, Lund, Sweden
| | - Vishal K Rajput
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden
| | - Santanu Mandal
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden
| | - Ian Cumpstey
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden
| | - Amaia Larumbe
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden
| | - Bader A Salameh
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden.,Chemistry Department, The Hashemite University, P. O. Box 150459, Zarka, 13115, Jordan
| | - Barbro Kahl-Knutsson
- Department of Laboratory Medicine, Section MIG, Lund University, BMC-C1228b, Klinikgatan 28, 221 84, Lund, Sweden
| | - Hilde van Hattum
- Department of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P. O. Box 80082, 3508 TB, Utrecht, Netherlands
| | - Monique van Scherpenzeel
- Department of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P. O. Box 80082, 3508 TB, Utrecht, Netherlands.,Translational Metabolic Laboratory, 51 Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, Netherlands
| | - Roland J Pieters
- Department of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P. O. Box 80082, 3508 TB, Utrecht, Netherlands
| | - Tariq Sethi
- Department of Respiratory Medicine and Allergy, Kings College, 41 Denmark Hill Campus, Bessemer Road, London, SE5 9RJ, UK
| | - Hans Schambye
- Galecto Biotech ApS, COBIS, Ole Maaloes vej 3, Copenhagen N, 2200, Denmark
| | - Stina Oredsson
- Department of Biology, Lund University, P. O. Box 118, 221 00, Lund, Sweden
| | - Hakon Leffler
- Department of Laboratory Medicine, Section MIG, Lund University, BMC-C1228b, Klinikgatan 28, 221 84, Lund, Sweden
| | - Helen Blanchard
- Institute for Glycomics, Griffith University, Gold Coast Campus, Queensland, 4222, Australia.
| | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, P. O. Box 124, 221 00, Lund, Sweden.
| |
Collapse
|
26
|
Blanchard H, Bum-Erdene K, Bohari MH, Yu X. Galectin-1 inhibitors and their potential therapeutic applications: a patent review. Expert Opin Ther Pat 2016; 26:537-54. [PMID: 26950805 DOI: 10.1517/13543776.2016.1163338] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Galectins have affinity for β-galactosides. Human galectin-1 is ubiquitously expressed in the body and its expression level can be a marker in disease. Targeted inhibition of galectin-1 gives potential for treatment of inflammatory disorders and anti-cancer therapeutics. AREAS COVERED This review discusses progress in galectin-1 inhibitor discovery and development. Patent applications pertaining to galectin-1 inhibitors are categorised as monovalent- and multivalent-carbohydrate-based inhibitors, peptides- and peptidomimetics. Furthermore, the potential of galectin-1 protein as a therapeutic is discussed along with consideration of the unique challenges that galectin-1 presents, including its monomer-dimer equilibrium and oxidized and reduced forms, with regard to delivering an intact protein to a pathologically relevant site. EXPERT OPINION Significant evidence implicates galectin-1's involvement in cancer progression, inflammation, and host-pathogen interactions. Conserved sequence similarity of the carbohydrate-binding sites of different galectins makes design of specific antagonists (blocking agents/inhibitors of function) difficult. Key challenges pertaining to the therapeutic use of galectin-1 are its monomer-dimer equilibrium, its redox state, and delivery of intact galectin-1 to the desired site. Developing modified forms of galectin-1 has resulted in increased stability and functional potency. Gene and protein therapy approaches that deliver the protein toward the target are under exploration as is exploitation of different inhibitor scaffolds.
