1
|
Tripathi DK, Naidu G, Nagar N, Pramanik SD, Roy P, Kumar D, Poluri KM. Deciphering the molecular interactions between monocyte chemoattractant protein and its potential inhibitor suramin. Int J Biol Macromol 2025; 307:141903. [PMID: 40064271 DOI: 10.1016/j.ijbiomac.2025.141903] [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: 12/05/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Chemokines, in coordination with glycosaminoglycans (GAGs) and G protein-coupled receptors (GPCRs), play a critical role in regulating inflammatory responses. Among these, monocyte chemoattractant protein-1, also known as CCL2 stands out for its role in coordinating with other immune molecules to direct macrophage migration, infiltration, and recruitment to inflamed tissues, highlighting this pathway as a promising target for therapeutic intervention. In the present study, suramin, a polysulfonated napthylurea compound, having structure similarity with heparin, initially developed therapeutic for treating Human African Trypanosomas [HAT] was analyzed for its repressive action against CCL2 arbitrated macrophage migration. The study delves into the binding interaction between suramin (SUR) and CCL2 monomer, elucidating the molecular and biophysical underpinnings of their interaction through various techniques, including isothermal calorimetry, fluorescence spectroscopy, fluorescence lifetime studies, CD spectroscopy, and 2D NMR spectroscopy. Additionally, in-silico mechanistic studies employing molecular dynamic simulations, MMPBSA, and decomposition analysis unravel the intricacies of CCL2-SUR interactions. The molecule is observed to be attenuating the migration of macrophages by interacting with nanomolar affinity (119 ± 11 nM) on the CCL2 with the region overlapping with the CCR2/GAG binding pocket. Thus, this study comprehensively identified suramin, as a possible GAG mimetic for scheming structure-based drug molecules exhibiting anti-inflammatory action by aiming the CCL2-CCR2-GAG axis.
Collapse
Affiliation(s)
- Deepak Kumar Tripathi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Goutami Naidu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Nupur Nagar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Siddhartha Das Pramanik
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Partha Roy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Dinesh Kumar
- Centre of Biomedical Research, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS) Campus, Lucknow 226014, Uttar Pradesh, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
2
|
Kabinger F, Doze V, Schmitzová J, Lidschreiber M, Dienemann C, Cramer P. Structural basis of SARS-CoV-2 polymerase inhibition by nonnucleoside inhibitor HeE1-2Tyr. Proc Natl Acad Sci U S A 2025; 122:e2419854122. [PMID: 40035759 PMCID: PMC11912441 DOI: 10.1073/pnas.2419854122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/18/2025] [Indexed: 03/06/2025] Open
Abstract
Targeting the RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 with small molecules is a promising therapeutic strategy against COVID-19, but potent and safe inhibitors are lacking. HeE1-2Tyr, a nonnucleoside inhibitor of Dengue virus RdRp, was also shown to inhibit SARS-CoV-2 RdRp in vitro and to have antiviral activity in cells, but the underlying mechanism remains unclear. Here, we elucidate the molecular mechanism of HeE1-2Tyr-mediated SARS-CoV-2 RdRp inhibition. Biochemical assays confirm that HeE1-2Tyr inhibits RdRp with an IC50 of 5 µM and show that it competes with RNA binding to RdRp in vitro. Structural analysis using cryo-EM reveals that a stack of three HeE1-2Tyr molecules binds to the RNA binding site of RdRp. The identification of the conserved HeE1-2Tyr binding site and its intriguing inhibition mechanism of three stacked molecules that outcompete RNA may facilitate further development of pan-corona nonnucleoside inhibitors.