Collapse
Affiliation(s)
- Helen Blanchard
- a Institute for Glycomics , Griffith University , Gold Coast Campus , Queensland , Australia
| | - Khuchtumur Bum-Erdene
- a Institute for Glycomics , Griffith University , Gold Coast Campus , Queensland , Australia
| | | | - Xing Yu
- a Institute for Glycomics , Griffith University , Gold Coast Campus , Queensland , Australia
| |
Collapse
|
27
|
Upadhyaya K, Hamidullah, Singh K, Arun A, Shukla M, Srivastava N, Ashraf R, Sharma A, Mahar R, Shukla SK, Sarkar J, Ramachandran R, Lal J, Konwar R, Tripathi RP. Identification of gallic acid based glycoconjugates as a novel tubulin polymerization inhibitors. Org Biomol Chem 2015; 14:1338-58. [PMID: 26659548 DOI: 10.1039/c5ob02113h] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A novel class of gallic acid based glycoconjugates were designed and synthesized as potential anticancer agents. Among all the compounds screened, compound 2a showed potent anticancer activity against breast cancer cells. The latter resulted in tubulin polymerization inhibition and induced G2/M cell cycle arrest, generation of reactive oxygen species, mitochondrial depolarization and subsequent apoptosis in breast cancer cells. In addition, ultraviolet-visible spectroscopy and fluorescence quenching studies of the compound with tubulin confirmed direct interaction of compounds with tubulin. Molecular modeling studies revealed that it binds at the colchicine binding site in tubulin. Further, 2a also exhibited potent in vivo anticancer activity in LA-7 syngeneic rat mammary tumor model. Current data projects its strong candidature to be developed as anticancer agent.
Collapse
Affiliation(s)
- Kapil Upadhyaya
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Marchiori MF, Pires Souto DE, Oliveira Bortot L, Francisco Pereira J, Kubota LT, Cummings RD, Dias-Baruffi M, Carvalho I, Campo VL. Synthetic 1,2,3-triazole-linked glycoconjugates bind with high affinity to human galectin-3. Bioorg Med Chem 2015; 23:3414-25. [DOI: 10.1016/j.bmc.2015.04.044] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 04/02/2015] [Accepted: 04/13/2015] [Indexed: 11/29/2022]
|
29
|
Cecioni S, Imberty A, Vidal S. Glycomimetics versus Multivalent Glycoconjugates for the Design of High Affinity Lectin Ligands. Chem Rev 2014; 115:525-61. [DOI: 10.1021/cr500303t] [Citation(s) in RCA: 381] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Samy Cecioni
- CERMAV, Université Grenoble Alpes and CNRS, BP 53, F-38041 Grenoble Cedex 9, France
- Institut
de Chimie et Biochimie Moléculaires et Supramoléculaires,
Laboratoire de Chimie Organique 2 - Glycochimie, UMR 5246, Université Lyon 1 and CNRS, 43 Boulevard du 11 Novembre 1918, F-69622, Villeurbanne, France
| | - Anne Imberty
- CERMAV, Université Grenoble Alpes and CNRS, BP 53, F-38041 Grenoble Cedex 9, France
| | - Sébastien Vidal
- Institut
de Chimie et Biochimie Moléculaires et Supramoléculaires,
Laboratoire de Chimie Organique 2 - Glycochimie, UMR 5246, Université Lyon 1 and CNRS, 43 Boulevard du 11 Novembre 1918, F-69622, Villeurbanne, France
| |
Collapse
|
30
|
Rajput VK, Leffler H, Nilsson UJ, Mukhopadhyay B. Synthesis and evaluation of iminocoumaryl and coumaryl derivatized glycosides as galectin antagonists. Bioorg Med Chem Lett 2014; 24:3516-20. [DOI: 10.1016/j.bmcl.2014.05.063] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/03/2014] [Accepted: 05/16/2014] [Indexed: 10/25/2022]
|
31
|
Rachel H, Chang-Chun L. Recent advances toward the development of inhibitors to attenuate tumor metastasis via the interruption of lectin-ligand interactions. Adv Carbohydr Chem Biochem 2014; 69:125-207. [PMID: 24274369 DOI: 10.1016/b978-0-12-408093-5.00005-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aberrant glycosylation is a well-recognized phenomenon that occurs on the surface of tumor cells, and the overexpression of a number of ligands (such as TF, sialyl Tn, and sialyl Lewis X) has been correlated to a worse prognosis for the patient. These unique carbohydrate structures play an integral role in cell-cell communication and have also been associated with more metastatic cancer phenotypes, which can result from binding to lectins present on cell surfaces. The most well studied metastasis-associated lectins are the galectins and selectins, which have been correlated to adhesion, neoangiogenesis, and immune-cell evasion processes. In order to slow the rate of metastatic lesion formation, a number of approaches have been successfully developed which involve interfering with the tumor lectin-substrate binding event. Through the generation of inhibitors, or by attenuating lectin and/or carbohydrate expression, promising results have been observed both in vitro and in vivo. This article briefly summarizes the involvement of lectins in the metastatic process and also describes different approaches used to prevent these undesirable carbohydrate-lectin binding events, which should ultimately lead to improvement in current cancer therapies.
Collapse
Affiliation(s)
- Hevey Rachel
- Alberta Glycomics Centre, Department of Chemistry, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
32
|
Blanchard H, Bum-Erdene K, Hugo MW. Inhibitors of Galectins and Implications for Structure-Based Design of Galectin-Specific Therapeutics. Aust J Chem 2014. [DOI: 10.1071/ch14362] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Galectins are a family of galactoside-specific lectins that are involved in a myriad of metabolic and disease processes. Due to roles in cancer and inflammatory and heart diseases, galectins are attractive targets for drug development. Over the last two decades, various strategies have been used to inhibit galectins, including polysaccharide-based therapeutics, multivalent display of saccharides, peptides, peptidomimetics, and saccharide-modifications. Primarily due to galectin carbohydrate binding sites having high sequence identities, the design and development of selective inhibitors targeting particular galectins, thereby addressing specific disease states, is challenging. Furthermore, the use of different inhibition assays by research groups has hindered systematic assessment of the relative selectivity and affinity of inhibitors. This review summarises the status of current inhibitors, strategies, and novel scaffolds that exploit subtle differences in galectin structures that, in conjunction with increasing available data on multiple galectins, is enabling the feasible design of effective and specific inhibitors of galectins.
Collapse
|
33
|
Berg R, Straub BF. Advancements in the mechanistic understanding of the copper-catalyzed azide-alkyne cycloaddition. Beilstein J Org Chem 2013; 9:2715-50. [PMID: 24367437 PMCID: PMC3869285 DOI: 10.3762/bjoc.9.308] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/30/2013] [Indexed: 12/21/2022] Open
Abstract
The copper-catalyzed azide-alkyne cycloaddition (CuAAC) is one of the most broadly applicable and easy-to-handle reactions in the arsenal of organic chemistry. However, the mechanistic understanding of this reaction has lagged behind the plethora of its applications for a long time. As reagent mixtures of copper salts and additives are commonly used in CuAAC reactions, the structure of the catalytically active species itself has remained subject to speculation, which can be attributed to the multifaceted aggregation chemistry of copper(I) alkyne and acetylide complexes. Following an introductory section on common catalyst systems in CuAAC reactions, this review will highlight experimental and computational studies from early proposals to very recent and more sophisticated investigations, which deliver more detailed insights into the CuAAC's catalytic cycle and the species involved. As diverging mechanistic views are presented in articles, books and online resources, we intend to present the research efforts in this field during the past decade and finally give an up-to-date picture of the currently accepted dinuclear mechanism of CuAAC. Additionally, we hope to inspire research efforts on the development of molecularly defined copper(I) catalysts with defined structural characteristics, whose main advantage in contrast to the regularly used precatalyst reagent mixtures is twofold: on the one hand, the characteristics of molecularly defined, well soluble catalysts can be tuned according to the particular requirements of the experiment; on the other hand, the understanding of the CuAAC reaction mechanism can be further advanced by kinetic studies and the isolation and characterization of key intermediates.