Collapse
Affiliation(s)
- Florian Kabinger
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Valerie Doze
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Jana Schmitzová
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Michael Lidschreiber
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Christian Dienemann
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Patrick Cramer
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| |
Collapse
|
3
|
Zwillinger M, Sőregi P, Sanchez F, Douat C, Csékei M, Huc I, Kotschy A. Development of Aromatic Foldamer Building Blocks Bearing Multiple Biogenic Side Chains. J Org Chem 2025; 90:3043-3052. [PMID: 39964779 DOI: 10.1021/acs.joc.4c02900] [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: 02/20/2025]
Abstract
Aromatic oligoamides, with their intrinsic rigidity and well-defined conformations, are recognized for their potential in medical applications. Similar structures are present in several naturally occurring antibiotics and have been explored for their ability to bind to various proteins and B-DNA (canonical right-handed DNA helix). This study introduces a synthetic approach to produce quinoline amino acid monomers bearing diversified side chain combinations in positions 4, 5, and 6 of the quinoline ring, designed to enhance the side chain density on helical foldamers. By increasing the number of side chains on each monomer, we aim to mimic the dense side chain presentation of α-peptides, thus improving the potential for protein surface recognition. This synthetic strategy involves efficient functionalization through cross-coupling reactions, enabling the installation of diverse side chains at strategic positions on the quinoline ring. The process has been optimized for automated solid-phase synthesis, successfully producing a 20-unit oligoamide with good purity. This foldamer, featuring multiple cationic, anionic, polar, and hydrophobic side chains, demonstrates the potential for molecular recognition in drug discovery and therapeutic applications. The methodology described here represents a significant advancement in the construction of aromatic oligoamide foldamers, providing a robust platform for further exploration of biological systems.
Collapse
Affiliation(s)
- Márton Zwillinger
- Servier Research Institute of Medicinal Chemistry, Záhony utca 7, Budapest 1031, Hungary
| | - Petra Sőregi
- Servier Research Institute of Medicinal Chemistry, Záhony utca 7, Budapest 1031, Hungary
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, Budapest 1117, Hungary
| | - Florian Sanchez
- Ludwig-Maximilians-Universität, Butenandtstr. 5-13, München 81377, Germany
| | - Céline Douat
- Ludwig-Maximilians-Universität, Butenandtstr. 5-13, München 81377, Germany
| | - Márton Csékei
- Servier Research Institute of Medicinal Chemistry, Záhony utca 7, Budapest 1031, Hungary
| | - Ivan Huc
- Ludwig-Maximilians-Universität, Butenandtstr. 5-13, München 81377, Germany
| | - András Kotschy
- Servier Research Institute of Medicinal Chemistry, Záhony utca 7, Budapest 1031, Hungary
| |
Collapse
|
4
|
Huang SH, Wang CC, Shen PC, Liu ZM, Chen SJ, Tien YC, Lu CC. Suramin enhances proliferation, migration, and tendon gene expression of human supraspinatus tenocytes. J Orthop Res 2025; 43:252-263. [PMID: 39358851 DOI: 10.1002/jor.25990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/23/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Rotator cuff tendinopathy is a common musculoskeletal disorder with limited pharmacological treatment strategies. This study aimed to investigate tenocytes' functional in vitro response from a ruptured supraspinatus tendon to suramin administration and to elucidate whether suramin can enhance tendon repair and modulate the inflammatory response to injury. Tenocytes were obtained from human supraspinatus tendons (n = 6). We investigated the effect of suramin on LPS-induced inflammatory responses and the underlying molecular mechanisms in THP-1 macrophages. Suramin enhanced the proliferation, cell viability, and migration of tenocytes. It also increased the protein expression of PCNA and Ki-67. Suramin-treated tenocytes exhibited increased expression of COL1A1, COL3A1, TNC, SCX, and VEGF. Suramin significantly reduced LPS-induced iNOS, COX2 synthesis, inflammatory cytokine TNF-α production, and inflammatory signaling by influencing the NF-κB pathways in THP-1 cells. Our results suggest that suramin holds great promise as a therapeutic option for treating rotator cuff tendinopathy.