Collapse
Affiliation(s)
- Regina Berg
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
| | - Bernd F Straub
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
| |
Collapse
|
34
|
Mangsang W, Sirion U, Saeeng R. One-pot synthesis of O-glycosyl triazoles by O-glycosylation–click reaction. Carbohydr Res 2013; 375:79-89. [DOI: 10.1016/j.carres.2013.04.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 04/20/2013] [Accepted: 04/24/2013] [Indexed: 11/25/2022]
|
35
|
Porciúncula González C, Castilla A, Garófalo L, Soule S, Irazoqui G, Giacomini C. Enzymatic synthesis of 2-aminoethyl β-d-galactopyranoside catalyzed by Aspergillus oryzae β-galactosidase. Carbohydr Res 2013; 368:104-10. [DOI: 10.1016/j.carres.2012.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 11/29/2012] [Accepted: 12/07/2012] [Indexed: 11/27/2022]
|
36
|
Irazoqui G, Bustamante MJ, Castilla A, Villagrán LV, Batista-Viera F, Brena BM, Giacomini C. Substrate-like inhibition of the transgalactosylation reaction catalyzed by β-galactosidase fromAspergillus oryzae. BIOCATAL BIOTRANSFOR 2013. [DOI: 10.3109/10242422.2012.762575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Dings RPM, Kumar N, Miller MC, Loren M, Rangwala H, Hoye TR, Mayo KH. Structure-based optimization of angiostatic agent 6DBF7, an allosteric antagonist of galectin-1. J Pharmacol Exp Ther 2012; 344:589-99. [PMID: 23232447 DOI: 10.1124/jpet.112.199646] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Galectin-1 (gal-1), which binds β-galactoside groups on various cell surface receptors, is crucial to cell adhesion and migration, and is found to be elevated in several cancers. Previously, we reported on 6DBF7, a dibenzofuran (DBF)-based peptidomimetic of the gal-1 antagonist anginex. In the present study, we used a structure-based approach to optimize 6DBF7. Initial NMR studies showed that 6DBF7 binds to gal-1 on one side of the β-sandwich away from the lectin's carbohydrate binding site. Although an alanine scan of 6DBF7 showed that the two cationic groups (lysines) in the partial peptide are crucial to its angiostatic activity, it is the hydrophobic face of the amphipath that appears to interact directly with the surface of gal-1. Based on this structural information, we designed and tested additional DBF analogs. In particular, substitution of the C-terminal Asp for alanine and branched alkyl side chains (Val, Leu, Ile) for linear ones (Nle, Nva) rendered the greatest improvements in activity. Flow cytometry with gal-1(-/-) splenocytes showed that 6DBF7 and two of its more potent analogs (DB16 and DB21) can fully inhibit fluorescein isothiocyanate-gal-1 binding. Moreover, heteronuclear single-quantum coherence NMR titrations showed that the presence of DB16 decreases gal-1 affinity for lactose, indicating that the peptidomimetic targets gal-1 as a noncompetitive, allosteric inhibitor of glycan binding. Using tumor mouse models (B16F10 melanoma, LS174 lung, and MA148 ovarian), we found that DB21 inhibits tumor angiogenesis and tumor growth significantly better than 6DBF7, DB16, or anginex. DB21 is currently being developed further and holds promise for the management of human cancer in the clinic.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Biochemistry, University of Minnesota, 321 Church Street, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Dings RPM, Miller MC, Nesmelova I, Astorgues-Xerri L, Kumar N, Serova M, Chen X, Raymond E, Hoye TR, Mayo KH. Antitumor agent calixarene 0118 targets human galectin-1 as an allosteric inhibitor of carbohydrate binding. J Med Chem 2012; 55:5121-9. [PMID: 22575017 DOI: 10.1021/jm300014q] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Calix[4]arene compound 0118 is an angiostatic agent that inhibits tumor growth in mice. Although 0118 is a topomimetic of galectin-1-targeting angiostatic amphipathic peptide Anginex, we had yet to prove that 0118 targets galectin-1. Galectin-1 is involved in pathological disorders like tumor endothelial cell adhesion and migration and therefore presents a relevant target for therapeutic intervention against cancer. Here, (15)N-(1)H HSQC NMR spectroscopy demonstrates that 0118 indeed targets galectin-1 at a site away from the lectin's carbohydrate binding site and thereby attenuates lactose binding to the lectin. Flow cytometry and agglutination assays show that 0118 attenuates binding of galectin-1 to cell surface glycans, and the inhibition of cell proliferation by 0118 is found to be correlated with the cellular expression of the lectin. In general, our data indicate that 0118 targets galectin-1 as an allosteric inhibitor of glycan/carbohydrate binding. This work contributes to the clinical development of antitumor calixarene compound 0118.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota Health Sciences Center, 6-155 Jackson Hall, 321 Church Street, Minneapolis, Minnesota 55455, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Synthesis of novel 3-deoxy-3-C-triazolylmethyl-allose derivatives and evaluation of their biological activity. OPEN CHEM 2012. [DOI: 10.2478/s11532-012-0002-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
AbstractRecently, monosaccharide-triazole conjugates have proved to possess a large variety of useful biological activities. This paper describes synthesis of a new series of 3-deoxy-3-C-triazolylmethyl-allose derivatives. These new compounds are obtained from acetonide-protected 3-deoxy-3-azidomethyl allose and commercial alkynes via Cu(I) catalyzed 1,3-dipolar cycloaddition. The obtained molecular scaffolds differ from those described earlier by the presence of a methylene linker (-CH2-) between the C(3) of allose and the triazole moiety. It was demonstrated that acetonide-protected monosaccharide, 3-deoxy-3-C-(4-phenyl-1H-1,2,3-triazol-1-yl)methyl-1,2:5,6-di-O-isopropylidene-α-d-allofuranose, inhibited α-L-fucosidase for 26% at 0.1 mM concentration, but a deprotected analog, 3-deoxy-3-C-(4-(4-tert-butylphenyl)-1H-1,2,3-triazol-1-yl)methyl-β-d-allofuranose, showed 15% inhibition of β-glucosidase at 1 mM concentration.
Collapse
|
40
|
Kónya K, Fekete S, Abrahám A, Patonay T. α-Azido ketones. Part 7: synthesis of 1,4-disubstituted triazoles by the "click" reaction of various terminal acetylenes with phenacyl azides or α-azidobenzo(hetera)cyclanones. Mol Divers 2012; 16:91-102. [PMID: 22307767 DOI: 10.1007/s11030-012-9360-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Accepted: 01/09/2012] [Indexed: 11/24/2022]
Abstract
Copper(I)-catalyzed alkyne-azide 1,3-cycloaddition reaction (CuAAC, Sharpless-Meldal reaction) of various α-azido ketones such as substituted 2-azidoacetophenones, 2-azidobenzosuberone and 3-azido(thio)chromanones with terminal alkynes was studied. The reaction resulted in the formation of the expected 1,2,3-triazoles in moderate to good yields although the reactivity was somewhat lower than in the case of simple azides. Reaction of ethynylchromones as alkynes gave interesting dichromonoid systems bridged by a triazole unit.