Collapse
Affiliation(s)
- Shih-Hao Huang
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Chien Wang
- Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Po-Chih Shen
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zi-Miao Liu
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shu-Jung Chen
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yin-Chun Tien
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Cheng-Chang Lu
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopaedic Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Llanos S, Di Geronimo B, Casajús E, Blanco-Romero E, Fernández-Leiro R, Méndez J. Interference of small compounds and Mg 2+ with dsRNA-binding fluorophores compromises the identification of SARS-CoV-2 RdRp inhibitors. Sci Rep 2024; 14:28250. [PMID: 39548173 PMCID: PMC11568178 DOI: 10.1038/s41598-024-78354-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024] Open
Abstract
The COVID-19 pandemic highlighted the need for the rapid development of antiviral therapies. Viral RNA-dependent RNA polymerases (RdRp) are promising targets, and numerous virtual screenings for potential inhibitors were conducted without validation of the identified hits. Here we have tested a set of presumed RdRp inhibitors in biochemical assays based on fluorometric detection of RdRp activity or on the electrophoretic separation or RdRp products. We find that fluorometric detection of RdRp activity is unreliable as a screening method because many small compounds interfere with fluorophore binding to dsRNA, and this effect is enhanced by the Mg2+ metal ions used by nucleic acid polymerases. The fact that fluorimetric detection of RdRp activity leads to false-positive hits underscores the requirement for independent validation methods. We also show that suramin, one of the proposed RdRp inhibitors that could be validated biochemically, is a multi-polymerase inhibitor. While this does not hinder its potential as an antiviral agent, it cannot be considered an specific inhibitor of SARS-CoV-2 RdRp.
Collapse
Affiliation(s)
- Susana Llanos
- DNA Replication Group, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 3 Melchor Fernandez Almagro, 28029, Madrid, Spain.
| | - Bruno Di Geronimo
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive NW, Atlanta, GA, 30332-0400, USA
| | - Ester Casajús
- Genome Integrity and Structural Biology Group, Structural Biology Programme, Spanish National Cancer Research Center (CNIO), 3 Melchor Fernandez Almagro, 28029, Madrid, Spain
| | - Elena Blanco-Romero
- DNA Replication Group, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 3 Melchor Fernandez Almagro, 28029, Madrid, Spain
- Genome Integrity and Structural Biology Group, Structural Biology Programme, Spanish National Cancer Research Center (CNIO), 3 Melchor Fernandez Almagro, 28029, Madrid, Spain
| | - Rafael Fernández-Leiro
- Genome Integrity and Structural Biology Group, Structural Biology Programme, Spanish National Cancer Research Center (CNIO), 3 Melchor Fernandez Almagro, 28029, Madrid, Spain
| | - Juan Méndez
- DNA Replication Group, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 3 Melchor Fernandez Almagro, 28029, Madrid, Spain.
| |
Collapse
|
6
|
Boniardi I, Corona A, Basquin J, Basquin C, Milia J, Nagy I, Tramontano E, Zinzula L. Suramin inhibits SARS-CoV-2 nucleocapsid phosphoprotein genome packaging function. Virus Res 2023; 336:199221. [PMID: 37704176 PMCID: PMC10514558 DOI: 10.1016/j.virusres.2023.199221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/27/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is fading, however its etiologic agent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues posing - despite the availability of licensed vaccines - a global health threat, due to the potential emergence of vaccine-resistant SARS-CoV-2 variants. This makes the development of new drugs against COVID-19 a persistent urgency and sets as research priority the validation of novel therapeutic targets within the SARS-CoV-2 proteome. Among these, a promising one is the SARS-CoV-2 nucleocapsid (N) phosphoprotein, a major structural component of the virion with indispensable role in packaging the viral genome into a ribonucleoprotein (RNP) complex, which also contributes to SARS-CoV-2 innate immune evasion by inhibiting the host cell type-I interferon (IFN-I) response. By combining miniaturized differential scanning fluorimetry with microscale thermophoresis, we found that the 100-year-old drug Suramin interacts with SARS-CoV-2 N-terminal domain (NTD) and C-terminal domain (CTD), thereby inhibiting their single-stranded RNA (ssRNA) binding function with low-micromolar Kd and IC50 values. Molecular docking suggests that Suramin interacts with basic NTD cleft and CTD dimer interface groove, highlighting three potentially druggable ssRNA binding sites. Electron microscopy shows that Suramin inhibits the formation in vitro of RNP complex-like condensates by SARS-CoV-2 N with a synthetic ssRNA. In a dose-dependent manner, Suramin also reduced SARS-CoV-2-induced cytopathic effect on Vero E6 and Calu-3 cells, partially reverting the SARS-CoV-2 N-inhibited IFN-I production in 293T cells. Our findings indicate that Suramin inhibits SARS-CoV-2 replication by hampering viral genome packaging, thereby representing a starting model for design of new COVID-19 antivirals.