Collapse
Affiliation(s)
- Krisztina Kónya
- Department of Organic Chemistry, University of Debrecen, P.O. Box 20, Debrecen, 4010, Hungary
| | | | | | | |
Collapse
|
41
|
Mayo KH. From Carbohydrate to Peptidomimetic Inhibitors of Galectins. ACS SYMPOSIUM SERIES 2012. [DOI: 10.1021/bk-2012-1115.ch003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Affiliation(s)
- Kevin H. Mayo
- Department of Biochemistry, Molecular Biology & Biophysics, 6-155 Jackson Hall, University of Minnesota, 321 Church Street, Minneapolis, Minnesota 55455
| |
Collapse
|
42
|
Leffler H, Nilsson UJ. Low-Molecular Weight Inhibitors of Galectins. ACS SYMPOSIUM SERIES 2012. [DOI: 10.1021/bk-2012-1115.ch002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Hakon Leffler
- Section MIG, Department of Laboratory Medicine, Lund University, Sölvegatan 23, SE-223 62, Lund, Sweden
- Center for Analysis and Synthesis, Lund University, POB 124, SE-221 00 Lund, Sweden
| | - Ulf J. Nilsson
- Section MIG, Department of Laboratory Medicine, Lund University, Sölvegatan 23, SE-223 62, Lund, Sweden
- Center for Analysis and Synthesis, Lund University, POB 124, SE-221 00 Lund, Sweden
| |
Collapse
|
43
|
Saraboji K, Håkansson M, Genheden S, Diehl C, Qvist J, Weininger U, Nilsson UJ, Leffler H, Ryde U, Akke M, Logan DT. The carbohydrate-binding site in galectin-3 is preorganized to recognize a sugarlike framework of oxygens: ultra-high-resolution structures and water dynamics. Biochemistry 2011; 51:296-306. [PMID: 22111949 PMCID: PMC3255464 DOI: 10.1021/bi201459p] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
![]()
The recognition of carbohydrates by proteins is a fundamental aspect of communication within and between living cells. Understanding the molecular basis of carbohydrate–protein interactions is a prerequisite for the rational design of synthetic ligands. Here we report the high- to ultra-high-resolution crystal structures of the carbohydrate recognition domain of galectin-3 (Gal3C) in the ligand-free state (1.08 Å at 100 K, 1.25 Å at 298 K) and in complex with lactose (0.86 Å) or glycerol (0.9 Å). These structures reveal striking similarities in the positions of water and carbohydrate oxygen atoms in all three states, indicating that the binding site of Gal3C is preorganized to coordinate oxygen atoms in an arrangement that is nearly optimal for the recognition of β-galactosides. Deuterium nuclear magnetic resonance (NMR) relaxation dispersion experiments and molecular dynamics simulations demonstrate that all water molecules in the lactose-binding site exchange with bulk water on a time scale of nanoseconds or shorter. Nevertheless, molecular dynamics simulations identify transient water binding at sites that agree well with those observed by crystallography, indicating that the energy landscape of the binding site is maintained in solution. All heavy atoms of glycerol are positioned like the corresponding atoms of lactose in the Gal3C complexes. However, binding of glycerol to Gal3C is insignificant in solution at room temperature, as monitored by NMR spectroscopy or isothermal titration calorimetry under conditions where lactose binding is readily detected. These observations make a case for protein cryo-crystallography as a valuable screening method in fragment-based drug discovery and further suggest that identification of water sites might inform inhibitor design.
Collapse
Affiliation(s)
- Kadhirvel Saraboji
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Box 124, Lund University, SE-221 00 Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Galectin-1-specific inhibitors as a new class of compounds to treat HIV-1 infection. Antimicrob Agents Chemother 2011; 56:154-62. [PMID: 22064534 DOI: 10.1128/aac.05595-11] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite significant improvements, antiretroviral therapies against HIV-1 are plagued by a high frequency of therapeutic failures that have been associated with acquisition of drug resistance. We recently reported that HIV-1 exploits a host glycan binding protein, galectin-1, to increase its attachment to host cells, thereby increasing its overall infectivity in susceptible cells. This finding suggests that host molecules such as galectin-1 could reduce the expected efficiency of HIV-1 drugs targeting early steps of the replicative cycle, such as attachment and entry processes. Thus, new classes of drugs that would interfere with galectin-1/HIV-1 interactions could benefit the current antiretroviral therapy. To further explore this possibility, experiments were conducted to discover leading compounds showing specific inhibition of galectin-1 activity in a cellular model of HIV-1 infection. Three lactoside compounds were found to modestly inhibit the interaction of galectin-1 with primary human CD4(+) T cells. Interestingly, these same inhibitors reduced the galectin-1-mediated increase in HIV-1 attachment to target cells in a much more efficient manner. More important, the tested lactoside derivatives also significantly decreased the galectin-1-dependent enhancement of HIV-1 infection. These observations deserve further attention when considering that the development of new drugs to prevent and treat HIV-1 infection remains a priority.
Collapse
|
45
|
Diehl C, Engström O, Delaine T, Håkansson M, Genheden S, Modig K, Leffler H, Ryde U, Nilsson UJ, Akke M. Protein flexibility and conformational entropy in ligand design targeting the carbohydrate recognition domain of galectin-3. J Am Chem Soc 2010; 132:14577-89. [PMID: 20873837 PMCID: PMC2954529 DOI: 10.1021/ja105852y] [Citation(s) in RCA: 189] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Indexed: 02/08/2023]
Abstract
Rational drug design is predicated on knowledge of the three-dimensional structure of the protein-ligand complex and the thermodynamics of ligand binding. Despite the fundamental importance of both enthalpy and entropy in driving ligand binding, the role of conformational entropy is rarely addressed in drug design. In this work, we have probed the conformational entropy and its relative contribution to the free energy of ligand binding to the carbohydrate recognition domain of galectin-3. Using a combination of NMR spectroscopy, isothermal titration calorimetry, and X-ray crystallography, we characterized the binding of three ligands with dissociation constants ranging over 2 orders of magnitude. (15)N and (2)H spin relaxation measurements showed that the protein backbone and side chains respond to ligand binding by increased conformational fluctuations, on average, that differ among the three ligand-bound states. Variability in the response to ligand binding is prominent in the hydrophobic core, where a distal cluster of methyl groups becomes more rigid, whereas methyl groups closer to the binding site become more flexible. The results reveal an intricate interplay between structure and conformational fluctuations in the different complexes that fine-tunes the affinity. The estimated change in conformational entropy is comparable in magnitude to the binding enthalpy, demonstrating that it contributes favorably and significantly to ligand binding. We speculate that the relatively weak inherent protein-carbohydrate interactions and limited hydrophobic effect associated with oligosaccharide binding might have exerted evolutionary pressure on carbohydrate-binding proteins to increase the affinity by means of conformational entropy.
Collapse
|
46
|
Zheng H, McDonald R, Hall DG. Boronic acid catalysis for mild and selective [3+2] dipolar cycloadditions to unsaturated carboxylic acids. Chemistry 2010; 16:5454-60. [PMID: 20373314 DOI: 10.1002/chem.200903484] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Herein, the concept of boronic acid catalysis (BAC) for the activation of unsaturated carboxylic acids is applied in several classic dipolar [3+2] cycloadditions involving azides, nitrile oxides, and nitrones as partners. These cycloadditions can be used to produce pharmaceutically interesting, small heterocyclic products, such as triazoles, isoxazoles, and isoxazolidines. These cycloadducts are formed directly and include a free carboxylic acid functionality that can be employed for further transformations, thereby avoiding prior masking or functionalization. In all cases, BAC provides faster reactions, under milder conditions, with much improved product yields and regioselectivities. In some instances, such as triazole formation from the reaction of azides with 2-alkynoic acids, catalysis with ortho-nitrophenylboronic acid circumvents the undesirable product decarboxylation observed when using thermal activation. By using NMR spectroscopic studies, the boronic acid catalyst was shown to provide activation by a LUMO-lowering effect in the unsaturated carboxylic acid, likely via a monoacylated hemiboronic ester intermediate.