Collapse
Affiliation(s)
- Irene Boniardi
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato 09042, Italy
| | - Jerome Basquin
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Claire Basquin
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Jessica Milia
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato 09042, Italy
| | - István Nagy
- Center of Research and Development, Eszterházy Károly Catholic University, Eger 3300, Hungary
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato 09042, Italy.
| | - Luca Zinzula
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried 82152, Germany.
| |
Collapse
|
7
|
Wang QJ, Wipf P. Small Molecule Inhibitors of Protein Kinase D: Early Development, Current Approaches, and Future Directions. J Med Chem 2023; 66:122-139. [PMID: 36538005 DOI: 10.1021/acs.jmedchem.2c01599] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Now entering its fourth decade, research on the biological function, small molecule inhibition, and disease relevance of the three known isoforms of protein kinase D, PKD1, PKD2, and PKD3, has entered a mature development stage. This mini-perspective focuses on the medicinal chemistry that provided a structurally diverse set of mainly active site inhibitors, which, for a brief time period, moved through preclinical development stages but have yet to be tested in clinical trials. In particular, between 2006 and 2012, a rapid expansion of synthetic efforts led to several moderately to highly PKD-selective chemotypes but did not yet achieve PKD subtype selectivity or resolve general toxicity and pharmacokinetic challenges. In addition to cancer, other unresolved medical needs in cardiovascular, inflammatory, and metabolic diseases would, however, benefit from a renewed focus on potent and selective PKD modulators.
Collapse
Affiliation(s)
- Qiming Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States.,School of Pharmacy, University of Eastern Finland, 70210 Kuopio, Finland
| |
Collapse
|
8
|
Sammons R, Bohanon AL, Kowtha A, Dejong A, Cho EJ, Kaoud TS, Dalby KN. High-Throughput Assay for Identifying Diverse Antagonists of the Binding Interaction between the ACE2 Receptor and the Dynamic Spike Proteins of SARS-CoV-2. ACS Infect Dis 2022; 8:2259-2270. [PMID: 36315931 PMCID: PMC9673917 DOI: 10.1021/acsinfecdis.2c00297] [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: 06/08/2022] [Indexed: 01/29/2023]
Abstract
SARS-CoV-2, a coronavirus strain that started a worldwide pandemic in early 2020, attaches to human cells by binding its spike (S) glycoprotein to a host receptor protein angiotensin-converting enzyme 2 (ACE2). Blocking the interaction between the S protein and ACE2 has emerged as an important strategy for preventing viral infection. We systematically developed and optimized an AlphaLISA assay to investigate binding events between ACE2 and the ectodomain of the SARS-CoV-2 S protein (S-614G: residues 1-1208 with a D614G mutation). Using S-614G permits discovering potential allosteric inhibitors that stabilize the S protein in a conformation that impedes its access to ACE2. Over 30,000 small molecules were screened in a high-throughput format for activity against S-614G and ACE2 binding using the AlphaLISA assay. A viral entry assay was used to validate hits using lentiviral particles pseudotyped with the full-length S protein of the Wuhan-1 strain. Two compounds identified in the screen, oleic acid and suramin, blocked the attachment of S-614G to ACE2 and S protein-driven cell entry into Calu-3 and ACE2-overexpressing HEK293T cells. Oleic acid inhibits S-614G binding to ACE2 far more potently than to the receptor-binding domain (RBD, residues 319-541 of SARS-CoV-2 S), potentially indicating a noncompetitive mechanism. The results indicate that using the full-length ectodomain of the S protein can be important for identifying allosteric inhibitors of ACE2 binding. The approach reported here represents a rapidly adaptable format for discovering receptor-binding inhibitors to S-proteins of future coronavirus strains.
Collapse
Affiliation(s)
- Rachel
M. Sammons
- Targeted
Therapeutic Drug Discovery and Development Program, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Amanda L. Bohanon
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
| | - Anvith Kowtha
- Division
of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Audrey Dejong
- Division
of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Eun Jeong Cho
- Targeted
Therapeutic Drug Discovery and Development Program, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Tamer S. Kaoud
- Targeted
Therapeutic Drug Discovery and Development Program, The University of Texas at Austin, Austin, Texas 78712, United States
- Division
of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Kevin N. Dalby
- Targeted
Therapeutic Drug Discovery and Development Program, The University of Texas at Austin, Austin, Texas 78712, United States
- Division
of Chemical Biology and Medicinal Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|