Collapse
Affiliation(s)
- Hongchao Zheng
- Department of Chemistry, Gunning-Lemieux Chemistry Centre, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| | | | | |
Collapse
|
47
|
Salameh BA, Cumpstey I, Sundin A, Leffler H, Nilsson UJ. 1H-1,2,3-triazol-1-yl thiodigalactoside derivatives as high affinity galectin-3 inhibitors. Bioorg Med Chem 2010; 18:5367-78. [PMID: 20538469 DOI: 10.1016/j.bmc.2010.05.040] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 05/13/2010] [Accepted: 05/14/2010] [Indexed: 01/23/2023]
Abstract
Galactose C3-triazole derivatives were synthesized by Cu(I)-catalyzed cycloaddition between acetylenes and galactose C3-azido derivatives. Evaluation against galectin-3, 7, 8N (N-terminal) and 9N (N-terminal) revealed 1,4-disubstituted triazoles to be high-affinity inhibitors of galectin-3 with selectivity over galectin-7, 8N, and 9N. Conformational analysis of 1,4-di- and 1,4,5-tri-substituted galactose C3-triazoles suggested that a triazole C5-substituent interfered sterically with the galectin proteins, which explained their poor affinities compared to the corresponding 1,4-disubstituted triazoles. Introduction of two 1,4-disubstituted triazole moieties onto thiodigalactoside resulted in affinities down to 29 nM for galectin-3.
Collapse
Affiliation(s)
- Bader A Salameh
- Chemistry Department, The Hashemite University, PO Box 150459, Zarka 13115, Jordan
| | | | | | | | | |
Collapse
|
48
|
Nesmelova IV, Ermakova E, Daragan VA, Pang M, Menéndez M, Lagartera L, Solís D, Baum LG, Mayo KH. Lactose binding to galectin-1 modulates structural dynamics, increases conformational entropy, and occurs with apparent negative cooperativity. J Mol Biol 2010; 397:1209-30. [PMID: 20184898 DOI: 10.1016/j.jmb.2010.02.033] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 01/04/2010] [Accepted: 02/18/2010] [Indexed: 11/30/2022]
Abstract
Galectins are a family of lectins with a conserved carbohydrate recognition domain that interacts with beta-galactosides. By binding cell surface glycoconjugates, galectin-1 (gal-1) is involved in cell adhesion and migration processes and is an important regulator of tumor angiogenesis. Here, we used heteronuclear NMR spectroscopy and molecular modeling to investigate lactose binding to gal-1 and to derive solution NMR structures of gal-1 in the lactose-bound and unbound states. Structure analysis shows that the beta-strands and loops around the lactose binding site, which are more open and dynamic in the unbound state, fold in around the bound lactose molecule, dampening internal motions at that site and increasing motions elsewhere throughout the protein to contribute entropically to the binding free energy. CD data support the view of an overall more open structure in the lactose-bound state. Analysis of heteronuclear single quantum coherence titration binding data indicates that lactose binds the two carbohydrate recognition domains of the gal-1 dimer with negative cooperativity, in that the first lactose molecule binds more strongly (K(1)=21+/-6 x 10(3) M(-1)) than the second (K(2)=4+/-2 x 10(3) M(-1)). Isothermal calorimetry data fit using a sequential binding model present a similar picture, yielding K(1)=20+/-10 x 10(3) M(-1) and K(2)=1.67+/-0.07 x 10(3) M(-1). Molecular dynamics simulations provide insight into structural dynamics of the half-loaded lactose state and, together with NMR data, suggest that lactose binding at one site transmits a signal through the beta-sandwich and loops to the second binding site. Overall, our results provide new insight into gal-1 structure-function relationships and to protein-carbohydrate interactions in general.
Collapse
Affiliation(s)
- Irina V Nesmelova
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
van Scherpenzeel M, Moret EE, Ballell L, Liskamp RMJ, Nilsson UJ, Leffler H, Pieters RJ. Synthesis and Evaluation of New Thiodigalactoside-Based Chemical Probes to Label Galectin-3. Chembiochem 2009; 10:1724-33. [DOI: 10.1002/cbic.200900198] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
50
|
Synthesis of 3-azido-3-deoxy-β-d-galactopyranosides. Carbohydr Res 2009; 344:1282-4. [DOI: 10.1016/j.carres.2009.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Accepted: 05/07/2009] [Indexed: 01/25/2023]
|